1
|
Batan D, Tseropoulos G, Kirkpatrick BE, Bishop C, Bera K, Khang A, Weiser-Evans M, Anseth KS. PTEN Regulates Myofibroblast Activation in Valvular Interstitial Cells Based on Subcellular Localization. Adv Biol (Weinh) 2025:e2400540. [PMID: 40229965 DOI: 10.1002/adbi.202400540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/03/2025] [Indexed: 04/16/2025]
Abstract
Aortic valve stenosis (AVS) is characterized by altered mechanics of the valve leaflets, which disrupts blood flow through the aorta and can cause left ventricle hypotrophy. These changes in the valve tissue result in the activation of resident valvular interstitial cells (VICs) into myofibroblasts, which have increased levels of αSMA in their stress fibers. The persistence of VIC myofibroblast activation is a hallmark of AVS. In recent years, the tumor suppressor gene phosphatase and tensin homolog (PTEN) has emerged as an important player in the regulation of fibrosis in various tissues (e.g., lung, skin), which motivated to investigate PTEN as a potential protective factor against matrix-induced myofibroblast activation in VICs. In aortic valve samples from humans, high levels of PTEN are found in healthy tissue and low levels of PTEN in diseased tissue. Then, using pharmacological inducers to treat VIC cultures, it is observed that PTEN overexpression prevented stiffness-induced myofibroblast activation, whereas genetic and pharmacological inhibition of PTEN further activated myofibroblasts. The increased nuclear PTEN localization is also observed in VICs cultured on stiff matrices, and nuclear PTEN also correlated with smaller nuclei, altered expression of histones, and a quiescent fibroblast phenotype. Together, these results suggest that PTEN not only suppresses VIC activation, but functions to promote quiescence, and can serve as a potential pharmacological target for the treatment of AVS.
Collapse
Affiliation(s)
- Dilara Batan
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, 80303, USA
- The BioFrontiers Institute, University of Colorado, Boulder, Colorado, 80303, USA
| | - Georgios Tseropoulos
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, 80303, USA
- The BioFrontiers Institute, University of Colorado, Boulder, Colorado, 80303, USA
| | - Bruce E Kirkpatrick
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, 80303, USA
- The BioFrontiers Institute, University of Colorado, Boulder, Colorado, 80303, USA
- Medical Scientist Training Program, School of Medicine, University of Colorado, Aurora, Colorado, 80045, USA
| | - Carrie Bishop
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, 80303, USA
| | - Kaustav Bera
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, 80303, USA
- The BioFrontiers Institute, University of Colorado, Boulder, Colorado, 80303, USA
| | - Alex Khang
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, 80303, USA
- The BioFrontiers Institute, University of Colorado, Boulder, Colorado, 80303, USA
| | - Mary Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado, Anschutz Medical Campus, 12700 East 19th Avenue, C281, Research Complex 2, Room 7101, Aurora, Colorado, 80045, USA
- Center for Fibrosis Research and Translation, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, 80045, USA
- Department of Medicine, Cardiovascular Pulmonary Research Program, University of Colorado, Anschutz Medical Campus, Aurora, Colorado, 80045, USA
| | - Kristi S Anseth
- Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, 80303, USA
- The BioFrontiers Institute, University of Colorado, Boulder, Colorado, 80303, USA
| |
Collapse
|
2
|
Liu W, Cai X, Duan S, Shen J, Wu J, Zhou Z, Yu K, He C, Wang Y. E3 ubiquitin ligase Smurf1 promotes cardiomyocyte pyroptosis by mediating ubiquitin-dependent degradation of TRIB2 in a rat model of heart failure. Int Rev Immunol 2025:1-15. [PMID: 39749701 DOI: 10.1080/08830185.2024.2434058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/27/2024] [Accepted: 11/17/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE Heart failure (HF) causes structural and functional changes in the heart, with the pyroptosis-mediated inflammatory response as the core link in HF pathogenesis. E3 ubiquitin ligases participate in cardiovascular disease progression. Here, we explored the underlying molecular mechanisms of E3 ubiquitin ligase Smurf1 in governing HF. METHODS HF rat/H9C2 cell models were established by doxorubicin intraperitoneal injections/hypoxia-reoxygenation (H/R), and treated with Smurf1 siRNA and oe-TRIB2 lentivirus plasmids or the NF-κB pathway inhibitor PDTC/si-smurf1, si-TRIB2, protease inhibitor MG132, or lysosomal inhibitor NH4Cl. The cardiac function/cardiac tissue pathological changes/fibrosis in HF rats were evaluated by echocardiography/H&E and Masson staining. GSDMD-N expression was determined by immunohistochemistry. Cell viability/lactate dehydrogenase (LDH) activity/IL-1β and IL-18 levels were measured by CCK-8/LDH kit/ELISA. The interaction between TRIB2 and Smurf1/TRIB2 ubiquitination levels was assessed by co-immunoprecipitation assay. The expression levels of Smurf1 and TRIB2 messenger RNA (mRNA) were determined by RT-qPCR. Levels of Smurf1/TRIB2/the NF-κB pathway-related factors/pyroptosis-related factors and TRIB2 mRNA were determined by Western blot/RT-qPCR. RESULTS Smurf1 was highly expressed in H/R-induced H9C2 cells/HF rats, while its knockdown up-regulated TRIB2 and repressed the NF-κB pathway, reduced cardiomyocyte pyroptosis, and attenuated HF. Mechanistically, Smurf1 promoted TRIB2 degradation through an ubiquitin-dependent manner and activated the NF-κB pathway under H/R conditions. TRIB2 silencing annulled Smurf1 knockdown-regulated NF-κB pathway and cardiomyocyte pyroptosis. TRIB2 overexpression inactivated the NF-κB pathway and reduced cardiomyocyte pyroptosis, thus retarding HF. CONCLUSION Smurf1 was highly expressed in HF rats, which promoted TRIB2 ubiquitination degradation and activated the NF-κB pathway, thereby promoting cardiomyocyte pyroptosis in HF rats.
Collapse
Affiliation(s)
- Wei Liu
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| | - Xin Cai
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| | - Shiying Duan
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| | - Jihua Shen
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| | - Jiayuan Wu
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| | - Zhengwei Zhou
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| | - Kaili Yu
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| | - Caihong He
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| | - Yuqin Wang
- Department of Cardiology, Loudi Central Hospital, Loudi City, Hunan Province, China
| |
Collapse
|
3
|
Yan T, Song S, Sun W, Ge Y. HAPLN1 knockdown inhibits heart failure development via activating the PKA signaling pathway. BMC Cardiovasc Disord 2024; 24:197. [PMID: 38580957 PMCID: PMC10996236 DOI: 10.1186/s12872-024-03861-8] [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: 10/08/2023] [Accepted: 03/26/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Heart failure (HF) is a heterogeneous syndrome that affects millions worldwide, resulting in substantial health and economic burdens. However, the molecular mechanism of HF pathogenesis remains unclear. METHODS HF-related key genes were screened by a bioinformatics approach.The impacts of HAPLN1 knockdown on Angiotensin II (Ang II)-induced AC16 cells were assessed through a series of cell function experiments. Enzyme-linked immunosorbent assay (ELISA) was used to measure levels of oxidative stress and apoptosis-related factors. The HF rat model was induced by subcutaneous injection isoprenaline and histopathologic changes in the cardiac tissue were assessed by hematoxylin and eosin (HE) staining and echocardiographic index. Downstream pathways regulated by HAPLN1 was predicted through bioinformatics and then confirmed in vivo and in vitro by western blot. RESULTS Six hub genes were screened, of which HAPLN1, FMOD, NPPB, NPPA, and COMP were overexpressed, whereas NPPC was downregulated in HF. Further research found that silencing HAPLN1 promoted cell viability and reduced apoptosis in Ang II-induced AC16 cells. HAPLN1 knockdown promoted left ventricular ejection fraction (LVEF) and left ventricular fraction shortening (LVFS), while decreasing left ventricular end-systolic volume (LVESV) in the HF rat model. HAPLN1 knockdown promoted the levels of GSH and suppressed the levels of MDA, LDH, TNF-α, and IL-6. Mechanistically, silencing HAPLN1 activated the PKA pathway, which were confirmed both in vivo and in vitro. CONCLUSION HAPLN1 knockdown inhibited the progression of HF by activating the PKA pathway, which may provide novel perspectives on the management of HF.
Collapse
Affiliation(s)
- Tao Yan
- Department of Cardiology, Zibo Municipal Hospital, Ward 1, No. 139 Huangong Road, Linzi District, Zibo City, Shandong Province, 255400, China
| | - Shushuai Song
- Department of Cardiology, Qingdao Fuwai Cardiovascular Hospital, No. 201 Nanjing Road, Shibei District, Qingdao City, Shandong Province, 266034, China
| | - Wendong Sun
- Department of Cardiology, Zibo Municipal Hospital, No. 139 Huangong Road, Linzi District, Zibo City, Shandong Province, 255400, China
| | - Yiping Ge
- Department of Cardiology, Qingdao Fuwai Cardiovascular Hospital, No. 201 Nanjing Road, Shibei District, Qingdao City, Shandong Province, 266034, China.
| |
Collapse
|
4
|
Wei W, Li C, Zhang B, Huang D, Li Z, Gao J. Total Glucosides of Paeony Ameliorate Myocardial Injury in Chronic Heart Failure Rats by Suppressing PARP-1. J Cardiovasc Transl Res 2024; 17:388-402. [PMID: 37831380 PMCID: PMC11052853 DOI: 10.1007/s12265-023-10440-3] [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: 04/16/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023]
Abstract
Total glucosides of paeony (TGP) have a potential protective effect on chronic heart failure (CHF) rats, but the mechanism remains unclear. PARP inhibition prevents the decrease in myocardial contractility. Therefore, we aim to investigate the effects and mechanisms of TGP on CHF and the role of PARP-1 in CHF. Left anterior descending ligation rats and adriamycin-treated H9C9 cells were used as CHF models, and captopril as a positive control for in vivo experiments. We found that TGP alleviated myocardial remodeling and improved cardiac morphology and function. TGP also reduced myocardial apoptosis and autophagy, decreased inflammatory factor release, and inhibited the PARP-1 and NF-κB proteins. Through cell transfection, we found that PAPR-1 knockdown inhibited NF-κB nuclear translocation. Additionally, TGP inhibited apoptosis, autophagy, and inflammation in CHF cells, while PARP-1 overexpression partially antagonized them. In conclusion, TGP has the potential to improve CHF and PARP-1 may be a potential target.
Collapse
Affiliation(s)
- Wenjuan Wei
- Department of Cardiology, The First People's Hospital of Xiaoshan District, No. 199, Shixin Nan Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Caiyan Li
- Department of Cardiology, The First People's Hospital of Xiaoshan District, No. 199, Shixin Nan Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Baoyong Zhang
- Department of Cardiology, The First People's Hospital of Xiaoshan District, No. 199, Shixin Nan Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Deyun Huang
- Department of Cardiology, The First People's Hospital of Xiaoshan District, No. 199, Shixin Nan Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China
| | - Zheming Li
- College of Pharmacy, Hangzhou Medical College, No. 481, Binwen Road, Binjiang District, Hangzhou, 310053, Zhejiang, China.
| | - Jiaer Gao
- Department of Cardiology, The First People's Hospital of Xiaoshan District, No. 199, Shixin Nan Road, Xiaoshan District, Hangzhou, 311200, Zhejiang, China.
| |
Collapse
|
5
|
Kraus L, Beavens B. The Current Therapeutic Role of Chromatin Remodeling for the Prognosis and Treatment of Heart Failure. Biomedicines 2023; 11:biomedicines11020579. [PMID: 36831115 PMCID: PMC9953583 DOI: 10.3390/biomedicines11020579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Cardiovascular diseases are a major cause of death globally, with no cure to date. Many interventions have been studied and suggested, of which epigenetics and chromatin remodeling have been the most promising. Over the last decade, major advancements have been made in the field of chromatin remodeling, particularly for the treatment of heart failure, because of innovations in bioinformatics and gene therapy. Specifically, understanding changes to the chromatin architecture have been shown to alter cardiac disease progression via variations in genomic sequencing, targeting cardiac genes, using RNA molecules, and utilizing chromatin remodeler complexes. By understanding these chromatin remodeling mechanisms in an injured heart, treatments for heart failure have been suggested through individualized pharmaceutical interventions as well as biomarkers for major disease states. By understanding the current roles of chromatin remodeling in heart failure, a potential therapeutic approach may be discovered in the future.
Collapse
|