1
|
Zamanian MY, Maleki S, Oghenemaro EF, Singh M, Mohammadi M, Alkhayyat AH, Sapaev IB, Kaur P, Shirsalimi N, Nagarwal A. Omentin-1 as a promising biomarker and therapeutic target in hypertension and heart failure: a comprehensive review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04008-y. [PMID: 40126671 DOI: 10.1007/s00210-025-04008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/02/2025] [Indexed: 03/26/2025]
Abstract
Omentin-1, a novel adipocytokine predominantly secreted by visceral adipose tissue, has emerged as a significant factor in cardiovascular health, particularly regarding hypertension (HTN) and heart failure (HF). This manuscript investigates the multifaceted roles of omentin-1 in these conditions, emphasizing its protective effects on vascular function and its potential as both a biomarker and therapeutic target. Clinical studies indicate that reduced circulating levels of omentin-1 are associated with metabolic syndrome (MetS) and increased cardiovascular risk, while animal studies demonstrate its ability to ameliorate endothelial dysfunction and lower blood pressure. Omentin-1 exerts its beneficial effects through various signaling pathways, including AMP-activated protein kinase (AMPK) and protein kinase B (Akt), thereby promoting vasodilation, enhancing insulin sensitivity, and mitigating inflammation. In the context of HF, particularly heart failure with preserved ejection fraction (HFpEF), omentin-1 levels exhibit a negative correlation with diastolic dysfunction and inflammatory markers, suggesting its role in cardiac protection. Additionally, the manuscript discusses the implications of omentin-1 in managing obesity-related cardiovascular diseases and its potential utility as a prognostic marker for adverse outcomes in HF patients. Collectively, omentin-1 represents a promising avenue for research in cardiovascular health, with the potential to inform novel therapeutic strategies aimed at improving outcomes in patients with HTN and HF. Further research is necessary to elucidate the details of omentin-1 function and evaluate its potential in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Mohammad Yasin Zamanian
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Saba Maleki
- Tehran Heart Center, Cardiovascular Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Guilan University of Medical Sciences (GUMS), Rasht, Guilan Province, Iran
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology and Biotechnology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Mandeep Singh
- Directorate of Sports and Physical Education, University of Jammu, Jammu, India
| | - Maryam Mohammadi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Ahmad Hussen Alkhayyat
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Computers Techniques Engineering, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Ibrokhim B Sapaev
- Tashkent Institute of Irrigation and Agricultural Mechanization Engineers" National Research University, Tashkent, Uzbekistan
- Scientific Researcher, University of Tashkent for Applied Sciences, Str. Gavhar 1, 100149, Tashkent, Uzbekistan
- Western Caspian University, Scientific Researcher, Baku, Azerbaijan
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Niyousha Shirsalimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6718773654, Iran.
| | - Amritesh Nagarwal
- Department of Cardiology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| |
Collapse
|
2
|
Mousavi SM, Jalali-Zefrei F, Shourmij M, Tabaghi S, Davari A, Khalili SB, Farzipour S, Salari A. Targeting Wnt Pathways with Small Molecules as New Approach in Cardiovascular Disease. Curr Cardiol Rev 2025; 21:108-122. [PMID: 39482911 PMCID: PMC12060913 DOI: 10.2174/011573403x333038241023153349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 11/03/2024] Open
Abstract
The increasing incidences of morbidity and mortality associated with cardiovascular diseases represent significant difficulties for clinical treatment and have a major impact on patient health. Wnt signaling pathways are highly conserved and are well known for their regulatory roles in embryonic development, tissue regeneration, and adult tissue homeostasis. Wnt signaling is classified into two distinct pathways: canonical Wnt/β-catenin signaling and noncanonical pathways, including planar cell polarity and Wnt/Ca2+ pathways. A growing body of experimental evidence suggests the involvement of both canonical and non-canonical Wnt signaling pathways in the development of cardiovascular diseases, including myocardial hypertrophy, arrhythmias, diabetic cardiomyopathy, arrhythmogenic cardiomyopathy, and myocardial infarction. Thus, to enhance patient quality of life, diagnosing and treating cardiac illnesses may require a thorough understanding of the molecular functions played by the Wnt pathway in these disorders. Many small-molecule inhibitors specifically target various components within the Wnt signaling pathways, such as Frizzled, Disheveled, Porcupine, and Tankyrase. This study aims to present an overview of the latest findings regarding the functions of Wnt signaling in human cardiac disorders and possible inhibitors of Wnt, which could lead to novel approaches for treating cardiac ailments.
Collapse
Affiliation(s)
- Seyed Mehdi Mousavi
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Jalali-Zefrei
- Department of radiology, Faculty of Medicine, Guilan University of Medical Science, Rasht, Iran
| | - Mohammad Shourmij
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Shiva Tabaghi
- Cardiovascular Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhesam Davari
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Saeed Bahador Khalili
- Department of Electronic Engineering, Universitat Rovira i Virgili, Tarragona, 43007, Spain
| | - Soghra Farzipour
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Arsalan Salari
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
3
|
Svetláková BB, Líšková VP, Barančík M. Wnt Signaling Inhibitors as Therapeutic Approach in Ischemic Heart Disease. Molecules 2024; 29:5958. [PMID: 39770047 PMCID: PMC11677181 DOI: 10.3390/molecules29245958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/05/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Wnt (wingless-type MMTV integration site family) signaling is an evolutionary conserved system highly active during embryogenesis, but in adult hearts has low activities under normal conditions. It is essential for a variety of physiological processes including stem cell regeneration, proliferation, migration, cell polarity, and morphogenesis, thereby ensuring homeostasis and regeneration of cardiac tissue. Its dysregulation and excessive activation during pathological conditions leads to morphological and functional changes in the heart resulting in impaired myocardial regeneration under pathological conditions such as myocardial infarction, heart failure, and coronary artery disease. Several groups of Wnt inhibitors have demonstrated the ability to modulate the Wnt pathway and thereby significantly reduce fibrosis and improve cardiac function after myocardial ischemia. Their inhibitory effect can be realized at multiple levels, which include the inhibition of Wnt ligands, the inhibition of Frizzled receptors, the stabilization of the β-catenin destruction complex, and the disruption of nuclear β-catenin interactions. In this review, we overview the function of Wnt signaling in responses of cardiac cells to pathological conditions, especially ischemic heart disease, with an emphasis on the use of inhibitors of this signaling as a therapeutic approach. Finally, we summarize the current knowledge about the potential of the targeting of Wnt signaling in therapeutic applications.
Collapse
Affiliation(s)
| | | | - Miroslav Barančík
- Centre of Experimental Medicine, Slovak Academy of Sciences, 841 04 Bratislava, Slovakia; (B.B.S.); (V.P.L.)
| |
Collapse
|
4
|
Shin J, Yoon Y, Oh DJ. Clinical impact of Wnt5a expression on persistence of acute kidney injury in patients with urosepsis. Ren Fail 2024; 46:2369176. [PMID: 38913943 PMCID: PMC11198152 DOI: 10.1080/0886022x.2024.2369176] [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: 02/26/2024] [Accepted: 06/12/2024] [Indexed: 06/26/2024] Open
Abstract
Abnormal Wnt5a expression is associated with dysregulated inflammation and organ dysfunction. However, the effect of Wnt5a activation on the duration of organ dysfunction remains unclear. This prospective study investigated the association between Wnt5a levels and persistent acute kidney injury (AKI) in patients with urosepsis. Serum creatinine and Wnt5a levels were measured on days 1 and 5 and at discharge in 87 patients diagnosed with urosepsis. Patients with urosepsis were classified into an improving acute kidney injury (AKI) group and a persistent or worsening AKI group according to the AKI stage on days 1 and 5. AKI recovery was defined as a discharge-to-baseline serum creatinine ratio of <1.5. Twenty-eight patients with urosepsis (32.2%) had persistent or worsening AKI, and their Wnt5a levels were higher on days 1 and 5 and at discharge than those with improving AKI. The association between Wnt5a levels and persistent or worsening AKI was maintained after adjusting for age, sex, baseline serum creatinine levels, and disease severity. Moreover, elevated Wnt5a levels were associated with an increased risk of major adverse kidney events. High Wnt5a levels at discharge were associated with unrecovered AKI and participants with AKI recovery had a steeper Wnt5a slope over time than those without recovery, irrespective of age, sex, baseline serum creatinine level, or disease severity. Assessment of Wnt5a expression was helpful in predicting AKI persistence and adverse outcomes in patients with urosepsis. Therefore, Wnt5a may serve as a valuable bio-marker for identifying the risk of persistence of AKI.
Collapse
Affiliation(s)
- Jungho Shin
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Yoosik Yoon
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Dong-Jin Oh
- Department of Internal Medicine, Myongji Hospital, Hanyang University College of Medicine, Goyang, South Korea
| |
Collapse
|
5
|
Chong A, Joshua J, Raheb S, Pires A, Colpitts M, Caswell JL, Fonfara S. Evaluation of potential novel biomarkers for feline hypertrophic cardiomyopathy. Res Vet Sci 2024; 180:105430. [PMID: 39395261 DOI: 10.1016/j.rvsc.2024.105430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Hypertrophic cardiomyopathy (HCM) is the most common cardiomyopathy in cats. The diagnosis can be difficult, requiring advanced echocardiographic skills. Additionally, circulating biomarkers (N-terminal pro-B type natriuretic peptide and cardiac troponin I) have several limitations when used for HCM screening. In previous work, we identified interleukin 18 (IL-18), insulin-like growth factor binding protein 2 (IGFBP-2), brain-type glycogen phosphorylase B (PYGB), and WNT Family Member 5 A (WNT5A) as myocardial genes that show significant differential expression between cats with HCM and healthy cats. The products of these genes are released into the circulation, and we hypothesized that IL-18, IGFBP-2, PYGB, and WNT5A serum RNA and protein concentrations differ between healthy cats, cats with subclinical HCM, and those with HCM and congestive heart failure (HCM + CHF). Reverse transcriptase quantitative polymerase chain reaction (RTqPCR) and enzyme-linked immunosorbent assay (ELISA) were applied to evaluate gene and protein expression, respectively, in the serum of eight healthy controls, eight cats with subclinical HCM, and six cats with HCM + CHF. Serum IGFBP-2 RNA concentrations were significantly different among groups and were highest in cats with subclinical HCM. Compared to healthy controls, serum IL-18 and WNT5A gene expression were significantly higher in cats with HCM + CHF, and WNT5A was higher in cats with subclinical HCM. No differences were observed for PYGB. These results indicate that further investigation via large scale clinical studies for IGFBP-2, WNT5A, and IL-18 may be valuable in diagnosing and staging feline HCM.
Collapse
Affiliation(s)
- Andrew Chong
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Jessica Joshua
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada; Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road E., Guelph, ON N1G 2W1, Canada
| | - Shari Raheb
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Ananda Pires
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Michelle Colpitts
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada
| | - Jeff L Caswell
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, 50 Stone Road E., Guelph, ON N1G 2W1, Canada
| | - Sonja Fonfara
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, 28 College Ave W, Guelph, Ontario N1G 2W1, Canada.
| |
Collapse
|
6
|
Luo B, Zheng R, Shi C, Chen D, Jin X, Hou J, Xu G, Hu B. DACT2 modulates atrial fibrillation through TGF/β and Wnt signaling pathways. Heliyon 2024; 10:e36050. [PMID: 39224277 PMCID: PMC11367123 DOI: 10.1016/j.heliyon.2024.e36050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia that seriously affects the quality of life of patients. Effective treatment and prevention are important to control the morbidity and mortality of AF. It has been found that cardiac fibrosis promotes the onset and progression of AF. It is now known that transforming growth factor β (TGF-β), an important fibrotic cytokine, plays an important role in cardiac fibrosis by inducing myofibroblast activation via the activation of classical (SMAD-based) and non-classical (non-SMAD-based) signaling pathways. In addition, specific activation of the Wnt/β-catenin pathway has been shown to promote the transformation of fibroblasts into myofibroblasts. In recent years, a new family of proteins, namely Disheveled-associated antagonist of beta-catenin (DACT) 2, can affect the Wnt/β-catenin and TGF-β signaling pathways by regulating the phosphorylation levels of these target proteins, which in turn affects the progression of fibrosis. The present study focuses on the effect of DACT2-guided β-catenin on atrial fibrosis. It is expected that the summarized information can be helpful in the treatment of AF.
Collapse
Affiliation(s)
- Bairu Luo
- Department of Clinical Pathology, Jiaxing University Master Degree Cultivation Base, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, 314001, ZJ, China
| | - Rui Zheng
- Department of Clinical Pathology, The 3rd Clinical Medical College Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310053, ZJ, China
| | - Chaoqun Shi
- Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, ZJ, China
| | - Deqing Chen
- Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, ZJ, China
| | - Xin Jin
- Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, ZJ, China
| | - Jian Hou
- Department of Cardiology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, 510080, GD, China
| | - Guangtao Xu
- Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, ZJ, China
| | - Bo Hu
- Department of Clinical Pathology, Jiaxing Hospital of Traditional Chinese Medicine, the 3rd Clinical Medical College Affiliated to Zhejiang Chinese Medical University, Jiaxing, 314001, ZJ, China
| |
Collapse
|
7
|
Zhang J, Li Y, Chen Y, Zhang J, Jia Z, He M, Liao X, He S, Bian JS, Nie XW. o 8G Site-Specifically Modified tRF-1-AspGTC: A Novel Therapeutic Target and Biomarker for Pulmonary Hypertension. Circ Res 2024; 135:76-92. [PMID: 38747146 DOI: 10.1161/circresaha.124.324421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 04/18/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Hypoxia and oxidative stress contribute to the development of pulmonary hypertension (PH). tRNA-derived fragments play important roles in RNA interference and cell proliferation, but their epitranscriptional roles in PH development have not been investigated. We aimed to gain insight into the mechanistic contribution of oxidative stress-induced 8-oxoguanine in pulmonary vascular remodeling. METHODS Through small RNA modification array analysis and quantitative polymerase chain reaction, a significant upregulation of the 8-oxoguanine -modified tRF-1-AspGTC was found in the lung tissues and the serum of patients with PH. RESULTS This modification occurs at the position 5 of the tRF-1-AspGTC (5o8G tRF). Inhibition of the 5o8G tRF reversed hypoxia-induced proliferation and apoptosis resistance in pulmonary artery smooth muscle cells. Further investigation unveiled that the 5o8G tRF retargeted mRNA of WNT5A (Wingless-type MMTV integration site family, member 5A) and CASP3 (Caspase3) and inhibited their expression. Ultimately, BMPR2 (Bone morphogenetic protein receptor 2) -reactive oxygen species/5o8G tRF/WNT5A signaling pathway exacerbated the progression of PH. CONCLUSIONS Our study highlights the role of site-specific 8-oxoguanine-modified tRF in promoting the development of PH. Our findings present a promising therapeutic avenue for managing PH and propose 5o8G tRF as a potential innovative marker for diagnosing this disease.
Collapse
Affiliation(s)
- Junting Zhang
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- The First Affiliated Hospital, Southern University of Science and Technology), China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- Post-Doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, China (Junting Zhang, Y.L., S.H.)
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China (Junting Zhang, Z.J., M.H., J.-S.B.)
| | - Yiying Li
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- The First Affiliated Hospital, Southern University of Science and Technology), China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- Post-Doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, China (Junting Zhang, Y.L., S.H.)
| | - Yuan Chen
- Lung Transplant Group, Wuxi People's Hospital Affiliated to Nanjing Medical University, China (Y.C.)
| | - Jianchao Zhang
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- The First Affiliated Hospital, Southern University of Science and Technology), China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
| | - Zihui Jia
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China (Junting Zhang, Z.J., M.H., J.-S.B.)
| | - Muhua He
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- The First Affiliated Hospital, Southern University of Science and Technology), China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China (Junting Zhang, Z.J., M.H., J.-S.B.)
| | - Xueyi Liao
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- The First Affiliated Hospital, Southern University of Science and Technology), China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
| | - Siyu He
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- The First Affiliated Hospital, Southern University of Science and Technology), China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- Post-Doctoral Scientific Research Station of Basic Medicine, Jinan University, Guangzhou, China (Junting Zhang, Y.L., S.H.)
| | - Jin-Song Bian
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- The First Affiliated Hospital, Southern University of Science and Technology), China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- Department of Pharmacology, Joint Laboratory of Guangdong-Hong Kong Universities for Vascular Homeostasis and Diseases, School of Medicine, Southern University of Science and Technology, Shenzhen, China (Junting Zhang, Z.J., M.H., J.-S.B.)
| | - Xiao-Wei Nie
- Shenzhen Institute of Respiratory Disease, Shenzhen Key Laboratory of Respiratory Disease, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
- The First Affiliated Hospital, Southern University of Science and Technology), China (Junting Zhang, Y.L., Jianchao Zhang, Z.J., M.H., X.L., S.H., J.-S.B., X.-W.N.)
| |
Collapse
|
8
|
Li X, Turaga D, Li RG, Tsai CR, Quinn JN, Zhao Y, Wilson R, Carlson K, Wang J, Spinner JA, Hickey EJ, Adachi I, Martin JF. The Macrophage Landscape Across the Lifespan of a Human Cardiac Allograft. Circulation 2024; 149:1650-1666. [PMID: 38344825 PMCID: PMC11105989 DOI: 10.1161/circulationaha.123.065294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 01/16/2024] [Indexed: 05/22/2024]
Abstract
BACKGROUND Much of our knowledge of organ rejection after transplantation is derived from rodent models. METHODS We used single-nucleus RNA sequencing to investigate the inflammatory myocardial microenvironment in human pediatric cardiac allografts at different stages after transplantation. We distinguished donor- from recipient-derived cells using naturally occurring genetic variants embedded in single-nucleus RNA sequencing data. RESULTS Donor-derived tissue resident macrophages, which accompany the allograft into the recipient, are lost over time after transplantation. In contrast, monocyte-derived macrophages from the recipient populate the heart within days after transplantation and form 2 macrophage populations: recipient MP1 and recipient MP2. Recipient MP2s have cell signatures similar to donor-derived resident macrophages; however, they lack signatures of pro-reparative phagocytic activity typical of donor-derived resident macrophages and instead express profibrotic genes. In contrast, recipient MP1s express genes consistent with hallmarks of cellular rejection. Our data suggest that recipient MP1s activate a subset of natural killer cells, turning them into a cytotoxic cell population through feed-forward signaling between recipient MP1s and natural killer cells. CONCLUSIONS Our findings reveal an imbalance of donor-derived and recipient-derived macrophages in the pediatric cardiac allograft that contributes to allograft failure.
Collapse
Affiliation(s)
- Xiao Li
- The Texas Heart Institute, Houston, TX, USA
| | - Diwakar Turaga
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Division of Critical Care Medicine, Texas Children’s Hospital, Houston TX, USA
| | - Rich G. Li
- The Texas Heart Institute, Houston, TX, USA
| | - Chang-Ru Tsai
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Julianna N. Quinn
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, TX, USA
| | - Yi Zhao
- The Texas Heart Institute, Houston, TX, USA
| | | | - Katherine Carlson
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, TX, USA
| | - Joseph A. Spinner
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Division of Cardiology, Texas Children’s Hospital, Houston, TX, USA
| | - Edward J. Hickey
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, USA
| | - Iki Adachi
- Department of Surgery, Baylor College of Medicine, Houston, Texas, USA
- Division of Congenital Heart Surgery, Texas Children’s Hospital, Houston, TX, USA
| | - James F. Martin
- The Texas Heart Institute, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
9
|
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
|
10
|
Trinh-Minh T, Chen CW, Tran Manh C, Li YN, Zhu H, Zhou X, Chakraborty D, Zhang Y, Rauber S, Dees C, Lin NY, Kah D, Gerum R, Bergmann C, Kreuter A, Reuter C, Groeber-Becker F, Eckes B, Distler O, Fabry B, Ramming A, Schambony A, Schett G, Distler JH. Noncanonical WNT5A controls the activation of latent TGF-β to drive fibroblast activation and tissue fibrosis. J Clin Invest 2024; 134:e159884. [PMID: 38747285 PMCID: PMC11093613 DOI: 10.1172/jci159884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/20/2024] [Indexed: 05/19/2024] Open
Abstract
Transforming growth factor β (TGF-β) signaling is a core pathway of fibrosis, but the molecular regulation of the activation of latent TGF-β remains incompletely understood. Here, we demonstrate a crucial role of WNT5A/JNK/ROCK signaling that rapidly coordinates the activation of latent TGF-β in fibrotic diseases. WNT5A was identified as a predominant noncanonical WNT ligand in fibrotic diseases such as systemic sclerosis, sclerodermatous chronic graft-versus-host disease, and idiopathic pulmonary fibrosis, stimulating fibroblast-to-myofibroblast transition and tissue fibrosis by activation of latent TGF-β. The activation of latent TGF-β requires rapid JNK- and ROCK-dependent cytoskeletal rearrangements and integrin αV (ITGAV). Conditional ablation of WNT5A or its downstream targets prevented activation of latent TGF-β, rebalanced TGF-β signaling, and ameliorated experimental fibrosis. We thus uncovered what we believe to be a novel mechanism for the aberrant activation of latent TGF-β in fibrotic diseases and provided evidence for targeting WNT5A/JNK/ROCK signaling in fibrotic diseases as a new therapeutic approach.
Collapse
Affiliation(s)
- Thuong Trinh-Minh
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Chih-Wei Chen
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Cuong Tran Manh
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Yi-Nan Li
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Honglin Zhu
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Xiang Zhou
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Debomita Chakraborty
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Yun Zhang
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| | - Simon Rauber
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Clara Dees
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Neng-Yu Lin
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
- Graduate Institute of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Delf Kah
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Richard Gerum
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Christina Bergmann
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Alexander Kreuter
- Clinic for Dermatology, Venereology and Allergology, HELIOS St. Elisabeth Clinic Oberhausen, North-Rhine-Westphalia, Germany
| | - Christiane Reuter
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research (ISC) Würzburg, Bavaria, Germany
| | - Florian Groeber-Becker
- Translational Center for Regenerative Therapies, Fraunhofer Institute for Silicate Research (ISC) Würzburg, Bavaria, Germany
| | - Beate Eckes
- Translational Matrix Biology, University of Cologne, Cologne, North-Rhine-Westphalia, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, North-Rhine-Westphalia, Germany
| | - Oliver Distler
- Rheumaklinik, University Hospital Zurich, Zurich, Switzerland
| | - Ben Fabry
- Department of Physics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Andreas Ramming
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Alexandra Schambony
- Division of Developmental Biology, Biology Department, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Bavaria, Germany
| | - Georg Schett
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Bavaria, Germany
- German Center for Immunotherapy, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University of Erlangen, Erlangen, Bavaria, Germany
| | - Jörg H.W. Distler
- Department of Rheumatology and
- Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, North-Rhine-Westphalia, Germany
| |
Collapse
|
11
|
Huang Y, Xue Q, Chang J, Wang X, Miao C. Wnt5a: A promising therapeutic target for inflammation, especially rheumatoid arthritis. Cytokine 2023; 172:156381. [PMID: 37806072 DOI: 10.1016/j.cyto.2023.156381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/05/2023] [Accepted: 09/22/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Wnt5a is a member of the Wnt protein family, which acts on classical or multiple non-classical Wnt signaling pathways by binding to different receptors. The expression regulation and signal transduction of Wnt5a is closely related to the inflammatory response. Abnormal activation of Wnt5a signaling is an important part of inflammation and rheumatoid arthritis (RA). OBJECTIVES This paper mainly focuses on Wnt5a protein and its mediated signaling pathway, summarizes the latest research progress of Wnt5a in the pathological process of inflammation and RA, and looks forward to the main directions of Wnt5a in RA research, aiming to provide a theoretical basis for the prevention and treatment of RA diseases by targeting Wnt5a. RESULTS Wnt5a is highly expressed in activated blood vessels, histocytes and synoviocytes in inflammatory diseases such as sepsis, sepsis, atherosclerosis and rheumatoid arthritis. It mediates the production of pro-inflammatory cytokines and chemokines, regulates the migration and recruitment of various immune effector cells, and thus participates in the inflammatory response. Wnt5a plays a pathological role in synovial inflammation and bone destruction of RA, and may be an important clinical therapeutic target for RA. CONCLUSION Wnt5a is involved in the pathological process of inflammation and interacts with inflammatory factors. Wnt5a may be a new target for regulating the progression of RA disease and intervening therapy because of its multi-modal effects on the etiology of RA, especially as a regulator of osteoclast activity and inflammation.
Collapse
Affiliation(s)
- Yurong Huang
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qiuyun Xue
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Jun Chang
- Department of Orthopaedics, the First Affiliated Hospital, Anhui Medical University, Hefei 230032, China; Anhui Public Health Clinical Center, Hefei, China.
| | - Xiao Wang
- Department of Clinical Nursing, School of Nursing, Anhui University of Chinese Medicine, Hefei, China.
| | - Chenggui Miao
- Department of Pharmacology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
12
|
Cai D, Wang X, Sun Y, Fan H, Zhou J, Yang Z, Qiu H, Wang J, Su J, Gong T, Jiang C, Liang P. Patient-specific iPSC-derived cardiomyocytes reveal aberrant activation of Wnt/β-catenin signaling in SCN5A-related Brugada syndrome. Stem Cell Res Ther 2023; 14:241. [PMID: 37679791 PMCID: PMC10486057 DOI: 10.1186/s13287-023-03477-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 08/29/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Mutations in the cardiac sodium channel gene SCN5A cause Brugada syndrome (BrS), an arrhythmic disorder that is a leading cause of sudden death and lacks effective treatment. An association between SCN5A and Wnt/β-catenin signaling has been recently established. However, the role of Wnt/β-catenin signaling in BrS and underlying mechanisms remains unknown. METHODS Three healthy control subjects and one BrS patient carrying a novel frameshift mutation (T1788fs) in the SCN5A gene were recruited in this study. Control and BrS patient-specific induced pluripotent stem cells (iPSCs) were generated from skin fibroblasts using nonintegrated Sendai virus. All iPSCs were differentiated into cardiomyocytes using monolayer-based differentiation protocol. Action potentials and sodium currents were recorded from control and BrS iPSC-derived cardiomyocytes (iPSC-CMs) by single-cell patch clamp. RESULTS BrS iPSC-CMs exhibited increased burden of arrhythmias and abnormal action potential profile featured by slower depolarization, decreased action potential amplitude, and increased beating interval variation. Moreover, BrS iPSC-CMs showed cardiac sodium channel (Nav1.5) loss-of-function as compared to control iPSC-CMs. Interestingly, the electrophysiological abnormalities and Nav1.5 loss-of-function observed in BrS iPSC-CMs were accompanied by aberrant activation of Wnt/β-catenin signaling. Notably, inhibition of Wnt/β-catenin significantly rescued Nav1.5 defects and arrhythmic phenotype in BrS iPSC-CMs. Mechanistically, SCN5A-encoded Nav1.5 interacts with β-catenin, and reduced expression of Nav1.5 leads to re-localization of β-catenin in BrS iPSC-CMs, which aberrantly activates Wnt/β-catenin signaling to suppress SCN5A transcription. CONCLUSIONS Our findings suggest that aberrant activation of Wnt/β-catenin signaling contributes to the pathogenesis of SCN5A-related BrS and point to Wnt/β-catenin as a potential therapeutic target.
Collapse
Affiliation(s)
- Dongsheng Cai
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Xiaochen Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Yaxun Sun
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Hangping Fan
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Jingjun Zhou
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Zongkuai Yang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hangyuan Qiu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China
| | - Jue Wang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Jun Su
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Tingyu Gong
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Chenyang Jiang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 Qingchun East Road, Hangzhou, 310016, China.
| | - Ping Liang
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, China.
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China.
| |
Collapse
|
13
|
Kishimoto H, Iwasaki M, Wada K, Horitani K, Tsukamoto O, Kamikubo K, Nomura S, Matsumoto S, Harada T, Motooka D, Okuzaki D, Takashima S, Komuro I, Kikuchi A, Shiojima I. Wnt5a-YAP signaling axis mediates mechanotransduction in cardiac myocytes and contributes to contractile dysfunction induced by pressure overload. iScience 2023; 26:107146. [PMID: 37456848 PMCID: PMC10338234 DOI: 10.1016/j.isci.2023.107146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 03/31/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Non-canonical Wnt signaling activated by Wnt5a/Wnt11 is required for the second heart field development in mice. However, the pathophysiological role of non-canonical Wnt signaling in the adult heart has not been fully elucidated. Here we show that cardiomyocyte-specific Wnt5a knockout mice exhibit improved systolic function and reduced expression of mechanosensitive genes including Nppb when subjected to pressure overload. In cultured cardiomyocytes, Wnt5a knockdown reduced Nppb upregulation induced by cyclic cell stretch. Upstream analysis revealed that TEAD1, a transcription factor that acts with Hippo pathway co-activator YAP, was downregulated both in vitro and in vivo by Wnt5a knockdown/knockout. YAP nuclear translocation was induced by cell stretch and attenuated by Wnt5a knockdown. Wnt5a knockdown-induced Nppb downregulation during cell stretch was rescued by Hippo inhibition, and the rescue effect was canceled by knockdown of YAP. These results collectively suggest that Wnt5a-YAP signaling axis mediates mechanotransduction in cardiomyocytes and contributes to heart failure progression.
Collapse
Affiliation(s)
- Hiroshi Kishimoto
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Masayoshi Iwasaki
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Kensaku Wada
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Keita Horitani
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| | - Osamu Tsukamoto
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Kenta Kamikubo
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Seitaro Nomura
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shinji Matsumoto
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Takeshi Harada
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Seiji Takashima
- Department of Medical Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Akira Kikuchi
- Department of Molecular Biology and Biochemistry, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka 565-0871, Japan
| | - Ichiro Shiojima
- Department of Medicine II, Kansai Medical University, Osaka 573-1010, Japan
| |
Collapse
|
14
|
Bekedam FT, Goumans MJ, Bogaard HJ, de Man FS, Llucià-Valldeperas A. Molecular mechanisms and targets of right ventricular fibrosis in pulmonary hypertension. Pharmacol Ther 2023; 244:108389. [PMID: 36940790 DOI: 10.1016/j.pharmthera.2023.108389] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/19/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023]
Abstract
Right ventricular fibrosis is a stress response, predominantly mediated by cardiac fibroblasts. This cell population is sensitive to increased levels of pro-inflammatory cytokines, pro-fibrotic growth factors and mechanical stimulation. Activation of fibroblasts results in the induction of various molecular signaling pathways, most notably the mitogen-activated protein kinase cassettes, leading to increased synthesis and remodeling of the extracellular matrix. While fibrosis confers structural protection in response to damage induced by ischemia or (pressure and volume) overload, it simultaneously contributes to increased myocardial stiffness and right ventricular dysfunction. Here, we review state-of-the-art knowledge of the development of right ventricular fibrosis in response to pressure overload and provide an overview of all published preclinical and clinical studies in which right ventricular fibrosis was targeted to improve cardiac function.
Collapse
Affiliation(s)
- F T Bekedam
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - M J Goumans
- Department of Cell and Chemical Biology, Leiden UMC, 2300 RC Leiden, the Netherlands
| | - H J Bogaard
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands
| | - F S de Man
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| | - A Llucià-Valldeperas
- Amsterdam UMC location Vrije Universiteit Amsterdam, PHEniX laboratory, Department of Pulmonary Medicine, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences, Pulmonary Hypertension and Thrombosis, Amsterdam, the Netherlands.
| |
Collapse
|
15
|
Wan S, Cui Z, Wu L, Zhang F, Liu T, Hu J, Tian J, Yu B, Liu F, Kou J, Li F. Ginsenoside Rd promotes omentin secretion in adipose through TBK1-AMPK to improve mitochondrial biogenesis via WNT5A/Ca 2+ pathways in heart failure. Redox Biol 2023; 60:102610. [PMID: 36652744 PMCID: PMC9860421 DOI: 10.1016/j.redox.2023.102610] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Ginsenoside Rd is an active ingredient in Panax ginseng CA Mey and can be absorbed into the adipose tissue. Adipokines play an important role in the treatment of cardiovascular diseases. However, the potential benefit of Rd on heart failure (HF) and the underlying mechanism associated with the crosstalk between adipocytes and cardiomyocytes remains to be illustrated. Here, the results identified that Rd improved cardiac function and inhibited cardiac pathological changes in transverse aortic constriction (TAC), coronary ligation (CAL) and isoproterenol (ISO)-induced HF mice. And Rd promoted the release of omentin from the adipose tissue and up-regulated omentin expression in lipopolysaccharide (LPS)-induced 3T3-L1 adipocytes. Further, Rd could increase TBK1 and AMPK phosphorylation in adipocytes. And also, the TBK1-AMPK signaling pathway regulated the expression of omentin in LPS-induced adipocytes. Moreover, the omentin mRNA expression was significantly decreased by TBK1 knockdown in LPS-induced 3T3-L1 adipocytes. Additionally, molecular docking and SPR analysis confirmed that Rd had a certain binding ability with TBK1, and co-treatment with TBK1 inhibitors or TBK1 knockdown partially abolished the effect of Rd on increasing the omentin expression and the ratio of p-AMPK to AMPK in adipocytes. Moreover, we found that circulating omentin level diminished in the HF patients compared with healthy subjects. Meanwhile, the adipose tissue-specific overexpression of omentin improved cardiac function, reduced myocardial infarct size and ameliorated cardiac pathological features in CAL-induced HF mice. Consistently, exogenous omentin reduced mtROS levels and restored ΔψM to improve oxygen and glucose deprivation (OGD)-induced cardiomyocytes injury. Further, omentin inhibited the WNT5A/Ca2+ signaling pathway and promoted mitochondrial biogenesis function to ameliorate myocardial ischemia injury. However, WNT5A knockdown inhibited the impairment of mitochondrial biogenesis and partially counteracted the cardioprotective effect of omentin in vitro. Therefore, this study indicated that Rd promoted omentin secretion from adipocytes through the TBK1-AMPK pathway to improve mitochondrial biogenesis function via WNT5A/Ca2+ signaling pathway to ameliorate myocardial ischemia injury, which provided a new therapeutic mechanism and potential drugs for the treatment of HF.
Collapse
Affiliation(s)
- Shiyao Wan
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - ZeKun Cui
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Lingling Wu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Fan Zhang
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Tao Liu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jingui Hu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiangwei Tian
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Boyang Yu
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Fuming Liu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Junping Kou
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Fang Li
- Jiangsu Key Laboratory of TCM Evaluation and Translational Research, Research Center for Traceability and Standardization of TCMs, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
16
|
Sutton NR, Malhotra R, Hilaire C, Aikawa E, Blumenthal RS, Gackenbach G, Goyal P, Johnson A, Nigwekar SU, Shanahan CM, Towler DA, Wolford BN, Chen Y. Molecular Mechanisms of Vascular Health: Insights From Vascular Aging and Calcification. Arterioscler Thromb Vasc Biol 2023; 43:15-29. [PMID: 36412195 PMCID: PMC9793888 DOI: 10.1161/atvbaha.122.317332] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/11/2022] [Indexed: 11/23/2022]
Abstract
Cardiovascular disease is the most common cause of death worldwide, especially beyond the age of 65 years, with the vast majority of morbidity and mortality due to myocardial infarction and stroke. Vascular pathology stems from a combination of genetic risk, environmental factors, and the biologic changes associated with aging. The pathogenesis underlying the development of vascular aging, and vascular calcification with aging, in particular, is still not fully understood. Accumulating data suggests that genetic risk, likely compounded by epigenetic modifications, environmental factors, including diabetes and chronic kidney disease, and the plasticity of vascular smooth muscle cells to acquire an osteogenic phenotype are major determinants of age-associated vascular calcification. Understanding the molecular mechanisms underlying genetic and modifiable risk factors in regulating age-associated vascular pathology may inspire strategies to promote healthy vascular aging. This article summarizes current knowledge of concepts and mechanisms of age-associated vascular disease, with an emphasis on vascular calcification.
Collapse
Affiliation(s)
- Nadia R. Sutton
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Rajeev Malhotra
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Cynthia Hilaire
- Division of Cardiology, Departments of Medicine and Bioengineering, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, 1744 BSTWR, 200 Lothrop St, Pittsburgh, PA, 15260 USA
| | - Elena Aikawa
- Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA USA
| | - Roger S. Blumenthal
- Johns Hopkins Ciccarone Center for the Prevention of Cardiovascular Disease; Baltimore, MD
| | - Grace Gackenbach
- Division of Cardiovascular Medicine, Michigan Medicine, Ann Arbor, Michigan, USA
| | - Parag Goyal
- Department of Medicine, Weill Cornell Medicine, New York, NY
| | - Adam Johnson
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Sagar U. Nigwekar
- Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, MA USA
| | - Catherine M. Shanahan
- School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London, UK
| | - Dwight A. Towler
- Department of Medicine | Endocrine Division and Pak Center for Mineral Metabolism Research, UT Southwestern Medical Center, Dallas, TX USA
| | - Brooke N. Wolford
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham and Research Department, Veterans Affairs Birmingham Medical Center, Birmingham, AL, USA
| |
Collapse
|
17
|
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: 72] [Impact Index Per Article: 24.0] [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
|
18
|
Liu J, Zhang R, Wang D, Lin Y, Bai C, Nie N, Gao S, Zhang Q, Chang H, Ren C. Elucidating the role of circNFIB in myocardial fibrosis alleviation by endogenous sulfur dioxide. BMC Cardiovasc Disord 2022; 22:492. [PMID: 36404310 PMCID: PMC9677687 DOI: 10.1186/s12872-022-02909-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 10/21/2022] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND To investigate the role of circNFIB in the alleviation of myocardial fibrosis by endogenous sulfur dioxide (SO2). METHODS We stimulated cultured neonatal rat cardiac fibroblasts with transforming growth factor-β1 (TGF-β1) and developed an in vitro myocardial fibrosis model. Lentivirus vectors containing aspartate aminotransferase 1 (AAT1) cDNA were used to overexpress AAT1, and siRNA was used to silence circNFIB. The SO2, collagen, circNFIB, Wnt/β-catenin, and p38 MAPK pathways were examined in each group. RESULTS In the in vitro TGF-β1-induced myocardial fibrosis model, endogenous SO2/AAT1 expression was significantly decreased, and collagen levels in the cell supernatant and type I and III collagen expression, as well as α-SMA expression, were all significantly increased. TGF-β1 also significantly reduced circNFIB expression. AAT1 overexpression significantly reduced myocardial fibrosis while significantly increasing circNFIB expression. Endogenous SO2 alleviated myocardial fibrosis after circNFIB expression was blocked. We discovered that circNFIB plays an important role in the alleviation of myocardial fibrosis by endogenous SO2 by inhibiting the Wnt/β-catenin and p38 MAPK pathways. CONCLUSION Endogenous SO2 promotes circNFIB expression, which inhibits the Wnt/β-catenin and p38 MAPK signaling pathways, consequently alleviating myocardial fibrosis.
Collapse
Affiliation(s)
- Jia Liu
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ranran Zhang
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dahai Wang
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yi Lin
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cui Bai
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Nana Nie
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shan Gao
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qiuye Zhang
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Chang
- grid.412521.10000 0004 1769 1119Department of pediatric nephrology and rheumotology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chongmin Ren
- grid.412521.10000 0004 1769 1119Department of orthopedic oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Cardiovascular diseases are the leading cause of death worldwide, largely due to the limited regenerative capacity of the adult human heart. In contrast, teleost zebrafish hearts possess natural regeneration capacity by proliferation of pre-existing cardiomyocytes after injury. Hearts of mice can regenerate if injured in a few days after birth, which coincides with the transient capacity for cardiomyocyte proliferation. This review tends to elaborate the roles and mechanisms of Wnt/β-catenin signaling in heart development and regeneration in mammals and non-mammalian vertebrates. RECENT FINDINGS Studies in zebrafish, mice, and human embryonic stem cells demonstrate the binary effect for Wnt/β-catenin signaling during heart development. Both Wnts and Wnt antagonists are induced in multiple cell types during cardiac development and injury repair. In this review, we summarize composites of the Wnt signaling pathway and their different action routes, followed by the discussion of their involvements in cardiac specification, proliferation, and patterning. We provide overviews about canonical and non-canonical Wnt activity during heart homeostasis, remodeling, and regeneration. Wnt/β-catenin signaling exhibits biphasic and antagonistic effects on cardiac specification and differentiation depending on the stage of embryogenesis. Inhibition of Wnt signaling is beneficial for cardiac wound healing and functional recovery after injury. Understanding of the roles and mechanisms of Wnt signaling pathway in injured animal hearts will contribute to the development of potential therapeutics for human diseased hearts.
Collapse
Affiliation(s)
- Dongliang Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jianjian Sun
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China.,Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510100, Guangdong, China
| | - Tao P Zhong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Molecular Medicine, School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
20
|
Effect of Secreted Frizzled-Related Protein 5 in Mice with Heart Failure. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1606212. [PMID: 35685732 PMCID: PMC9173992 DOI: 10.1155/2022/1606212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 05/20/2022] [Indexed: 11/18/2022]
Abstract
Although some progress has been made in its treatment, heart failure is still one of the most important health problems that endanger public health. This study aims to explore the myocardial protective effect of secreted frizzled-related protein 5 (SFRP5) on mice with heart failure. The mouse model of heart failure was established by using the isoproterenol (ISO) hydrochloride gradient modeling method. The treatment group was injected with 0.02 mg/kg/24 h SFRP5 recombinant protein intraperitoneally 30 minutes after the injection of isoproterenol, and the ISO + phosphate-buffered saline (PBS) group was injected with the same amount of PBS. After intraperitoneal injection of SFRP5 recombinant protein in mice with heart failure, the inflammatory response was reduced, and the left ventricular systolic and diastolic function of heart failure mice and the pathological structure of the myocardial tissue were improved. Compared with the ISO group, the expression level of SFRP5 protein in the ISO + SFRP5 group was increased significantly, the expression levels of Wnt5a and JNK protein were decreased markedly, and the enzyme activities of SOD and GSH-Px in the serum were observably increased, but they were lower than those parameters in the normal group. The SFRP5 recombinant protein has a protective effect on isoproterenol-induced heart failure in mice. The mechanism of action may be related to inhibiting the Wnt5A/JNK signaling pathway and reducing oxidative stress and inflammation. SFRP5 may be one of the therapeutic targets of heart failure.
Collapse
|
21
|
Lin T, Tao J, Chen Y, Zhang Y, Li F, Zhang Y, Han X, Zhao Z, Liu G, Li H. Selenium Deficiency Leads to Changes in Renal Fibrosis Marker Proteins and Wnt/β-Catenin Signaling Pathway Components. Biol Trace Elem Res 2022; 200:1127-1139. [PMID: 33895963 DOI: 10.1007/s12011-021-02730-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 04/18/2021] [Indexed: 01/03/2023]
Abstract
Renal fibrosis is the final result of the progression of chronic kidney disease (CKD) to end-stage renal disease (ESRD). Earlier studies confirmed that selenium (Se) displays a close association with kidney diseases. However, the correlation between Se and fibrosis has rarely been explored. Thus, this article mainly aimed to investigate the effect of Se deficiency on renal fibrosis and the Wnt/β-catenin signaling pathway. Twenty BALB/c mice were fed a diet containing 0.02-mg/kg Se (Se-deficient diet) or 0.18-mg/kg Se (standard diet) for 20 weeks. A human glomerular mesangial cell (HMC) cell line was transfected with lentiviral TRNAU1AP-shRNA vector to establish a stable Se deficiency model in vitro. As indicated in this study, the glutathione (GSH) content in the Se-deficient group displayed an obvious decline compared with that in the control group, whereas the content of malondialdehyde (MDA) was obviously elevated. The results of Masson staining showed fibrosis around the renal tubules, and the results of immunohistochemistry showed that the area of positive fibronectin expression increased. In the Se-deficient group, the levels of collagen I, collagen III, matrix metalloproteinase 9 (MMP9), and other fibrosis-related proteins changed significantly in vivo and in vitro. Compared with the control group, the TRNAU1AP-shRNA group showed markedly reduced cell proliferation and migration abilities. Our data indicate that Se deficiency can cause kidney damage and renal fibrosis. Furthermore, the Wnt pathway is critical for the development of tissue and organ fibrosis. The data of this study demonstrated that the expression of Wnt5a, β-catenin, and dishevelled 1 (Dvl-1) was significantly upregulated in the Se-deficient group. Therefore, the Wnt/β-catenin pathway may play an important role in renal fibrosis caused by Se deficiency.
Collapse
Affiliation(s)
- Tingting Lin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Jiaqi Tao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Ying Chen
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Yitong Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Fenglan Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Yutong Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Xueqing Han
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Zihui Zhao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Guiyan Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China
| | - Hui Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Baojian Road 157, Nangang District, Harbin City, 150086, Heilongjiang, China.
| |
Collapse
|
22
|
Biogenesis and Function of Extracellular Vesicles in Pathophysiological Processes Skeletal Muscle Atrophy. Biochem Pharmacol 2022; 198:114954. [DOI: 10.1016/j.bcp.2022.114954] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022]
|
23
|
Reid LV, Spalluto CM, Watson A, Staples KJ, Wilkinson TMA. The Role of Extracellular Vesicles as a Shared Disease Mechanism Contributing to Multimorbidity in Patients With COPD. Front Immunol 2021; 12:754004. [PMID: 34925327 PMCID: PMC8675939 DOI: 10.3389/fimmu.2021.754004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/04/2021] [Indexed: 01/27/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is one of the leading causes of death worldwide. Individuals with COPD typically experience a progressive, debilitating decline in lung function as well as systemic manifestations of the disease. Multimorbidity, is common in COPD patients and increases the risk of hospitalisation and mortality. Central to the genesis of multimorbidity in COPD patients is a self-perpetuating, abnormal immune and inflammatory response driven by factors including ageing, pollutant inhalation (including smoking) and infection. As many patients with COPD have multiple concurrent chronic conditions, which require an integrative management approach, there is a need to greater understand the shared disease mechanisms contributing to multimorbidity. The intercellular transfer of extracellular vesicles (EVs) has recently been proposed as an important method of local and distal cell-to-cell communication mediating both homeostatic and pathological conditions. EVs have been identified in many biological fluids and provide a stable capsule for the transfer of cargo including proteins, lipids and nucleic acids. Of these cargo, microRNAs (miRNAs), which are short 17-24 nucleotide non-coding RNA molecules, have been amongst the most extensively studied. There is evidence to support that miRNA are selectively packaged into EVs and can regulate recipient cell gene expression including major pathways involved in inflammation, apoptosis and fibrosis. Furthermore changes in EV cargo including miRNA have been reported in many chronic diseases and in response to risk factors including respiratory infections, noxious stimuli and ageing. In this review, we discuss the potential of EVs and EV-associated miRNA to modulate shared pathological processes in chronic diseases. Further delineating these may lead to the identification of novel biomarkers and therapeutic targets for patients with COPD and multimorbidities.
Collapse
Affiliation(s)
- Laura V Reid
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - C Mirella Spalluto
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Alastair Watson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom.,Birmingham Medical School, University of Birmingham, Birmingham, United Kingdom
| | - Karl J Staples
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| | - Tom M A Wilkinson
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,National Institute for Health Research Southampton Biomedical Research Centre, Southampton Centre for Biomedical Research, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
24
|
Zhang S, Liu H, Fang Q, He H, Lu X, Wang Y, Fan X. Shexiang Tongxin Dropping Pill Protects Against Chronic Heart Failure in Mice via Inhibiting the ERK/MAPK and TGF-β Signaling Pathways. Front Pharmacol 2021; 12:796354. [PMID: 34925046 PMCID: PMC8682969 DOI: 10.3389/fphar.2021.796354] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Chronic heart failure (CHF) is a major public health problem with high mortality and morbidity worldwide. Shexiang Tongxin Dropping Pill (STDP) is a widely used traditional Chinese medicine preparation for coronary heart disease and growing evidence proves that STDP exerts beneficial effects on CHF in the clinic. However, the molecular mechanism of the therapeutic effects of STDP on CHF remains largely unknown. Objective: This study aimed to elucidate the mechanism of action of STDP against CHF by integrating network pharmacology analysis and whole-transcriptome sequencing. Methods: First, the mouse model of CHF was established by the transverse aortic constriction (TAC) surgery, and the efficacy of STDP against CHF was evaluated by assessing the alterations in cardiac function, myocardial fibrosis, and cardiomyocyte hypertrophy with echocardiography, Masson’s trichrome staining, and wheat germ agglutinin staining. Next, a CHF disease network was constructed by integrating cardiovascular disease-related genes and the transcriptome sequencing data, which was used to explore the underlying mechanism of action of STDP. Then, the key targets involved in the effects of STDP on CHF were determined by network analysis algorithms, and pathway enrichment analysis was performed to these key genes. Finally, important targets in critical pathway were verified in vivo. Results: STDP administration obviously improved cardiac function, relieved cardiomyocyte hypertrophy, and ameliorated myocardial fibrosis in CHF mice. Moreover, STDP significantly reversed the imbalanced genes that belong to the disease network of CHF in mice with TAC, and the number of genes with the reverse effect was 395. Pathway analysis of the crucial genes with recovery efficiency revealed that pathways related to fibrosis and energy metabolism were highly enriched, while TGF-β pathway and ERK/MAPK pathway were predicted to be significantly affected. Consistently, validation experiments confirmed that inhibiting ERK/MAPK and TGF-β signaling pathways via reduction of the phosphorylation level of Smad3 and ERK1/2 is the important mechanism of STDP against CHF. Conclusion: Our data demonstrated that STDP can recover the imbalanced CHF network disturbed by the modeling of TAC through the multi-target and multi-pathway manner in mice, and the mechanisms are mainly related to inhibition of ERK/MAPK and TGF-β signaling pathways.
Collapse
Affiliation(s)
- Shuying Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hanbing Liu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianqian Fang
- Inner Mongolia Conba Pharmaceutical Co., Ltd., Hohhot, China
| | - Houhong He
- Zhejiang Conba Pharmaceutical Co., Ltd., Hangzhou, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Yi Wang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaohui Fan
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| |
Collapse
|
25
|
Yin C, Ye Z, Wu J, Huang C, Pan L, Ding H, Zhong L, Guo L, Zou Y, Wang X, Wang Y, Gao P, Jin X, Yan X, Zou Y, Huang R, Gong H. Elevated Wnt2 and Wnt4 activate NF-κB signaling to promote cardiac fibrosis by cooperation of Fzd4/2 and LRP6 following myocardial infarction. EBioMedicine 2021; 74:103745. [PMID: 34911029 PMCID: PMC8669316 DOI: 10.1016/j.ebiom.2021.103745] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 11/25/2021] [Accepted: 11/26/2021] [Indexed: 11/29/2022] Open
Abstract
Background Acute myocardial infarction (AMI)-induced excessive myocardial fibrosis exaggerates cardiac dysfunction. However, serum Wnt2 or Wnt4 level in AMI patients, and the roles in cardiac fibrosis are largely unkown. Methods AMI and non-AMI patients were enrolled to examine serum Wnt2 and Wnt4 levels by ELISA analysis. The AMI patients were followed-up for one year. MI mouse model was built by ligation of left anterior descending branch (LAD). Findings Serum Wnt2 or Wnt4 level was increased in patients with AMI, and the elevated Wnt2 and Wnt4 were correlated to adverse outcome of these patients. Knockdown of Wnt2 and Wnt4 significantly attenuated myocardial remodeling and cardiac dysfunction following experimental MI. In vitro, hypoxia enhanced the secretion and expression of Wnt2 and Wnt4 in neonatal rat cardiac myocytes (NRCMs) or fibroblasts (NRCFs). Mechanistically, the elevated Wnt2 or Wnt4 activated β-catenin /NF-κB signaling to promote pro-fibrotic effects in cultured NRCFs. In addition, Wnt2 or Wnt4 upregulated the expression of these Wnt co-receptors, frizzled (Fzd) 2, Fzd4 and (ow-density lipoprotein receptor-related protein 6 (LRP6). Further analysis revealed that Wnt2 or Wnt4 activated β-catenin /NF-κB by the co-operation of Fzd4 or Fzd2 and LRP6 signaling, respectively. Interpretation Elevated Wnt2 and Wnt4 activate β-catenin/NF-κB signaling to promote cardiac fibrosis by cooperation of Fzd4/2 and LRP6 in fibroblasts, which contributes to adverse outcome of patients with AMI, suggesting that systemic inhibition of Wnt2 and Wnt4 may improve cardiac dysfunction after MI.
Collapse
Affiliation(s)
- Chao Yin
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Zhishuai Ye
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100053, China
| | - Jian Wu
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Chenxing Huang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Le Pan
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Huaiyu Ding
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Zhong
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Guo
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yan Zou
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiang Wang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Ying Wang
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Pan Gao
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xuejuan Jin
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoxiang Yan
- Department of Vascular and Cardiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunzeng Zou
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Rongchong Huang
- Department of Cardiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100053, China.
| | - Hui Gong
- NHC Key Laboratory of Viral Heart Diseases, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
26
|
Zou Y, Pan L, Shen Y, Wang X, Huang C, Wang H, Jin X, Yin C, Wang Y, Jia J, Qian J, Zou Y, Gong H, Ge J. Cardiac Wnt5a and Wnt11 promote fibrosis by the crosstalk of FZD5 and EGFR signaling under pressure overload. Cell Death Dis 2021; 12:877. [PMID: 34564708 PMCID: PMC8464604 DOI: 10.1038/s41419-021-04152-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/20/2021] [Accepted: 09/09/2021] [Indexed: 12/16/2022]
Abstract
Progressive cardiac fibrosis accelerates the development of heart failure. Here, we aimed to explore serum Wnt5a and Wnt11 levels in hypertension patients, the roles of Wnt5a and Wnt11 in cardiac fibrosis and potential mechanisms under pressure overload. The pressure overload mouse model was built by transverse aortic constriction (TAC). Cardiac fibrosis was analyzed by Masson's staining. Serum Wnt5a or Wnt11 was elevated and associated with diastolic dysfunction in hypertension patients. TAC enhanced the expression and secretion of Wnt5a or Wnt11 from cardiomyocytes (CMs), cardiac fibroblasts (CFs), and cardiac microvascular endothelial cells (CMECs). Knockdown of Wnt5a and Wnt11 greatly improved cardiac fibrosis and function at 4 weeks after TAC. In vitro, shWnt5a or shWnt11 lentivirus transfection inhibited pro-fibrotic effects in CFs under mechanical stretch (MS). Similarly, conditional medium from stretched-CMs transfected with shWnt5a or shWnt11 lentivirus significantly suppressed the pro-fibrotic effects induced by conditional medium from stretched-CMs. These data suggested that CMs- or CFs-derived Wnt5a or Wnt11 showed a pro-fibrotic effect under pressure overload. In vitro, exogenous Wnt5a or Wnt11 activated ERK and p38 (fibrotic-related signaling) pathway, promoted the phosphorylation of EGFR, and increased the expression of Frizzled 5 (FZD5) in CFs. Inhibition or knockdown of EGFR greatly attenuated the increased FZD5, p-p38, and p-ERK levels, and the pro-fibrotic effect induced by Wnt5a or Wnt11 in CFs. Si-FZD5 transfection suppressed the increased p-EGFR level, and the fibrotic-related effects in CFs treated with Wnt5a or Wnt11. In conclusion, pressure overload enhances the secretion of Wnt5a or Wnt11 from CMs and CFs which promotes cardiac fibrosis by activation the crosstalk of FZD5 and EGFR. Thus, Wnt5a or Wnt11 may be a novel therapeutic target for the prevention of cardiac fibrosis under pressure overload.
Collapse
Affiliation(s)
- Yan Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Le Pan
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yi Shen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xiang Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenxing Huang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Hao Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Xuejuan Jin
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chao Yin
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ying Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jianguo Jia
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Juying Qian
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Hui Gong
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| | - Junbo Ge
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
27
|
Działo E, Czepiel M, Tkacz K, Siedlar M, Kania G, Błyszczuk P. WNT/β-Catenin Signaling Promotes TGF-β-Mediated Activation of Human Cardiac Fibroblasts by Enhancing IL-11 Production. Int J Mol Sci 2021; 22:ijms221810072. [PMID: 34576234 PMCID: PMC8468519 DOI: 10.3390/ijms221810072] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiac fibrosis is a pathological process associated with the development of heart failure. TGF-β and WNT signaling have been implicated in pathogenesis of cardiac fibrosis, however, little is known about molecular cross-talk between these two pathways. The aim of this study was to examine the effect of exogenous canonical WNT3a and non-canonical WNT5a in TGF-β-activated human cardiac fibroblasts. We found that WNT3a and TGF-β induced a β-catenin-dependent response, whereas WNT5a prompted AP-1 activity. TGF-β triggered profibrotic signatures in cardiac fibroblasts, and co-stimulation with WNT3a or co-activation of the β-catenin pathway with the GSK3β inhibitor CHIR99021 enhanced collagen I and fibronectin production and development of active contractile stress fibers. In the absence of TGF-β, neither WNT3a nor CHIR99021 exerted profibrotic responses. On a molecular level, in TGF-β-activated fibroblasts, WNT3a enhanced phosphorylation of TAK1 and production and secretion of IL-11 but showed no effect on the Smad pathway. Neutralization of IL-11 activity with the blocking anti-IL-11 antibody effectively reduced the profibrotic response of cardiac fibroblasts activated with TGF-β and WNT3a. In contrast to canonical WNT3a, co-activation with non-canonical WNT5a suppressed TGF-β-induced production of collagen I. In conclusion, WNT/β-catenin signaling promotes TGF-β-mediated fibroblast-to-myofibroblast transition by enhancing IL-11 production. Thus, the uncovered mechanism broadens our knowledge on a molecular basis of cardiac fibrogenesis and defines novel therapeutic targets for fibrotic heart diseases.
Collapse
Affiliation(s)
- Edyta Działo
- Department of Clinical Immunology, Jagiellonian University Medical College, 30-663 Cracow, Poland; (E.D.); (M.C.); (K.T.); (M.S.)
| | - Marcin Czepiel
- Department of Clinical Immunology, Jagiellonian University Medical College, 30-663 Cracow, Poland; (E.D.); (M.C.); (K.T.); (M.S.)
| | - Karolina Tkacz
- Department of Clinical Immunology, Jagiellonian University Medical College, 30-663 Cracow, Poland; (E.D.); (M.C.); (K.T.); (M.S.)
| | - Maciej Siedlar
- Department of Clinical Immunology, Jagiellonian University Medical College, 30-663 Cracow, Poland; (E.D.); (M.C.); (K.T.); (M.S.)
| | - Gabriela Kania
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland;
| | - Przemysław Błyszczuk
- Department of Clinical Immunology, Jagiellonian University Medical College, 30-663 Cracow, Poland; (E.D.); (M.C.); (K.T.); (M.S.)
- Center of Experimental Rheumatology, Department of Rheumatology, University Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland;
- Correspondence: ; Tel.: +48-12-658-24-86
| |
Collapse
|
28
|
Valizadeh A, Asghari S, Mansouri P, Alemi F, Majidinia M, Mahmoodpoor A, Yousefi B. The roles of signaling pathways in cardiac regeneration. Curr Med Chem 2021; 29:2142-2166. [PMID: 34521319 DOI: 10.2174/0929867328666210914115411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/05/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
In recent years, knowledge of cardiac regeneration mechanisms has dramatically expanded. Regeneration can replace lost parts of organs, common among animal species. The heart is commonly considered an organ with terminal development, which has no reparability potential during post-natal life; however, some intrinsic regeneration capacity has been reported for cardiac muscle, which opens novel avenues in cardiovascular disease treatment. Different endogenous mechanisms were studied for cardiac repairing and regeneration in recent decades. Survival, proliferation, inflammation, angiogenesis, cell-cell communication, cardiomyogenesis, and anti-aging pathways are the most important mechanisms that have been studied in this regard. Several in vitro and animal model studies focused on proliferation induction for cardiac regeneration reported promising results. These studies have mainly focused on promoting proliferation signaling pathways and demonstrated various signaling pathways such as Wnt, PI3K/Akt, IGF-1, TGF-β, Hippo, and VEGF signaling cardiac regeneration. Therefore, in this review, we intended to discuss the connection between different critical signaling pathways in cardiac repair and regeneration.
Collapse
Affiliation(s)
- Amir Valizadeh
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Samira Asghari
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Parinaz Mansouri
- Students Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Forough Alemi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Urmia University of Medical Sciences, Urmia. Iran
| | - Ata Mahmoodpoor
- Stem Cell and Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Bahman Yousefi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
29
|
Jiang W, Xiong Y, Li X, Yang Y. Cardiac Fibrosis: Cellular Effectors, Molecular Pathways, and Exosomal Roles. Front Cardiovasc Med 2021; 8:715258. [PMID: 34485413 PMCID: PMC8415273 DOI: 10.3389/fcvm.2021.715258] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/20/2021] [Indexed: 01/18/2023] Open
Abstract
Cardiac fibrosis, a common pathophysiologic process in most heart diseases, refers to an excess of extracellular matrix (ECM) deposition by cardiac fibroblasts (CFs), which can lead to cardiac dysfunction and heart failure subsequently. Not only CFs but also several other cell types including macrophages and endothelial cells participate in the process of cardiac fibrosis via different molecular pathways. Exosomes, ranging in 30-150 nm of size, have been confirmed to play an essential role in cellular communications by their bioactive contents, which are currently a hot area to explore pathobiology and therapeutic strategy in multiple pathophysiologic processes including cardiac fibrosis. Cardioprotective factors such as RNAs and proteins packaged in exosomes make them an excellent cell-free system to improve cardiac function without significant immune response. Emerging evidence indicates that targeting selective molecules in cell-derived exosomes could be appealing therapeutic treatments in cardiac fibrosis. In this review, we summarize the current understandings of cellular effectors, molecular pathways, and exosomal roles in cardiac fibrosis.
Collapse
Affiliation(s)
- Wenyang Jiang
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuyan Xiong
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaosong Li
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
30
|
Chavkin NW, Sano S, Wang Y, Oshima K, Ogawa H, Horitani K, Sano M, MacLauchlan S, Nelson A, Setia K, Vippa T, Watanabe Y, Saucerman JJ, Hirschi KK, Gokce N, Walsh K. The Cell Surface Receptors Ror1/2 Control Cardiac Myofibroblast Differentiation. J Am Heart Assoc 2021; 10:e019904. [PMID: 34155901 PMCID: PMC8403294 DOI: 10.1161/jaha.120.019904] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/22/2021] [Indexed: 12/25/2022]
Abstract
Background A hallmark of heart failure is cardiac fibrosis, which results from the injury-induced differentiation response of resident fibroblasts to myofibroblasts that deposit extracellular matrix. During myofibroblast differentiation, fibroblasts progress through polarization stages of early proinflammation, intermediate proliferation, and late maturation, but the regulators of this progression are poorly understood. Planar cell polarity receptors, receptor tyrosine kinase-like orphan receptor 1 and 2 (Ror1/2), can function to promote cell differentiation and transformation. In this study, we investigated the role of the Ror1/2 in a model of heart failure with emphasis on myofibroblast differentiation. Methods and Results The role of Ror1/2 during cardiac myofibroblast differentiation was studied in cell culture models of primary murine cardiac fibroblast activation and in knockout mouse models that underwent transverse aortic constriction surgery to induce cardiac injury by pressure overload. Expression of Ror1 and Ror2 were robustly and exclusively induced in fibroblasts in hearts after transverse aortic constriction surgery, and both were rapidly upregulated after early activation of primary murine cardiac fibroblasts in culture. Cultured fibroblasts isolated from Ror1/2 knockout mice displayed a proinflammatory phenotype indicative of impaired myofibroblast differentiation. Although the combined ablation of Ror1/2 in mice did not result in a detectable baseline phenotype, transverse aortic constriction surgery led to the death of all mice by day 6 that was associated with myocardial hyperinflammation and vascular leakage. Conclusions Together, these results show that Ror1/2 are essential for the progression of myofibroblast differentiation and for the adaptive remodeling of the heart in response to pressure overload.
Collapse
Affiliation(s)
- Nicholas W. Chavkin
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Cell BiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Soichi Sano
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
- Department of CardiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Ying Wang
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyXinqiao HospitalArmy Medical UniversityChongqingChina
| | - Kosei Oshima
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Hayato Ogawa
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Keita Horitani
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Miho Sano
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologyGraduate School of MedicineOsaka City UniversityOsakaJapan
| | - Susan MacLauchlan
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Anders Nelson
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of PharmacologyUniversity of VirginiaCharlottesvilleVA
| | - Karishma Setia
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Tanvi Vippa
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
| | - Yosuke Watanabe
- Vascular Biology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
| | - Jeffrey J. Saucerman
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVA
| | - Karen K. Hirschi
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Department of Cell BiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Cardiovascular Research CenterSchool of MedicineYale UniversityNew HavenCT
| | - Noyan Gokce
- Boston University School of MedicineBostonMA
| | - Kenneth Walsh
- Cardiovascular Research CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Hematovascular Biology CenterSchool of MedicineUniversity of VirginiaCharlottesvilleVA
- Molecular Cardiology/Whitaker Cardiovascular InstituteBoston University School of MedicineBostonMA
- Department of CardiologySchool of MedicineUniversity of VirginiaCharlottesvilleVA
| |
Collapse
|
31
|
Skaria T, Bachli E, Schoedon G. Transcriptional Regulation of Drug Metabolizing CYP Enzymes by Proinflammatory Wnt5A Signaling in Human Coronary Artery Endothelial Cells. Front Pharmacol 2021; 12:619588. [PMID: 34079452 PMCID: PMC8165381 DOI: 10.3389/fphar.2021.619588] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 03/29/2021] [Indexed: 12/31/2022] Open
Abstract
Downregulation of drug metabolizing enzymes and transporters by proinflammatory mediators in hepatocytes, enterocytes and renal tubular epithelium is an established mechanism affecting pharmacokinetics. Emerging evidences indicate that vascular endothelial cell expression of drug metabolizing enzymes and transporters may regulate pharmacokinetic pathways in heart to modulate local drug bioavailability and toxicity. However, whether inflammation regulates pharmacokinetic pathways in human cardiac vascular endothelial cells remains largely unknown. The lipid modified protein Wnt5A is emerging as a critical mediator of proinflammatory responses and disease severity in sepsis, hypertension and COVID-19. In the present study, we employed transcriptome profiling and gene ontology analyses to investigate the regulation of expression of drug metabolizing enzymes and transporters by Wnt5A in human coronary artery endothelial cells. Our study shows for the first time that Wnt5A induces the gene expression of CYP1A1 and CYP1B1 enzymes involved in phase I metabolism of a broad spectrum of drugs including chloroquine (the controversial drug for COVID-19) that is known to cause toxicity in myocardium. Further, the upregulation of CYP1A1 and CYP1B1 expression is preserved even during inflammatory crosstalk between Wnt5A and the prototypic proinflammatory IL-1β in human coronary artery endothelial cells. These findings stimulate further studies to test the critical roles of vascular endothelial cell CYP1A1 and CYP1B1, and the potential of vascular-targeted therapy with CYP1A1/CYP1B1 inhibitors in modulating myocardial pharmacokinetics in Wnt5A-associated inflammatory and cardiovascular diseases.
Collapse
Affiliation(s)
- Tom Skaria
- Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich, Zürich, Switzerland.,School of Biotechnology, National Institute of Technology Calicut, Kerala, India
| | - Esther Bachli
- Department of Medicine, Uster Hospital, Uster, Switzerland
| | - Gabriele Schoedon
- Inflammation Research Unit, Division of Internal Medicine, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
32
|
Ueland T, Abraityte A, Norum H, Varathalingam S, Gullestad L, Aukrust P, Andreassen AK. Circulating regulators of the wingless pathway in precapillary pulmonary hypertension. Respirology 2021; 26:574-581. [PMID: 33830565 DOI: 10.1111/resp.14048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/18/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVE Dysregulated Wnt signalling has been implicated in pulmonary hypertension (PH). We hypothesized that plasma levels of secreted Wnt proteins would be increased in patients with precapillary PH, correlate with indices of vascular resistance and cardiac function and give information on long-term prognosis. METHODS We measured the Wnt ligand Wnt5a and secreted Wnt antagonists Dickkopf (DKK) DKK1, DKK3, secreted frizzled-related protein 3 (sFRP3), Wnt inhibitory factor-1 (WIF1) and sclerostin (SOST) in 106 patients with precapillary PH and 40 healthy controls. A second sample was obtained after a median of 4 months (n = 52). During a median of 90 months follow-up, 67 patients died. RESULTS Our main findings were (i) Precapillary PH is characterized by enhanced systemic Wnt activity as reflected by elevated plasma levels of Wnt5a and secreted antagonists irrespective of diagnostic subgroups. (ii) WIF1 and in particular Wnt5a correlated with pulmonary vascular resistance and cardiac dysfunction. (iii) High levels of Wnt5a, sFRP3, DKK3 and WIF1 were associated with poor prognosis in age- and sex-adjusted analysis (hazard ratios per log/SD change ~1.4) and for DKK3 after further adjustment with right arterial pressure, pulmonary oxygen saturation, cardiac index, N-terminal pro B-type natriuretic peptide and peak oxygen uptake (VO2 ). Finally, an elevation of Wnt5a and DKK3 during follow-up was independently associated with poor prognosis. CONCLUSION Our data indicate that Wnt signalling pathways could be implicated in the pathogenesis of precapillary PH, and that some of the Wnt-related molecules (i.e., Wnt5a and DKK3) should be further investigated in these patients.
Collapse
Affiliation(s)
- Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Aurelija Abraityte
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Hilde Norum
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Sharanga Varathalingam
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Lars Gullestad
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Center for Heart Failure Research, University of Oslo, Oslo, Norway.,Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K. G. Jebsen Cardiac Research Center, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway.,Faculty of Medicine, University of Oslo, Oslo, Norway.,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.,Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway.,K. G. Jebsen Inflammation Research Center, University of Oslo, Oslo, Norway
| | - Arne K Andreassen
- Department of Cardiology, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
33
|
Su X, Shen Y, Jin Y, Weintraub NL, Tang YL. Identification of critical molecular pathways involved in exosome-mediated improvement of cardiac function in a mouse model of muscular dystrophy. Acta Pharmacol Sin 2021; 42:529-535. [PMID: 32601364 PMCID: PMC8115234 DOI: 10.1038/s41401-020-0446-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 05/17/2020] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive disease characterized by skeletal muscle atrophy, respiratory failure, and cardiomyopathy. Our previous studies have shown that transplantation with allogeneic myogenic progenitor-derived exosomes (MPC-Exo) can improve cardiac function in X-linked muscular dystrophy (Mdx) mice. In the present study we explored the molecular mechanisms underlying this beneficial effect. We quantified gene expression in the hearts of two strains of Mdx mice (D2.B10-DmdMdx/J and Utrntm1Ked-DmdMdx/J). Two days after MPC-Exo or control treatment, we performed unbiased next-generation RNA-sequencing to identify differentially expressed genes (DEGs) in treated Mdx hearts. Venn diagrams show a set of 780 genes that were ≥2-fold upregulated, and a set of 878 genes that were ≥2-fold downregulated, in both Mdx strains following MPC-Exo treatment as compared with control. Gene ontology (GO) and protein-protein interaction (PPI) network analysis showed that these DEGs were involved in a variety of physiological processes and pathways with a complex connection. qRT-PCR was performed to verify the upregulated ATP2B4 and Bcl-2 expression, and downregulated IL-6, MAPK8 and Wnt5a expression in MPC-Exo-treated Mdx hearts. Western blot analysis verified the increased level of Bcl-2 and decreased level of IL-6 protein in MPC-Exo-treated Mdx hearts compared with control treatment, suggesting that anti-apoptotic and anti-inflammatory effects might be responsible for heart function improvement by MPC-Exo. Based on these findings, we believed that these DEGs might be therapeutic targets that can be explored to develop new strategies for treating DMD.
Collapse
Affiliation(s)
- Xuan Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yan Shen
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yue Jin
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Yao-Liang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
34
|
Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH, Myung P, Plikus MV, Nie Q. Inference and analysis of cell-cell communication using CellChat. Nat Commun 2021; 12:1088. [PMID: 33597522 PMCID: PMC7889871 DOI: 10.1038/s41467-021-21246-9] [Citation(s) in RCA: 3703] [Impact Index Per Article: 925.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023] Open
Abstract
Understanding global communications among cells requires accurate representation of cell-cell signaling links and effective systems-level analyses of those links. We construct a database of interactions among ligands, receptors and their cofactors that accurately represent known heteromeric molecular complexes. We then develop CellChat, a tool that is able to quantitatively infer and analyze intercellular communication networks from single-cell RNA-sequencing (scRNA-seq) data. CellChat predicts major signaling inputs and outputs for cells and how those cells and signals coordinate for functions using network analysis and pattern recognition approaches. Through manifold learning and quantitative contrasts, CellChat classifies signaling pathways and delineates conserved and context-specific pathways across different datasets. Applying CellChat to mouse and human skin datasets shows its ability to extract complex signaling patterns. Our versatile and easy-to-use toolkit CellChat and a web-based Explorer ( http://www.cellchat.org/ ) will help discover novel intercellular communications and build cell-cell communication atlases in diverse tissues.
Collapse
Affiliation(s)
- Suoqin Jin
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Christian F Guerrero-Juarez
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Lihua Zhang
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Ivan Chang
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA, USA
- Research Cyberinfrastructure Center, University of California, Irvine, Irvine, CA, USA
| | - Raul Ramos
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
| | - Chen-Hsiang Kuan
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
- Division of Plastic Surgery, Department of Surgery, National Taiwan University, Taipei, Taiwan
| | - Peggy Myung
- Department of Dermatology, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Maksim V Plikus
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA, USA.
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
35
|
Hsueh YC, Hodgkinson CP, Gomez JA. The role of Sfrp and DKK proteins in cardiomyocyte development. Physiol Rep 2021; 9:e14678. [PMID: 33587322 PMCID: PMC7883806 DOI: 10.14814/phy2.14678] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/11/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
In this review, we summarize the role of Wnt proteins in cardiomyogenesis. More specifically, we focus on how the development of cardiomyocytes from precursor cells involves a complex interplay between Wnt canonical β-catenin signaling pathways and Wnt noncanonical signaling pathways involving PCP and JNK. We also describe recent literature which suggests that endogenous Wnt inhibitors such as the Sfrp and DKK proteins play important roles in regulating the cardiomyocyte differentiation.
Collapse
Affiliation(s)
- Ying-Chang Hsueh
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC, USA
| | - Conrad P Hodgkinson
- Mandel Center for Heart and Vascular Research, and the Duke Cardiovascular Research Center, Duke University Medical Center, Durham, NC, USA
| | - Jose A Gomez
- Department of Medicine, Clinical Pharmacology Division, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
36
|
Wang Z, Cui M, Shah AM, Tan W, Liu N, Bassel-Duby R, Olson EN. Cell-Type-Specific Gene Regulatory Networks Underlying Murine Neonatal Heart Regeneration at Single-Cell Resolution. Cell Rep 2020; 33:108472. [PMID: 33296652 PMCID: PMC7774872 DOI: 10.1016/j.celrep.2020.108472] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 10/09/2020] [Accepted: 11/11/2020] [Indexed: 12/22/2022] Open
Abstract
The adult mammalian heart has limited capacity for regeneration following injury, whereas the neonatal heart can readily regenerate within a short period after birth. Neonatal heart regeneration is orchestrated by multiple cell types intrinsic to the heart, as well as immune cells that infiltrate the heart after injury. To elucidate the transcriptional responses of the different cellular components of the mouse heart following injury, we perform single-cell RNA sequencing on neonatal hearts at various time points following myocardial infarction and couple the results with bulk tissue RNA-sequencing data collected at the same time points. Concomitant single-cell ATAC sequencing exposes underlying dynamics of open chromatin landscapes and regenerative gene regulatory networks of diverse cardiac cell types and reveals extracellular mediators of cardiomyocyte proliferation, angiogenesis, and fibroblast activation. Together, our data provide a transcriptional basis for neonatal heart regeneration at single-cell resolution and suggest strategies for enhancing cardiac function after injury.
Collapse
Affiliation(s)
- Zhaoning Wang
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Miao Cui
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Akansha M Shah
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Wei Tan
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Ning Liu
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, The Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
37
|
Chen Q, Jiao J, Wang Y, Mai Z, Ren J, He S, Li X, Chen Z. Egr-1 mediates low-dose arecoline induced human oral mucosa fibroblast proliferation via transactivation of Wnt5a expression. BMC Mol Cell Biol 2020; 21:80. [PMID: 33167868 PMCID: PMC7653895 DOI: 10.1186/s12860-020-00325-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/27/2020] [Indexed: 01/08/2023] Open
Abstract
Background Arecoline is an alkaloid natural product found in the areca nut that can induce oral submucous fibrosis and subsequent development of cancer. However, numerous studies have shown that arecoline may inhibit fibroblast proliferation and prevent collagen synthesis. Results High doses of arecoline (> 32 μg/ml) could inhibit human oral fibroblast proliferation, while low doses of arecoline (< 16 μg/ml) could promote the proliferation of human oral fibroblasts. Wnt5a was found to be both sufficient and necessary for the promotion of fibroblast proliferation. Egr-1 could mediate the expression of Wnt5a in fibroblasts, while NF-κB, FOXO1, Smad2, and Smad3 did not. Treatment with siRNAs specific to Egr-1, Egr inhibitors, or Wnt5a antibody treatment could all inhibit arecoline-induced Wnt5a upregulation and fibroblast proliferation. Conclusions Egr-1 mediates the effect of low dose arecoline treatment on human oral mucosa fibroblast proliferation by transactivating the expression of Wnt5a. Therefore, Egr inhibitors and Wnt5a antibodies are potential therapies for treatment of oral submucosal fibrosis and oral cancer.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, No.600 Tianhe road, Guangzhou, 510630, China
| | - Jiuyang Jiao
- Department of Oral & Maxillofacial Surgery & Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Youyuan Wang
- Department of Oral & Maxillofacial Surgery & Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhihui Mai
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, No.600 Tianhe road, Guangzhou, 510630, China
| | - Jing Ren
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, No.600 Tianhe road, Guangzhou, 510630, China
| | - Sijie He
- The fourth people's hospital of Nanhai district of Foshan city, Foshan, China.
| | - Xiaolan Li
- Guanghua School of stomatology & hospital of stomatology, Guangdong province key laboratory of stomatology, Sun Yat-sen University, Guangzhou, China.
| | - Zheng Chen
- Department of Stomatology, the Third Affiliated Hospital of Sun Yat-sen University, No.600 Tianhe road, Guangzhou, 510630, China.
| |
Collapse
|
38
|
Abstract
Myocardial fibrosis, the expansion of the cardiac interstitium through deposition of extracellular matrix proteins, is a common pathophysiologic companion of many different myocardial conditions. Fibrosis may reflect activation of reparative or maladaptive processes. Activated fibroblasts and myofibroblasts are the central cellular effectors in cardiac fibrosis, serving as the main source of matrix proteins. Immune cells, vascular cells and cardiomyocytes may also acquire a fibrogenic phenotype under conditions of stress, activating fibroblast populations. Fibrogenic growth factors (such as transforming growth factor-β and platelet-derived growth factors), cytokines [including tumour necrosis factor-α, interleukin (IL)-1, IL-6, IL-10, and IL-4], and neurohumoral pathways trigger fibrogenic signalling cascades through binding to surface receptors, and activation of downstream signalling cascades. In addition, matricellular macromolecules are deposited in the remodelling myocardium and regulate matrix assembly, while modulating signal transduction cascades and protease or growth factor activity. Cardiac fibroblasts can also sense mechanical stress through mechanosensitive receptors, ion channels and integrins, activating intracellular fibrogenic cascades that contribute to fibrosis in response to pressure overload. Although subpopulations of fibroblast-like cells may exert important protective actions in both reparative and interstitial/perivascular fibrosis, ultimately fibrotic changes perturb systolic and diastolic function, and may play an important role in the pathogenesis of arrhythmias. This review article discusses the molecular mechanisms involved in the pathogenesis of cardiac fibrosis in various myocardial diseases, including myocardial infarction, heart failure with reduced or preserved ejection fraction, genetic cardiomyopathies, and diabetic heart disease. Development of fibrosis-targeting therapies for patients with myocardial diseases will require not only understanding of the functional pluralism of cardiac fibroblasts and dissection of the molecular basis for fibrotic remodelling, but also appreciation of the pathophysiologic heterogeneity of fibrosis-associated myocardial disease.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- Department of Medicine (Cardiology), The Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, 1300 Morris Park Avenue Forchheimer G46B, Bronx, NY 10461, USA
| |
Collapse
|
39
|
Shin J, Yoon Y, Oh DJ. Evaluation of the Wnt signaling pathway as a prognostic marker in patients with urosepsis. Mol Cell Biochem 2020; 473:15-23. [PMID: 32588279 DOI: 10.1007/s11010-020-03804-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 06/18/2020] [Indexed: 12/30/2022]
Abstract
The Wnt signaling pathway has critical roles in dysregulated inflammation during sepsis; however, its impacts on clinical outcomes remain uncertain. This prospective observational study investigated the association between the Wnt pathway and clinical outcomes in patients with urosepsis. The study included 38 patients with urosepsis and 20 healthy individuals. Wnt3a and Wnt5a levels were measured at admission. The primary outcome was the occurrence of major adverse kidney events (MAKE), defined as new renal replacement therapy, stage 3 acute kidney injury, or death. Both Wnt3a and Wnt5a levels were higher in the patient group than in the control (P = 0.001 and P < 0.001, respectively). The primary outcome occurred in 13 (34.2%) subjects. The levels of Wnt5a were higher in subjects with MAKE than in those without MAKE (P = 0.015); however, Wnt3a levels showed no significant difference. Moreover, Wnt5a levels could be a marker to predict the possibility of MAKE (area under the curve 0.74 [0.57-0.92]; P = 0.016). Serum creatinine levels on day 0, day 5, and on discharge day were evaluated. The levels of creatinine on discharge day were higher in patients with high Wnt5a levels, compared to those with low Wnt5a levels (P = 0.030); however, no difference in Wnt5a levels was observed on day 0 and 5. Wnt3a and Wnt5a levels increased in patients with urosepsis. Moreover, evaluation of Wnt5a levels might help to predict the occurrence of MAKE and renal recovery in these patients.
Collapse
Affiliation(s)
- Jungho Shin
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Yoosik Yoon
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Dong-Jin Oh
- Department of Internal Medicine, Myongji Hospital, Hanyang University College of Medicine, 5 Hwasu-ro 14, Deogyang-gu, Goyang, 10475, Korea.
| |
Collapse
|
40
|
Abstract
Cardiovascular disease (CVD) is still a factor of mortality in the whole world. Through canonical and noncanonical pathways and with different receptors, the Wnt/β-catenin signaling pathway plays an essential role in response to heart injuries. Wnt regulates the mobilization and proliferation of cells in endothelium and epicardium in an infarcted heart. Therefore, with its profibrotic effects as well as its antagonism with other proteins, Wnt/β-catenin signaling pathway leads to beneficial effects on fibrosis and cardiac remodeling in myocardium. In addition, Wnt increases the proliferation and differentiation of cardiac progenitors in an ischemic heart. Complex interactions and dual activity of Wnt, the changes in its expression, and mutations that can change its activity during heart development have an adverse effect on cardiac myocardium after injury. However, targeting the Wnt in myocardium with cellular and molecular pathways can be suggested to improve and repair ischemic heart. Given these challenges, in this review article, we deal with the role of Wnt/β-catenin signaling pathway as well as its interactions with other cells and molecules in an ischemic myocardium.
Collapse
|
41
|
Xu W, Geng H, Liu X, Wang X, Li R, Lv Q, Liu Y, Wang J, Yang M, Jones PM, Liang J. Wingless-type MMTV integration site family member 5a: a novel biomarker regulated in type 2 diabetes mellitus and diabetic kidney disease. J Diabetes Metab Disord 2019; 18:525-532. [PMID: 31890678 PMCID: PMC6915239 DOI: 10.1007/s40200-019-00461-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 10/29/2019] [Indexed: 12/14/2022]
Abstract
Objectives Type 2 diabetes mellitus (T2DM) is sustained by insulin resistance (IR) and reduced β-cell mass, which is largely due to insulin secretory dysfunction. Wnt5a protein is essential to islet formation and β-cell migration in the development of pancreas in vertebrates. Levels of the Wnt5a protein antagonist plasma secreted frizzled-related protein 5 (Sfrp5) were elevated in patients with T2DM. However, the association between Wnt5a, T2DM patients and diabetic kidney disease (DKD) is unknown. We aim to investigate the circulating Wnt5a levels in in different clinical stages of T2DM and evaluate its correlation of duration of diabetes mellitus chronic complication. Methods A total of 329 participants (187 males, 142 females; age range 40 to 80 years) were enrolled in this study. Serum Wnt5a levels were measured by an enzyme-linked immunosorbent assay (ELISA). The demographic and clinical parameters evaluated in subjects with new onset T2DM, onset T2DM after treatment and DKD at different clinical phases. Results Wnt5a was significantly down-regulated in newly diagnosed T2DM patients and gradually increased after 3 months of treatment. Interesting, serum wnt5a was gradually increased in patients with long-term diabetes and kidney disease compared to patients with T2DM and onset DKD. Conclusions We speculated that the Wnt5a protein might regulate islet function and be involved in the onset of diabetes as a protective factor. It may be one of the inflammatory factors adversely involved in the progression of diabetic nephropathy.
Collapse
Affiliation(s)
- Wei Xu
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, SE1 1UL UK
| | - Houfa Geng
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| | - Xuekui Liu
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| | - Xiuli Wang
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| | - Rui Li
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| | - Qian Lv
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| | - Yin Liu
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| | - Jie Wang
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| | - Manqing Yang
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| | - Peter M. Jones
- Department of Diabetes, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, SE1 1UL UK
| | - Jun Liang
- Department of Endocrinology, Xuzhou Central Hospital, Xuzhou Institute of Medical Sciences, Xuzhou Clinical School of Nanjing Medical University, Affiliated Hospital of Medical School of Southeast University, Xuzhou, 221009 Jiangsu China
| |
Collapse
|
42
|
Du Y, Zhao Y, Zhu Y, Hu C, Zhang J, Ji Q, Liu W, Han H, Yang L, Zhang D, Tong S, Wang Z, Guo Y, Liu X, Zhou Y. High Serum Secreted Frizzled-Related Protein 5 Levels Associates with Early Improvement of Cardiac Function Following ST-Segment Elevation Myocardial Infarction Treated by Primary Percutaneous Coronary Intervention. J Atheroscler Thromb 2019; 26:868-878. [PMID: 30773518 PMCID: PMC6800391 DOI: 10.5551/jat.47019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
AIM Several members of secreted frizzled-related protein (SFRP) are involved in the process of myocardial ischemia-reperfusion injury. However, little is known about the role of SFRP5 in patients with acute ST-segment elevation myocardial infarction (STEMI). METHODS In this cross-sectional study, 85 patients with first-time anterior STEMI who underwent timely primary percutaneous coronary intervention (PCI) and 35 patients without coronary artery disease (CAD) were enrolled. Serum SFRP5 levels were measured using an enzyme-linked immunosorbent assay kit. Patients with STEMI were divided into low-SFRP5 and high-SFRP5 groups according to their median baseline serum SFRP5 levels. To evaluate cardiac function and structure after infarction, the left ventricular ejection fraction (LVEF) and left ventricular end-diastolic volume (LVEDV) were measured using echocardiography. The associations between changes in LVEF and reduced LVEF (≤ 50%) and clinical variables were determined by univariate and multivariate analyses. RESULTS Baseline serum SFRP5 levels were significantly higher in patients with STEMI than in those without CAD (23.3 ng/mL vs 19.8 ng/mL, P=0.008), although they decreased over time. Also, baseline serum SFRP5 levels were inversely correlated with peak hypersensitive cardiac troponin I (hs-cTnI) levels (r=-0.234, P=0.025) and peak hypersensitive C-reactive protein (hs-CRP) levels (r=-0.262, P=0.015). A multivariate linear regression model showed that changes in LVEF were positively correlated with serum SFRP5 levels at baseline (β= 0.249, 95% confidence interval (CI) 0.018-0.245, P=0.024) and 24 h after admission (β=0.220, 95% CI 0.003-0.264, P=0.045). At 3 months, LVEF in patients with high SFRP5 levels was significantly improved over baseline [(60.8±7.1) % vs (56.1±7.5) %, P=0.001]. LVEF was also significantly higher in patients with high SFRP5 levels than in those with low at the 3-month follow-up [(60.8±7.1) % vs (56.8±8.9) %, P=0.028]. Consequently, high serum SFRP5 levels at baseline were associated with a decreased risk of reduced LVEF at 3 months, independent of peak hs-cTnI and baseline cardiac function (hazard ratio 0.190, 95% CI 0.036-0.996; P=0.049). CONCLUSIONS High serum SFRP5 levels measured during the acute phase of STEMI were significantly associated with promoting myocardial recovery at an early phase following primary PCI, suggesting that SFRP5 is a potential therapeutic target in acute STEMI.
Collapse
Affiliation(s)
- Yu Du
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yingxin Zhao
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yong Zhu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Chenping Hu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Jianwei Zhang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Qingwei Ji
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Wei Liu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Hongya Han
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Lixia Yang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Dai Zhang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Shan Tong
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Zhijian Wang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yonghe Guo
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Xiaoli Liu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yujie Zhou
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| |
Collapse
|
43
|
Wang Y, Sano S, Oshima K, Sano M, Watanabe Y, Katanasaka Y, Yura Y, Jung C, Anzai A, Swirski FK, Gokce N, Walsh K. Wnt5a-Mediated Neutrophil Recruitment Has an Obligatory Role in Pressure Overload-Induced Cardiac Dysfunction. Circulation 2019; 140:487-499. [PMID: 31170826 DOI: 10.1161/circulationaha.118.038820] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Although the complex roles of macrophages in myocardial injury are widely appreciated, the function of neutrophils in nonischemic cardiac pathology has received relatively little attention. METHODS To examine the regulation and function of neutrophils in pressure overload-induced cardiac hypertrophy, mice underwent treatment with Ly6G antibody to deplete neutrophils and then were subjected to transverse aortic constriction. RESULTS Neutrophil depletion diminished transverse aortic constriction-induced hypertrophy and inflammation and preserved cardiac function. Myeloid deficiency of Wnt5a, a noncanonical Wnt, suppressed neutrophil infiltration to the hearts of transverse aortic constriction-treated mice and produced a phenotype that was similar to the neutropenic conditions. Conversely, mice overexpressing Wnt5a in myeloid cells displayed greater hypertrophic growth, inflammation, and cardiac dysfunction. Neutrophil depletion reversed the Wnt5a overexpression-induced cardiac pathology and eliminated differences in cardiac parameters between wild-type and myeloid-specific Wnt5a transgenic mice. CONCLUSIONS These findings reveal that Wnt5a-regulated neutrophil infiltration has a critical role in pressure overload-induced heart failure.
Collapse
Affiliation(s)
- Ying Wang
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.).,The First Affiliated Hospital of Chongqing Medical University, People's Republic of China (Y. Wang)
| | - Soichi Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.)
| | - Kosei Oshima
- Molecular Cardiology (K.O., Y.K.), Boston University School of Medicine, MA
| | - Miho Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.)
| | - Yosuke Watanabe
- Whitaker Cardiovascular Institute, and Vascular Biology Section (Y. Watanabe), Boston University School of Medicine, MA
| | | | - Yoshimitsu Yura
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.)
| | - Changhee Jung
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.).,Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea (C.J.)
| | - Atsushi Anzai
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston (A.A., F.K.S.)
| | - Filip K Swirski
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston (A.A., F.K.S.)
| | - Noyan Gokce
- Cardiovascular Medicine (N.G.), Boston University School of Medicine, MA.,Cardiology, Boston Medical Center, MA (N.G.). Dr Watanabe is currently at the Department of Internal Medicine II, University of Yamanashi, Faculty of Medicine, Chuo, Yamanashi, Japan. Dr Katanasaka is currently at the Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Japan. Dr Anzai is currently at the Department of Cardiology, Keio University, School of Medicine, Tokyo, Japan
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (Y. Wang, S.S., M.S., Y.Y., C.J., K.W.)
| |
Collapse
|
44
|
Chen D, Yu F, Wu F, Bai M, Lou F, Liao X, Wang C, Ye L. The role of Wnt7B in the mediation of dentinogenesis via the ERK1/2 pathway. Arch Oral Biol 2019; 104:123-132. [PMID: 31181411 DOI: 10.1016/j.archoralbio.2019.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/24/2019] [Accepted: 05/09/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVES This study investigates the role of Wnt7b in mouse dentin formation. DESIGN C57BL/6 mouse tooth germs at different developmental stages were collected to measure the expression of Wnt7b by immunohistochemical staining. The morphology of mandibles of Dmp1-cre;ROSA26-Wnt7b transgenic mice and ROSA26-Wnt7b littermates was analyzed by Micro-CT and HE staining. The ultramicrostructure of dentin was scanned with an electron microscope. Primary mouse dental papillae cells (MDPCs) and odontoblastic cell line (A11) were cultured and infected with adenovirus to overexpress Wnt7b. Cell proliferation and cell apoptosis were evaluated using CCK-8 and flow cytometry. Osteogenic differentiation of MDPCs and A11 was assessed by Alizarin red staining, and qPCR detection of osteogenic gene expression. The activation of signaling pathways was measured by the use of western blot analysis. The ERK1/2 inhibitor was used to test the effect of Wnt7b regulated cell differentiation. RESULTS Wnt7b was expressed principally in the mouse odontoblast layer after the early bell stage. In transgenic mice, Wnt7b was over-expressed in tooth mesenchyme, with a thinner predentin layer and thicker intertubular dentin. Both the micro-hardness value and the Ca/Pi ratio of dentin of transgenic mice were higher. Wnt7b promoted proliferation and mineralization of MDPCs and A11. The protein level of p-ERK1/2 was found to be higher in A11 infected with Ad-Wnt7b. The ERK signaling pathway inhibitor partly rescued the Wnt7b-induced differentiation of A11. CONCLUSIONS Wnt7b enhances dentinogenesis by increasing the proliferation and differentiation of dental mesenchymal cells partly through ERK1/2 pathway.
Collapse
Affiliation(s)
- Dian Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu 610041, PR China
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu 610041, PR China
| | - Fanzi Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu 610041, PR China
| | - Mingru Bai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu 610041, PR China
| | - Feng Lou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu 610041, PR China
| | - Xueyang Liao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu 610041, PR China
| | - Chenglin Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu 610041, PR China
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Chengdu 610041, PR China.
| |
Collapse
|
45
|
Wang Z, Zhao J, Wang T, Du X, Xie J. Fine-particulate matter aggravates cigarette smoke extract-induced airway inflammation via Wnt5a-ERK pathway in COPD. Int J Chron Obstruct Pulmon Dis 2019; 14:979-994. [PMID: 31190784 PMCID: PMC6512785 DOI: 10.2147/copd.s195794] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Exposure to environmental particulate matter (PM) ≤2.5 μm in diameter (PM2.5) and smoking are common contributors to COPD, and pertinent research implicates both factors in pulmonary inflammation. Using in vivo mouse and in vitro human cellular models, we investigated the joint impact of PM2.5 pollution, and cigarette smoke (CS) in mice or cigarette-smoke extract (CSE) in cells on COPD inflammation, and explored potential mechanisms. Methods Tissue changes in lungs of C57BL/6 mice exposed to PM2.5 and CS were studied by light microscopy, H&E, immunochemistry, and immunofluorescence-stained sections. Levels of inflammatory factors induced by PM2.5/CS in mice and PM2.5/CSE in 16HBE cells were also monitored by quantitative reverse-transcription (qRT)-PCR and ELISA. Expression of genes related to the Wnt5a-signaling pathway was assessed at transcriptional and protein levels using immunofluorescence, qRT-PCR, and Western blotting. Results Inflammatory response to combined exposure of PM2.5 and CS or CSE in mouse and 16HBE cells surpassed responses incited separately. Although separate PM2.5 and CS/CSE exposure upregulated the expression of Wnt5a (a member of the Wnt-secreted glycoprotein family), combined PM2.5 and CS/CSE exposure produced a steeper rise in Wnt5a levels. Use of a Wnt5a antagonist (BOX5) successfully blocked related inflammatory effects. ERK phosphorylation appeared to mediate the effects of Wnt5a in the COPD model, promoting PM2.5 aggravation of CS/CSE-induced airway inflammation. Conclusion Our findings suggest that combined PM2.5 and CS/CSE exposure induce airway inflammation and Wnt5a expression in vivo in mice and in vitro in 16HBE cells. Furthermore, PM2.5 seems to aggravate CS/CSE-induced inflammation via the Wnt5a–ERK pathway in the context of COPD.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China,
| | - Junling Zhao
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China,
| | - Ting Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China,
| | - Xiaohui Du
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China,
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China,
| |
Collapse
|
46
|
WNT3a and WNT5a Transported by Exosomes Activate WNT Signaling Pathways in Human Cardiac Fibroblasts. Int J Mol Sci 2019; 20:ijms20061436. [PMID: 30901906 PMCID: PMC6472055 DOI: 10.3390/ijms20061436] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/28/2022] Open
Abstract
WNT signaling plays an important role in fibrotic processes in the heart. Recently, exosomes have been proposed as novel extracellular transporters for WNT proteins. In this study, we analyzed whether WNT3a and WNT5a carried by exosomes could activate downstream molecular pathways in human cardiac fibroblasts. Exosomes were isolated from conditioned medium of control, WNT3a- and WNT5a-producing L cells by differential ultracentrifugations. Obtained exosomes showed size ranging between 20–150 nm and expressed exosomal markers ALG-2-interacting protein X (ALIX) and CD63. Treatment with WNT3a-rich exosomes inhibited activity of glycogen synthase kinase 3β (GSK3β), induced nuclear translocation of β-catenin, and activated T-cell factor (TCF)/lymphoid enhancer factor (LEF) transcription factors as well as expression of WNT/β-catenin responsive genes in cardiac fibroblasts, but did not coactivate extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and activator protein 1 (AP-1) signaling pathways. In contrast, exosomes produced by WNT5a-producing L cells failed to activate β-catenin-dependent response, but successfully triggered phosphorylation of ERK1/2 and JNK and stimulated IL-6 production. In conclusion, exosomes containing WNT proteins can functionally contribute to cardiac fibrosis by activating profibrotic WNT pathways on cardiac fibroblasts and may represent a novel mechanism of spreading profibrotic signals in the heart.
Collapse
|
47
|
Feng Y, Liang Y, Zhu X, Wang M, Gui Y, Lu Q, Gu M, Xue X, Sun X, He W, Yang J, Johnson RL, Dai C. The signaling protein Wnt5a promotes TGFβ1-mediated macrophage polarization and kidney fibrosis by inducing the transcriptional regulators Yap/Taz. J Biol Chem 2018; 293:19290-19302. [PMID: 30333225 DOI: 10.1074/jbc.ra118.005457] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 10/11/2018] [Indexed: 12/24/2022] Open
Abstract
M2 macrophage polarization is known to underlie kidney fibrosis. We previously reported that most of the members of the Wnt family of signaling proteins are induced in fibrotic kidneys. Dysregulation of the signaling protein Wnt5a is associated with fibrosis, but little is known about the role of Wnt5a in regulating M2 macrophage activation that results in kidney fibrosis. Here, using murine Raw 264.7 cells and bone marrow-derived macrophages, we found that Wnt5a enhanced transforming growth factor β1 (TGFβ1)-induced macrophage M2 polarization as well as expression of the transcriptional regulators Yes-associated protein (Yap)/transcriptional coactivator with PDZ-binding motif (Taz). Verteporfin blockade of Yap/Taz inhibited both Wnt5a- and TGFβ1-induced macrophage M2 polarization. In mouse models of kidney fibrosis, shRNA-mediated knockdown of Wnt5a expression diminished kidney fibrosis, macrophage Yap/Taz expression, and M2 polarization. Moreover, genetic ablation of Taz in macrophages attenuated kidney fibrosis and macrophage M2 polarization in mice. Collectively, these results indicate that Wnt5a promotes kidney fibrosis by stimulating Yap/Taz-mediated macrophage M2 polarization.
Collapse
Affiliation(s)
- Ye Feng
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Yan Liang
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Xingwen Zhu
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Mingjie Wang
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Yuan Gui
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Qingmiao Lu
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Mengru Gu
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Xian Xue
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Xiaoli Sun
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Weichun He
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Junwei Yang
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| | - Randy L Johnson
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| | - Chunsun Dai
- From the Center for Kidney Disease, 2nd Affiliated Hospital, Nanjing Medical University, 262 North Zhongshan Road, Nanjing, 210003 Jiangsu, China and
| |
Collapse
|
48
|
Meyer IS, Leuschner F. The role of Wnt signaling in the healing myocardium: a focus on cell specificity. Basic Res Cardiol 2018; 113:44. [PMID: 30327885 DOI: 10.1007/s00395-018-0705-y] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/01/2018] [Accepted: 10/09/2018] [Indexed: 12/18/2022]
Abstract
Various cell types are involved in the healing process after myocardial infarction (MI). Besides cardiac resident cells (such as cardiomyocytes, fibroblasts and endothelial cells) already present at the lesion site, a massive influx of leukocytes (mainly monocytes and neutrophils) is observed within hours after the ischemic event. So far, little is known about modes of interaction of these cells. Wnt signaling is an evolutionary conserved signaling cassette known to play an important role in cell-cell communication. While the overall reactivation of Wnt signaling upon ischemic injury is well described, the precise expression pattern of Wnt proteins, however, is far from understood. We here describe known Wnt components that partake in MI healing and differentiate cell-specific aspects. The secretion of Wnt proteins and their antagonists in the context of cardiac inflammation after MI appear to be tightly regulated in a spatial-temporal manner. Overall, we aim to stress the importance of elucidating not only Wnt component-specific aspects, but also their sometimes contradicting effects in different target cells. A better understanding of Wnt signaling in MI healing may eventually lead to the development of successful therapeutic approaches in an often considered "un-druggable" pathway.
Collapse
Affiliation(s)
- Ingmar Sören Meyer
- Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Florian Leuschner
- Department of Internal Medicine III, University Hospital Heidelberg, University of Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
49
|
Frangogiannis NG. Cardiac fibrosis: Cell biological mechanisms, molecular pathways and therapeutic opportunities. Mol Aspects Med 2018; 65:70-99. [PMID: 30056242 DOI: 10.1016/j.mam.2018.07.001] [Citation(s) in RCA: 582] [Impact Index Per Article: 83.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/23/2018] [Indexed: 12/13/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic companion of most myocardial diseases, and is associated with systolic and diastolic dysfunction, arrhythmogenesis, and adverse outcome. Because the adult mammalian heart has negligible regenerative capacity, death of a large number of cardiomyocytes results in reparative fibrosis, a process that is critical for preservation of the structural integrity of the infarcted ventricle. On the other hand, pathophysiologic stimuli, such as pressure overload, volume overload, metabolic dysfunction, and aging may cause interstitial and perivascular fibrosis in the absence of infarction. Activated myofibroblasts are the main effector cells in cardiac fibrosis; their expansion following myocardial injury is primarily driven through activation of resident interstitial cell populations. Several other cell types, including cardiomyocytes, endothelial cells, pericytes, macrophages, lymphocytes and mast cells may contribute to the fibrotic process, by producing proteases that participate in matrix metabolism, by secreting fibrogenic mediators and matricellular proteins, or by exerting contact-dependent actions on fibroblast phenotype. The mechanisms of induction of fibrogenic signals are dependent on the type of primary myocardial injury. Activation of neurohumoral pathways stimulates fibroblasts both directly, and through effects on immune cell populations. Cytokines and growth factors, such as Tumor Necrosis Factor-α, Interleukin (IL)-1, IL-10, chemokines, members of the Transforming Growth Factor-β family, IL-11, and Platelet-Derived Growth Factors are secreted in the cardiac interstitium and play distinct roles in activating specific aspects of the fibrotic response. Secreted fibrogenic mediators and matricellular proteins bind to cell surface receptors in fibroblasts, such as cytokine receptors, integrins, syndecans and CD44, and transduce intracellular signaling cascades that regulate genes involved in synthesis, processing and metabolism of the extracellular matrix. Endogenous pathways involved in negative regulation of fibrosis are critical for cardiac repair and may protect the myocardium from excessive fibrogenic responses. Due to the reparative nature of many forms of cardiac fibrosis, targeting fibrotic remodeling following myocardial injury poses major challenges. Development of effective therapies will require careful dissection of the cell biological mechanisms, study of the functional consequences of fibrotic changes on the myocardium, and identification of heart failure patient subsets with overactive fibrotic responses.
Collapse
Affiliation(s)
- Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer G46B, Bronx, NY, 10461, USA.
| |
Collapse
|
50
|
Hoseth EZ, Krull F, Dieset I, Mørch RH, Hope S, Gardsjord ES, Steen NE, Melle I, Brattbakk HR, Steen VM, Aukrust P, Djurovic S, Andreassen OA, Ueland T. Exploring the Wnt signaling pathway in schizophrenia and bipolar disorder. Transl Psychiatry 2018; 8:55. [PMID: 29507296 PMCID: PMC5838215 DOI: 10.1038/s41398-018-0102-1] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 01/05/2023] Open
Abstract
The Wnt signaling pathway plays a crucial role in neurodevelopment and in regulating the function and structure of the adult nervous system. Schizophrenia (SCZ) and bipolar disorder (BD) are severe mental disorders with evidence of subtle neurodevelopmental, structural and functional neuronal abnormalities. We aimed to elucidate the role of aberrant regulation of the Wnt system in these disorders by evaluating plasma levels of secreted Wnt modulators in patients (SCZ = 551 and BD = 246) and healthy controls (HCs = 639) using enzyme immune-assay. We also investigated the expression of 141 Wnt-related genes in whole blood in a subsample (SCZ = 338, BD = 241, and HCs = 263) using microarray analysis. Both SCZ and BD had dysregulated mRNA expression of Wnt-related genes favoring attenuated canonical (beta-catenin-dependent) signaling, and there were also indices of enhanced non-canonical Wnt signaling. In particular, FZD7, which may activate all Wnt pathways, but favors non-canonical signaling, and NFATc3, a downstream transcription factor and readout of the non-canonical Wnt/Ca2+ pathway, were significantly increased in SCZ and BD (p < 3 × 10-4). Furthermore, patients had lower plasma levels of soluble dickkopf 1 and sclerostin (p < 0.01) compared with HC. Our findings suggest that SCZ and BD are characterized by abnormal Wnt gene expression and plasma protein levels, and we propose that drugs targeting the Wnt pathway may have a role in the treatment of severe mental disorders.
Collapse
Affiliation(s)
- Eva Z. Hoseth
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway ,Division of Mental Health and Addiction, Møre and Romsdal Hospital Trust, Kristiansund, Norway
| | - Florian Krull
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ingrid Dieset
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ragni H. Mørch
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Sigrun Hope
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Departent of Neurohabilitation, Division of Neurology, Oslo University Hospital, Oslo, Norway
| | - Erlend S. Gardsjord
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Nils Eiel Steen
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Ingrid Melle
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Hans-Richard Brattbakk
- 0000 0004 1936 7443grid.7914.bNORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Oslo, Norway ,0000 0000 9753 1393grid.412008.fDr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Oslo, Norway
| | - Vidar M. Steen
- 0000 0004 1936 7443grid.7914.bNORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Oslo, Norway ,0000 0000 9753 1393grid.412008.fDr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Oslo, Norway
| | - Pål Aukrust
- 0000 0004 0389 8485grid.55325.34Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 0389 8485grid.55325.34Instiute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway ,0000 0004 1936 8921grid.5510.1K.G. Jensen Inflammatory Research Center, University of Oslo, Oslo, Norway
| | - Srdjan Djurovic
- 0000 0004 0389 8485grid.55325.34Department of Medical Genetics, Oslo University Hospital, Oslo, Norway ,0000 0004 1936 7443grid.7914.bNORMENT, KG Jebsen Centre for Psychosis Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ole A. Andreassen
- 0000 0004 0389 8485grid.55325.34NORMENT, KG Jebsen Centre for Psychosis Research, Institute of Clinical Medicine, University of Oslo, and Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Thor Ueland
- Research Institute for Internal Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway. .,Instiute of Clinical Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway. .,K.G. Jensen Inflammatory Research Center, University of Oslo, Oslo, Norway. .,K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway.
| |
Collapse
|