1
|
Imran M, Altamimi ASA, Afzal M, Babu MA, Goyal K, Ballal S, Sharma P, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H. Targeting senescence and GATA4 in age-related cardiovascular disease: a comprehensive approach. Biogerontology 2025; 26:45. [PMID: 39831933 DOI: 10.1007/s10522-025-10189-z] [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: 10/31/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
The growing prevalence of age-related cardiovascular diseases (CVDs) poses significant health challenges, necessitating the formulation of novel treatment approaches. GATA4, a vital transcription factor identified for modulating cardiovascular biology and cellular senescence, is recognized for its critical involvement in CVD pathogenesis. This review collected relevant studies from PubMed, Google Scholar, and Science Direct using search terms like 'GATA4,' 'cellular senescence,' 'coronary artery diseases,' 'hypertension,' 'heart failure,' 'arrhythmias,' 'congenital heart diseases,' 'cardiomyopathy,' and 'cardiovascular disease.' Additionally, studies investigating the molecular mechanisms underlying GATA4-mediated regulation of GATA4 and senescence in CVDs were analyzed to provide comprehensive insights into this critical aspect of potential treatment targeting. Dysregulation of GATA4 is involved in a variety of CVDs, as demonstrated by both experimental and clinical research, comprising CAD, hypertension, congenital heart diseases, cardiomyopathy, arrhythmias, and cardiac insufficiency. Furthermore, cellular senescence enhances the advancement of age-related CVDs. These observations suggested that therapies targeting GATA4, senescence pathways, or both as necessary may be an effective intervention in CVD progression and prognosis. Addressing age-related CVDs by targeting GATA4 and senescence is a broad mechanism approach. It implies further investigation of the molecular nature of these processes and elaboration of an effective therapeutic strategy. This review highlights the importance of GATA4 and senescence in CVD pathogenesis, emphasizing their potential as therapeutic targets for age-related CVDs.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha, 91911, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, UP, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Fadiyah Jadid Alanazi
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Sakaka 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| |
Collapse
|
2
|
Pathan SS, Pugazenthi A, Dixon BREA, Wensel TG, Rosengart TK, Mathison M. Activation of a GPCR, ORL1 Receptor: A Novel Therapy to Prevent Heart Failure Progression. J Cardiovasc Dev Dis 2024; 11:355. [PMID: 39590198 PMCID: PMC11595155 DOI: 10.3390/jcdd11110355] [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: 09/07/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 11/28/2024] Open
Abstract
The number of ischemic heart failure (HF) patients is growing dramatically worldwide. However, there are at present no preventive treatments for HF. Our previous study showed that Gata4 overexpression improved cardiac function after myocardial infarction in rat hearts. We also found that Gata4 overexpression significantly increased the expression of a Pnoc gene, an endogenous ligand for the cell membrane receptor ORL1. We hypothesized that the activation of the ORL1 receptor would suppress HF in a rat ischemic heart model. Adult Sprague Dawley rats (8 weeks old, six males and six females) underwent left anterior descending coronary artery ligation. Three weeks later, normal saline or MCOPPB (ORL1 activator, 2.5 mg/kg/day) intraperitoneal injection was started, and continued 5 days a week for 3 months. Echocardiography was performed six times: pre-operative, 3 days after coronary artery ligation, pre-MCOPPB or saline injection, and 1, 2, and 3 months after saline or MCOPPB injection started. Animals were euthanized after 3 months' follow-up and the hearts were harvested for histological analysis. The ORL1 activator, MCOPPB, significantly improved cardiac function after myocardial infarction in rats (ejection fraction, MCOPPB vs. saline at euthanasia, 67 ± 3% vs. 43 ± 2%, p < 0.001). MCOPPB also decreased fibrosis and induced angiogenesis. Thus, the ORL1 activator, MCOPPB, may be a novel treatment for preventing HF progression.
Collapse
Affiliation(s)
- Saliha S. Pathan
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| | - Aarthi Pugazenthi
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| | - Beverly R. E. A. Dixon
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| | - Theodore G. Wensel
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA;
| | - Todd K. Rosengart
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| | - Megumi Mathison
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA; (S.S.P.); (A.P.); (B.R.E.A.D.); (T.K.R.)
| |
Collapse
|
3
|
Pathan S, Pugazenthi A, Dixon BR, Wensel TG, Rosengart TK, Mathison M. Activation of a GPCR, ORL1 receptor: A novel therapy to prevent heart failure progression. RESEARCH SQUARE 2024:rs.3.rs-4578315. [PMID: 39070633 PMCID: PMC11275996 DOI: 10.21203/rs.3.rs-4578315/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Purpose The number of ischemic heart failure (HF) patients is growing dramatically worldwide. However, there are at present no preventive treatments for HF. Our previous study showed that Gata4 overexpression improved cardiac function after myocardial infarction in the rat heart. We also found that Gata4 overexpression significantly increased a Pnoc gene expression, an endogenous ligand for cell membrane receptor, ORL1. We hypothesized that an activation of ORL1 receptor would suppress HF in a rat ischemic heart model. Method Adult Sprague Dawley rats (8 weeks old, 6 males and 6 females) underwent left anterior descending coronary artery ligation. Three weeks later, normal saline or MCOPPB (ORL1 activator, 2.5mg/kg/day) intraperitoneal injection was started, and continued 5 days a week, for 3 months. Echocardiography was performed six times, pre-operative, 3 days after coronary artery ligation, pre-MCOPPB or saline injection, and 1, 2, and 3 months after saline or MCOPPB injection started. Animals were euthanized after 3 months follow up and the heart was harvested for histological analysis. Results ORL1 activator, MCOPPB, significantly improved cardiac function after myocardial infarction in rat (Ejection fraction, MCOPPB vs saline at euthanasia, 67 ± 3 vs 43 ± 2, p < 0.001). MCOPPB also decreased fibrosis and induced angiogenesis. Conclusion ORL1 activator, MCOPPB, may be a novel treatment for preventing HF progression.
Collapse
|
4
|
Zhao J, Hu J, Zhang R, Deng J. CEBPD REGULATES OXIDATIVE STRESS AND INFLAMMATORY RESPONSES IN HYPERTENSIVE CARDIAC REMODELING. Shock 2023; 60:713-723. [PMID: 37752084 DOI: 10.1097/shk.0000000000002228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
ABSTRACT Hypertension seems to inevitably cause cardiac remodeling, increasing the mortality of patients. This study aimed to explore the molecular mechanism of CCAAT/enhancer-binding protein delta (CEBPD)-mediated oxidative stress and inflammation in hypertensive cardiac remodeling. The hypertensive murine model was established through angiotensin-II injection, and hypertensive mice underwent overexpressed CEBPD vector injection, cardiac function evaluation, and observation of histological changes. The cell model was established by angiotensin-II treatment and transfected with overexpressed CEBPD vector. Cell viability and surface area and oxidative stress (reactive oxygen species/superoxide dismutase/lactate dehydrogenase/malondialdehyde) were assessed, and inflammatory factors (TNF-α/IL-1β/IL-6/IL-10) were determined both in vivo and in vitro . The levels of CEBPD, miR-96-5p, inositol 1,4,5-trisphosphate receptor 1 (IP3R), natriuretic peptide B, and natriuretic peptide A, collagen I, and collagen III in tissues and cells were determined. The binding relationships of CEBPD/miR-96-5p/IP3R 3' untranslated region were validated. CEBPD was reduced in cardiac tissue of hypertensive mice, and CEBPD upregulation improved cardiac function and attenuated fibrosis and hypertrophy, along with reductions of reactive oxygen species/lactate dehydrogenase/malondialdehyde/TNF-α/IL-1β/IL-6 and increases in superoxide dismutase/IL-10. CEBPD enriched on the miR-96-5p promoter to promote miR-96-5p expression, whereas CEBPD and miR-96-5p negatively regulated IP3R. miR-96-5p silencing/IP3R overexpression reversed the alleviative role of CEBPD overexpression in hypertensive mice. In summary, CEBPD promoted miR-96-5p to negatively regulate IP3R expression to inhibit oxidative stress and inflammation, thereby alleviating hypertensive cardiac remodeling.
Collapse
Affiliation(s)
- Jinghong Zhao
- Department of Cardiology, Nanchong Central Hospital, Nanchong, China
| | | | | | | |
Collapse
|
5
|
Stem cell laden nano and micro collagen/PLGA bimodal fibrous patches for myocardial regeneration. Biomater Res 2022; 26:79. [PMID: 36514148 DOI: 10.1186/s40824-022-00319-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 11/13/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Although the use of cardiac patches is still controversial, cardiac patch has the significance in the field of the tissue engineered cardiac regeneration because it overcomes several shortcomings of intra-myocardial injection by providing a template for cells to form a cohesive sheet. So far, fibrous scaffolds fabricated using electrospinning technique have been increasingly explored for preparation of cardiac patches. One of the problems with the use of electrospinning is that nanofibrous structures hardly allow the infiltration of cells for development of 3D tissue construct. In this respect, we have prepared novel bi-modal electrospun scaffolds as a feasible strategy to address the challenges in cardiac tissue engineering . METHODS Nano/micro bimodal composite fibrous patch composed of collagen and poly (D, L-lactic-co-glycolic acid) (Col/PLGA) was fabricated using an independent nozzle control multi-electrospinning apparatus, and its feasibility as the stem cell laden cardiac patch was systemically investigated. RESULTS Nano/micro bimodal distributions of Col/PLGA patches without beaded fibers were obtained in the range of the 4-6% collagen concentration. The poor mechanical properties of collagen and the hydrophobic property of PLGA were improved by co-electrospinning. In vitro experiments using bone marrow-derived mesenchymal stem cells (BMSCs) revealed that Col/PLGA showed improved cyto-compatibility and proliferation capacity compared to PLGA, and their extent increased with increase in collagen content. The results of tracing nanoparticle-labeled as well as GFP transfected BMSCs strongly support that Col/PLGA possesses the long-term stem cells retention capability, thereby allowing stem cells to directly function as myocardial and vascular endothelial cells or to secrete the recovery factors, which in turn leads to improved heart function proved by histological and echocardiographic findings. CONCLUSION Col/PLGA bimodal cardiac patch could significantly attenuate cardiac remodeling and fully recover the cardiac function, as a consequence of their potent long term stem cell engraftment capability.
Collapse
|
6
|
Liu D, Zhan Y, Ono K, Yin Y, Wang L, Wei M, Ji L, Liu M, Liu G, Zhou X, Zheng M. Pharmacological activation of estrogenic receptor G protein-coupled receptor 30 attenuates angiotensin II-induced atrial fibrosis in ovariectomized mice by modulating TGF-β1/smad pathway. Mol Biol Rep 2022; 49:6341-6355. [PMID: 35513634 DOI: 10.1007/s11033-022-07444-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 04/01/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND G-protein-coupled ER (GPR30) plays an important role in cardioprotection. Recent studies have shown that the GPR30-specific agonist G-1 reduces the degree of myocardial fibrosis in rats with myocardial infarction, reduces the morbidity associated with atrial fibrillation, and inhibits the proliferation of cardiac fibroblasts in animal experiments. Nevertheless, the underlying mechanism of myocardial fibrosis and atrial fibrillation remains unclear. In this study, we explored the mechanism underlying the effect of GPR30 on atrial fibrosis and atrial fibrillation in OVX mice. METHODS We established an animal model of atrial fibrillation induced by Ang II (derived from OVX C57BL/6 female mice) and observed the role of G-1 in cardiac function by echocardiography, hemodynamics, morphology and fibrosis-related and apoptosis-related protein expression by Masson's trichrome, immunofluorescence, western blotting and TUNEL staining. RESULTS Echocardiography and body surface ECG showed that G-1 combined with Ang II significantly reduced atrial fibrosis and atrial fibrillation compared to Ang II alone. The G-1 treatment group exhibited changes in the mRNA and protein expression of apoptosis-related genes. Moreover, G-1 treatment also altered the levels of inflammation-related proteins and mRNAs. In primary cultured cardiac fibroblasts (CFSs), proliferation was significantly increased in response to Ang II, and G-1 inhibited cell proliferation and apoptosis. CONCLUSION GPR30 is a potential therapeutic target for alleviating atrial fibrosis in OVX mice by upregulating Smad7 expression to inhibit the TGF-β/Smad pathway.
Collapse
Affiliation(s)
- Da Liu
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China
- Graduate School of Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050000, Hebei, China
| | - Yinge Zhan
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China
| | - Katsushige Ono
- Department of Pathological Physiology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama, Oita, 879-5593, Japan
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China
| | - Le Wang
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China
| | - Mei Wei
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China
| | - Lishuang Ji
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China
| | - Mei Liu
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China
| | - Xin Zhou
- Department of Cardiology, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District 300052, Tianjin, China
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medicical University, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China.
- Hebei Key Laboratory of Heart and Metabolism, 89 Donggang Road, Yuhua District, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
7
|
Yao J, Wu XY, Yu Q, Yang SF, Yuan J, Zhang ZQ, Xue JS, Jiang Q, Chen MB, Xue GH, Cao C. The requirement of phosphoenolpyruvate carboxykinase 1 for angiogenesis in vitro and in vivo. SCIENCE ADVANCES 2022; 8:eabn6928. [PMID: 35622925 PMCID: PMC9140980 DOI: 10.1126/sciadv.abn6928] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/12/2022] [Indexed: 05/23/2023]
Abstract
We here examined the potential biological function of phosphoenolpyruvate carboxykinase 1 (PCK1) in angiogenesis. shRNA- or CRISPR-Cas9-induced PCK1 depletion potently inhibited endothelial cell proliferation, migration, sprouting, and tube formation, whereas ectopic PCK1 overexpression exerted opposite activity. In HUVECs, Gαi3 expression and Akt activation were decreased following PCK1 depletion, but were augmented by ectopic PCK1 overexpression. In vivo, retinal expression of PCK1 gradually increased from postnatal day 1 (P1) to P5. The intravitreous injection of endothelial-specific PCK1 shRNA adenovirus at P1 potently inhibited the radial extension of vascular plexus at P5. Conditional endothelial knockdown of PCK1 in adult mouse retina increased vascular leakage and the number of acellular capillaries while decreasing the number of RGCs in murine retinas. In diabetic retinopathy patients, PCK1 mRNA and protein levels were up-regulated in retinal tissues. Together, PCK1 is essential for angiogenesis possibly by mediating Gαi3 expression and Akt activation.
Collapse
Affiliation(s)
- Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Xin-yuan Wu
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Qing Yu
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Shuo-fei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Jin Yuan
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Zhi-qing Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| | - Jin-song Xue
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
| | - Min-bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Guan-hua Xue
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Cong Cao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
8
|
Amini P, Amrovani M, Nassaj ZS, Ajorlou P, Pezeshgi A, Ghahrodizadehabyaneh B. Hypertension: Potential Player in Cardiovascular Disease Incidence in Preeclampsia. Cardiovasc Toxicol 2022; 22:391-403. [PMID: 35347585 DOI: 10.1007/s12012-022-09734-w] [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: 10/23/2021] [Accepted: 02/17/2022] [Indexed: 11/28/2022]
Abstract
Preeclampsia (PE) is one of the complications, that threatens pregnant mothers during pregnancy. According to studies, it accounts for 3-7% of all pregnancies, and also is effective in preterm delivery. PE is the third leading cause of death in pregnant women. High blood pressure in PE can increase the risk of developing cardiovascular disease (CVD) in cited individuals, and is one of the leading causes of death in PE individuals. Atrial natriuretic peptide (ANP), Renin-Angiotensin system and nitric oxide (NO) are some of involved factors in regulating blood pressure. Therefore, by identifying the signaling pathways, that are used by these molecules to regulate and modulate blood pressure, appropriate treatment strategies can be provided to reduce blood pressure through target therapy in PE individuals; consequently, it can reduce CVD risk and mortality.
Collapse
Affiliation(s)
- Parya Amini
- Atherosclerosis Research Center, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Mehran Amrovani
- High Institute for Education and Research in Transfusion Medicine, Tehran, Iran
| | - Zohre Saleh Nassaj
- Center for Health Related Social and Behavioral Sciences Research, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Parisa Ajorlou
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Aiyoub Pezeshgi
- Internal Medicine Department, Zanjan University of Medical Sciences, Zanjan, Iran.
| | | |
Collapse
|
9
|
Zhang C, Zhao H, Song X, Wang J, Zhao S, Deng H, He L, Zhou X, Yin X, Zhang K, Zhang Y, Wu Z, Chen Q, Du J, Yu D, Zhang S, Deng W. Transcription factor GATA4 drives RNA polymerase III-directed transcription and transformed cell proliferation through a filamin A/GATA4/SP1 pathway. J Biol Chem 2022; 298:101581. [PMID: 35038452 PMCID: PMC8857480 DOI: 10.1016/j.jbc.2022.101581] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 11/27/2022] Open
Abstract
RNA polymerase III (pol III) products play fundamental roles in a variety of cellular processes, including protein synthesis and cancer cell proliferation. In addition, dysregulation of pol III-directed transcription closely correlates with tumorigenesis. It is therefore of interest to identify novel pathways or factors governing pol III-directed transcription. Here, we show that transcription factor (TF) GATA binding protein 4 (GATA4) expression in SaOS2 cells was stimulated by the silencing of filamin A (FLNA), a repressor of pol III-directed transcription, suggesting that GATA4 is potentially associated with the regulation of pol III-directed transcription. Indeed, we show that GATA4 expression positively correlates with pol III-mediated transcription and tumor cell proliferation. Mechanistically, we found that GATA4 depletion inhibits the occupancies of the pol III transcription machinery factors at the loci of pol III target genes by reducing expression of both TFIIIB subunit TFIIB-related factor 1 and TFIIIC subunit general transcription factor 3C subunit 2 (GTF3C2). GATA4 has been shown to activate specificity factor 1 (Sp1) gene transcription by binding to the Sp1 gene promoter, and Sp1 has been confirmed to activate pol III gene transcription by directly binding to both Brf1 and Gtf3c2 gene promoters. Thus, the findings from this study suggest that GATA4 links FLNA and Sp1 signaling to form an FLNA/GATA4/Sp1 axis to modulate pol III-directed transcription and transformed cell proliferation. Taken together, these results provide novel insights into the regulatory mechanism of pol III-directed transcription.
Collapse
Affiliation(s)
- Cheng Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Houliang Zhao
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaoye Song
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Juan Wang
- School of Materials and Metallurgy, Wuhan University of Science and Technology, Wuhan, China
| | - Shasha Zhao
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Huan Deng
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Liu He
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiangyu Zhou
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaomei Yin
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Kewei Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Yue Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Zhongyu Wu
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Qiyue Chen
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Jiannan Du
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Deen Yu
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China
| | - Shihua Zhang
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.
| | - Wensheng Deng
- School of Life Science and Health, Wuhan University of Science and Technology, Wuhan, China.
| |
Collapse
|
10
|
Zhao K, Mao Y, Ye X, Ma J, Sun L, Li P, Li Y. MicroRNA-210-5p alleviates cardiac fibrosis via targeting transforming growth factor-beta type I receptor in rats on high sodium chloride (NaCl)-based diet. Eur J Pharmacol 2021; 912:174587. [PMID: 34678242 DOI: 10.1016/j.ejphar.2021.174587] [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: 07/30/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022]
Abstract
The present study was designed to explore whether high sodium chloride (NaCl)-based diet (HSD) caused cardiac fibrosis regardless of blood pressure in Sprague-Dawley (SD) rats, and to further determine the effects and the underlying mechanisms of microRNA (miR)-210-5p on HSD-induced cardiac fibrosis in rats or NaCl-induced cardiac fibroblast activation in neonatal rat cardiac fibroblasts (NRCFs). The SD rats received 8% HSD, and NRCFs were treated with NaCl. The levels of collagen I, alpha-smooth muscle actin (α-SMA) and transforming growth factor-beta 1 (TGF-β1) were increased in the heart of hypertension (HTN), hypertension-prone (HP) and hypertension-resistant (HR) rats on HSD in vivo. NaCl increased the levels of collagen I, α-SMA and TGF-β1 in NRCFs in vitro. The level of miR-210-5p was reduced in both NBD-induced rats' hearts and NaCl-treated NRCFs, which was consistent with the results of miR high-throughput sequencing in NRCFs. The HSD or NaCl-induced increases of collagen I, α-SMA and TGF-β1 were inhibited by miR-210-5p agomiR in vitro and in vivo, respectively. miR-210-5p antagomiR could mimic the pathological effects of NaCl in NRCFS. Bioinformatics analysis and luciferase reporter assays demonstrated that TGF-β type I receptor (TGFBR1) was a direct target gene of miR-210-5p. These results indicated that HSD resulted in cardiac fibrosis regardless of blood pressure. The upregulation of miR-210-5p could attenuate cardiac fibroblast activation in NRCFS via targeting TGFBR1. Thus, upregulating miR-210-5p might be a strategy for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yukang Mao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoman Ye
- Intensive Care Unit, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiazheng Ma
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Litao Sun
- SEU-FEI Nano-Pico Center, Key Laboratory of MEMS of Ministry of Education, Southeast University, Nanjing, 210096, China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yong Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
11
|
Triptolide impairs glycolysis by suppressing GATA4/Sp1/PFKP signaling axis in mouse Sertoli cells. Toxicol Appl Pharmacol 2021; 425:115606. [PMID: 34087332 DOI: 10.1016/j.taap.2021.115606] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 11/23/2022]
Abstract
Triptolide (TP), a primary bioactive ingredient isolated from the traditional Chinese herbal medicine Tripterygium wilfordii Hook. F. (TWHF), has attracted great interest for its therapeutic biological activities in inflammation and autoimmune disease. However, its clinical use is limited by severe testicular toxicity, and the underlying mechanism has not been elucidated. Our preliminary evidence demonstrated that TP disrupted glucose metabolism and caused testicular toxicity. During spermatogenesis, Sertoli cells (SCs) provide lactate as an energy source to germ cells by glycolysis. The transcription factors GATA-binding protein 4 (GATA4) and specificity protein 1 (Sp1) can regulate glycolysis. Based on this evidence, we speculate that TP causes abnormal glycolysis in SCs by influencing the expression of the transcription factors GATA4 and Sp1. The mechanism of TP-induced testicular toxicity was investigated in vitro and in vivo. The data indicated that TP decreased glucose consumption, lactate production, and the mRNA levels of glycolysis-related transporters and enzymes. TP also downregulated the protein expression of the transcription factors GATA4 and Sp1, as well as the glycolytic enzyme phosphofructokinase platelet (PFKP). Phosphorylated GATA4 and nuclear GATA4 protein levels were reduced in a dose- and time-dependent manner after TP incubation. Similar effects were observed in shGata4-treated TM4 cells and BALB/c mice administered 0.4 mg/kg TP for 28 days, and glycolysis was also inhibited. Gata4 knockdown downregulated Sp1 and PFKP expression. Furthermore, the Sp1 inhibitor plicamycin inhibited PFKP protein levels in TM4 cells. In conclusion, TP inhibited GATA4-mediated glycolysis by suppressing Sp1-dependent PFKP expression in SCs and caused testicular toxicity.
Collapse
|
12
|
Jurado Acosta A, Rysä J, Szabo Z, Moilanen AM, Serpi R, Ruskoaho H. Phosphorylation of GATA4 at serine 105 is required for left ventricular remodelling process in angiotensin II-induced hypertension in rats. Basic Clin Pharmacol Toxicol 2020; 127:178-195. [PMID: 32060996 PMCID: PMC7496669 DOI: 10.1111/bcpt.13398] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/11/2020] [Accepted: 02/12/2020] [Indexed: 12/25/2022]
Abstract
In this study, we investigated whether local intramyocardial GATA4 overexpression affects the left ventricular (LV) remodelling process and the importance of phosphorylation at serine 105 (S105) for the actions of GATA4 in an angiotensin II (AngII)‐induced hypertension rat model. Adenoviral constructs overexpressing wild‐type GATA4 or GATA4 mutated at S105 were delivered into the anterior LV free wall. AngII (33.3 µg/kg/h) was administered via subcutaneously implanted minipumps. Cardiac function and structure were examined by echocardiography, followed by histological immunostainings of LV sections and gene expression measurements by RT‐qPCR. The effects of GATA4 on cultured neonatal rat ventricular fibroblasts were evaluated. In AngII‐induced hypertension, GATA4 overexpression repressed fibrotic gene expression, reversed the hypertrophic adult‐to‐foetal isoform switch of myofibrillar genes and prevented apoptosis, whereas histological fibrosis was not affected. Overexpression of GATA4 mutated at S105 resulted in LV chamber dilatation, cardiac dysfunction and had minor effects on expression of myocardial remodelling genes. Fibrotic gene expression in cardiac fibroblasts was differently affected by overexpression of wild‐type or mutated GATA4. Our results indicate that GATA4 reduces AngII‐induced responses by interfering with pro‐fibrotic and hypertrophic gene expressions. GATA4 actions on LV remodelling and fibroblasts are dependent on phosphorylation site S105.
Collapse
Affiliation(s)
- Alicia Jurado Acosta
- Pharmacology and Toxicology, Biomedicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Zoltan Szabo
- Pharmacology and Toxicology, Biomedicine Research Unit, Faculty of Medicine, University of Oulu, Oulu, Finland
| | - Anne-Mari Moilanen
- Cancer and Translational Medicine Research Unit, University of Oulu, Oulu, Finland.,Oulu University Hospital and Medical Research Center Oulu, Oulu, Finland
| | - Raisa Serpi
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland
| | - Heikki Ruskoaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|