1
|
Zhou L, Mei S, Ma X, Wuyun Q, Cai Z, Chen C, Ding H, Yan J. Multi-omics insights into the pathogenesis of diabetic cardiomyopathy: epigenetic and metabolic profiles. Epigenomics 2025; 17:33-48. [PMID: 39623870 PMCID: PMC11727868 DOI: 10.1080/17501911.2024.2435257] [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: 09/22/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
AIM Diabetic cardiomyopathy (DbCM), a complex metabolic disease, greatly threatens human health due to therapeutic limitations. Multi-omics approaches facilitate the elucidation of its intrinsic pathological changes. METHODS Metabolomics, RNA-seq, proteomics, and assay of transposase-accessible chromatin (ATAC-seq) were utilized to elucidate multidimensional molecular alterations in DbCM. RESULTS In the heart and plasma of mice with DbCM, metabolomic analysis demonstrated significant differences in branched-chain amino acids (BCAAs) and lipids. Subsequent RNA-seq and proteomics showed that the key genes, including BCKDHB, PPM1K, Cpt1b, Fabp4, Acadm, Acadl, Acadvl, HADH, HADHA, HADHB, Eci1, Eci2, PDK4, and HMGCS2, were aberrantly regulated, contributing to the disorder of BCAAs and fatty acids. ATAC-seq analysis underscored the pivotal role of epigenetic regulation by revealing dynamic shifts in chromatin accessibility and a robust positive correlation with gene expression patterns in diabetic cardiomyopathy mice. Furthermore, motif analysis identified that KLF15 as a critical transcription factor in DbCM, regulating the core genes implicated with BCAAs metabolism. CONCLUSION Our research delved into the metabolic alterations and epigenetic landscape and revealed that KLF15 may be a promising candidate for therapeutic intervention in DbCM.
Collapse
Affiliation(s)
- Li Zhou
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Shuai Mei
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiaozhu Ma
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Qidamugai Wuyun
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Ziyang Cai
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Chen Chen
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Hu Ding
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jiangtao Yan
- Division of Cardiology, Departments of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
2
|
Bereczki Z, Benczik B, Balogh OM, Marton S, Puhl E, Pétervári M, Váczy-Földi M, Papp ZT, Makkos A, Glass K, Locquet F, Euler G, Schulz R, Ferdinandy P, Ágg B. Mitigating off-target effects of small RNAs: conventional approaches, network theory and artificial intelligence. Br J Pharmacol 2025; 182:340-379. [PMID: 39293936 DOI: 10.1111/bph.17302] [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: 11/30/2023] [Revised: 05/07/2024] [Accepted: 06/17/2024] [Indexed: 09/20/2024] Open
Abstract
Three types of highly promising small RNA therapeutics, namely, small interfering RNAs (siRNAs), microRNAs (miRNAs) and the RNA subtype of antisense oligonucleotides (ASOs), offer advantages over small-molecule drugs. These small RNAs can target any gene product, opening up new avenues of effective and safe therapeutic approaches for a wide range of diseases. In preclinical research, synthetic small RNAs play an essential role in the investigation of physiological and pathological pathways as silencers of specific genes, facilitating discovery and validation of drug targets in different conditions. Off-target effects of small RNAs, however, could make it difficult to interpret experimental results in the preclinical phase and may contribute to adverse events of small RNA therapeutics. Out of the two major types of off-target effects we focused on the hybridization-dependent, especially on the miRNA-like off-target effects. Our main aim was to discuss several approaches, including sequence design, chemical modifications and target prediction, to reduce hybridization-dependent off-target effects that should be considered even at the early development phase of small RNA therapy. Because there is no standard way of predicting hybridization-dependent off-target effects, this review provides an overview of all major state-of-the-art computational methods and proposes new approaches, such as the possible inclusion of network theory and artificial intelligence (AI) in the prediction workflows. Case studies and a concise survey of experimental methods for validating in silico predictions are also presented. These methods could contribute to interpret experimental results, to minimize off-target effects and hopefully to avoid off-target-related adverse events of small RNA therapeutics. LINKED ARTICLES: This article is part of a themed issue Non-coding RNA Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v182.2/issuetoc.
Collapse
Affiliation(s)
- Zoltán Bereczki
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bettina Benczik
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Olivér M Balogh
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Szandra Marton
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Eszter Puhl
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
| | - Mátyás Pétervári
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Sanovigado Kft, Budapest, Hungary
| | - Máté Váczy-Földi
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zsolt Tamás Papp
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - András Makkos
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Kimberly Glass
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Fabian Locquet
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Gerhild Euler
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Giessen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
3
|
Heger J, Partsch S, Harjung C, Varga ZV, Baranyai T, Weiß J, Kremer L, Locquet F, Leszek P, Ágg B, Benczik B, Ferdinandy P, Schulz R, Euler G. YB-1 Is a Novel Target for the Inhibition of α-Adrenergic-Induced Hypertrophy. Int J Mol Sci 2023; 25:401. [PMID: 38203580 PMCID: PMC10778708 DOI: 10.3390/ijms25010401] [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: 10/05/2023] [Revised: 12/20/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Cardiac hypertrophy resulting from sympathetic nervous system activation triggers the development of heart failure. The transcription factor Y-box binding protein 1 (YB-1) can interact with transcription factors involved in cardiac hypertrophy and may thereby interfere with the hypertrophy growth process. Therefore, the question arises as to whether YB-1 influences cardiomyocyte hypertrophy and might thereby influence the development of heart failure. YB-1 expression is downregulated in human heart biopsies of patients with ischemic cardiomyopathy (n = 8), leading to heart failure. To study the impact of reduced YB-1 in cardiac cells, we performed small interfering RNA (siRNA) experiments in H9C2 cells as well as in adult cardiomyocytes (CMs) of rats. The specificity of YB-1 siRNA was analyzed by a miRNA-like off-target prediction assay identifying potential genes. Testing three high-scoring genes by transfecting cardiac cells with YB-1 siRNA did not result in downregulation of these genes in contrast to YB-1, whose downregulation increased hypertrophic growth. Hypertrophic growth was mediated by PI3K under PE stimulation, as well by downregulation with YB-1 siRNA. On the other hand, overexpression of YB-1 in CMs, caused by infection with an adenovirus encoding YB-1 (AdYB-1), prevented hypertrophic growth under α-adrenergic stimulation with phenylephrine (PE), but not under stimulation with growth differentiation factor 15 (GDF15; n = 10-16). An adenovirus encoding the green fluorescent protein (AdGFP) served as the control. YB-1 overexpression enhanced the mRNA expression of the Gq inhibitor regulator of G-protein signaling 2 (RGS2) under PE stimulation (n = 6), potentially explaining its inhibitory effect on PE-induced hypertrophic growth. This study shows that YB-1 protects cardiomyocytes against PE-induced hypertrophic growth. Like in human end-stage heart failure, YB-1 downregulation may cause the heart to lose its protection against hypertrophic stimuli and progress to heart failure. Therefore, the transcription factor YB-1 is a pivotal signaling molecule, providing perspectives for therapeutic approaches.
Collapse
Affiliation(s)
- Jacqueline Heger
- Institute of Physiology, Justus Liebig University, 35392 Giessen, Germany; (S.P.); (C.H.); (J.W.); (L.K.); (F.L.); (R.S.); (G.E.)
| | - Stefan Partsch
- Institute of Physiology, Justus Liebig University, 35392 Giessen, Germany; (S.P.); (C.H.); (J.W.); (L.K.); (F.L.); (R.S.); (G.E.)
| | - Claudia Harjung
- Institute of Physiology, Justus Liebig University, 35392 Giessen, Germany; (S.P.); (C.H.); (J.W.); (L.K.); (F.L.); (R.S.); (G.E.)
| | - Zoltán V. Varga
- HCEMM-SU Cardiometabolic Immunology Research Group, 1094 Budapest, Hungary;
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1094 Budapest, Hungary; (T.B.); (B.Á.); (B.B.); (P.F.)
| | - Tamás Baranyai
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1094 Budapest, Hungary; (T.B.); (B.Á.); (B.B.); (P.F.)
| | - Johannes Weiß
- Institute of Physiology, Justus Liebig University, 35392 Giessen, Germany; (S.P.); (C.H.); (J.W.); (L.K.); (F.L.); (R.S.); (G.E.)
| | - Lea Kremer
- Institute of Physiology, Justus Liebig University, 35392 Giessen, Germany; (S.P.); (C.H.); (J.W.); (L.K.); (F.L.); (R.S.); (G.E.)
| | - Fabian Locquet
- Institute of Physiology, Justus Liebig University, 35392 Giessen, Germany; (S.P.); (C.H.); (J.W.); (L.K.); (F.L.); (R.S.); (G.E.)
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, Cardinal Stefan Wyszyński Institute of Cardiology, 04-628 Warszawa, Poland;
| | - Bence Ágg
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1094 Budapest, Hungary; (T.B.); (B.Á.); (B.B.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Bettina Benczik
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1094 Budapest, Hungary; (T.B.); (B.Á.); (B.B.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Péter Ferdinandy
- Cardiometabolic and MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1094 Budapest, Hungary; (T.B.); (B.Á.); (B.B.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University, 35392 Giessen, Germany; (S.P.); (C.H.); (J.W.); (L.K.); (F.L.); (R.S.); (G.E.)
| | - Gerhild Euler
- Institute of Physiology, Justus Liebig University, 35392 Giessen, Germany; (S.P.); (C.H.); (J.W.); (L.K.); (F.L.); (R.S.); (G.E.)
| |
Collapse
|
4
|
Ruppert M, Korkmaz-Icöz S, Benczik B, Ágg B, Nagy D, Bálint T, Sayour AA, Oláh A, Barta BA, Benke K, Ferdinandy P, Karck M, Merkely B, Radovits T, Szabó G. Pressure overload-induced systolic heart failure is associated with characteristic myocardial microRNA expression signature and post-transcriptional gene regulation in male rats. Sci Rep 2023; 13:16122. [PMID: 37752166 PMCID: PMC10522609 DOI: 10.1038/s41598-023-43171-1] [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: 02/06/2023] [Accepted: 09/20/2023] [Indexed: 09/28/2023] Open
Abstract
Although systolic function characteristically shows gradual impairment in pressure overload (PO)-evoked left ventricular (LV) hypertrophy (LVH), rapid progression to congestive heart failure (HF) occurs in distinct cases. The molecular mechanisms for the differences in maladaptation are unknown. Here, we examined microRNA (miRNA) expression and miRNA-driven posttranscriptional gene regulation in the two forms of PO-induced LVH (with/without systolic HF). PO was induced by aortic banding (AB) in male Sprague-Dawley rats. Sham-operated animals were controls. The majority of AB animals demonstrated concentric LVH and slightly decreased systolic function (termed as ABLVH). In contrast, in some AB rats severely reduced ejection fraction, LV dilatation and increased lung weight-to-tibial length ratio was noted (referred to as ABHF). Global LV miRNA sequencing revealed fifty differentially regulated miRNAs in ABHF compared to ABLVH. Network theoretical miRNA-target analysis predicted more than three thousand genes with miRNA-driven dysregulation between the two groups. Seventeen genes with high node strength value were selected for target validation, of which five (Fmr1, Zfpm2, Wasl, Ets1, Atg16l1) showed decreased mRNA expression in ABHF by PCR. PO-evoked systolic HF is associated with unique miRNA alterations, which negatively regulate the mRNA expression of Fmr1, Zfmp2, Wasl, Ets1 and Atg16l1.
Collapse
Affiliation(s)
- Mihály Ruppert
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary.
| | - Sevil Korkmaz-Icöz
- Department of Cardiac Surgery, University of Heidelberg, Heidelberg, Germany
- Department of Cardiac Surgery, University Hospital Halle (Saale), Halle, Germany
| | - Bettina Benczik
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Dávid Nagy
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary
| | - Tímea Bálint
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary
| | - Alex Ali Sayour
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary
| | - Attila Oláh
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary
| | - Bálint András Barta
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary
| | - Kálmán Benke
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Matthias Karck
- Department of Cardiac Surgery, University of Heidelberg, Heidelberg, Germany
| | - Béla Merkely
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary
| | - Tamás Radovits
- Experimental Research Laboratory, Heart and Vascular Center, Semmelweis University, Városmajor u. 68, 1122, Budapest, Hungary
| | - Gábor Szabó
- Department of Cardiac Surgery, University of Heidelberg, Heidelberg, Germany
- Department of Cardiac Surgery, University Hospital Halle (Saale), Halle, Germany
| |
Collapse
|
5
|
Song R, Ma S, Xu J, Ren X, Guo P, Liu H, Li P, Yin F, Liu M, Wang Q, Yu L, Liu J, Duan B, Rahman NA, Wołczyński S, Li G, Li X. A novel polypeptide encoded by the circular RNA ZKSCAN1 suppresses HCC via degradation of mTOR. Mol Cancer 2023; 22:16. [PMID: 36691031 PMCID: PMC9869513 DOI: 10.1186/s12943-023-01719-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND hsa_circ_0001727 (circZKSCAN1) has been reported to be a tumor-associated circRNA by sponging microRNAs. Intriguingly, we found that circZKSCAN1 encoded a secretory peptide (circZKSaa) in the liver. The present study aims to elucidate the potential role and molecular mechanism of circZKSaa in the regulation of hepatocellular carcinoma (HCC) progression. METHODS The circRNA profiling datasets (RNA-seq data GSE143233 and GSE140202) were reanalyzed and circZKSCAN1 was selected for further study. Mass spectrometry, polysome fractionation assay, dual-luciferase reporter, and a series of experiments showed that circZKSCAN1 encodes circZKSaa. Cell proliferation, apoptosis, and tumorigenesis in nude mice were examined to investigate the functions of circZKSaa. Mechanistically, the relationship between the circZKSaa and mTOR in HCC was verified by immunoprecipitation analyses, mass spectrometry, and immunofluorescence staining analyses. RESULTS Receiver operating characteristic (ROC) analysis demonstrated that the secretory peptide circZKSaa encoded by circZKSCAN1 might be the potential biomarker for HCC tissues. Through a series of experiments, we found that circZKSaa inhibited HCC progression and sensitize HCC cells to sorafenib. Mechanistically, we found that the sponge function of circZKSCAN1 to microRNA is weak in HCC, while overexpression of circZKSaa promoted the interaction of FBXW7 with the mammalian target of rapamycin (mTOR) to promote the ubiquitination of mTOR, thereby inhibiting the PI3K/AKT/mTOR pathway. Furthermore, we found that the high expression of cicZKSCAN1 in sorafenib-treated HCC cells was regulated by QKI-5. CONCLUSIONS These results reveal that a novel circZKSCAN1-encoded peptide acts as a tumor suppressor on PI3K/AKT/mTOR pathway, and sensitizes HCC cells to sorafenib via ubiquitination of mTOR. These findings demonstrated that circZKSaa has the potential to serve as a therapeutic target and biomarker for HCC treatment.
Collapse
Affiliation(s)
- Runjie Song
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Shuoqian Ma
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Jiajia Xu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Xin Ren
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Peilan Guo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Huijiao Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Peng Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Fan Yin
- Department of Oncology, The Second Medical Centre & National Clinical Research Center of Geriatric Disease, Chinese PLA General Hospital, Beijing, 100071, China
| | - Mei Liu
- Department of Pathology, Chinese PLA General Hospital, Beijing, 100071, China
| | - Qiang Wang
- Department of Urology, Peking University People's Hospital, Beijing, 100044, China
| | - Lei Yu
- Department of Thoracic Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jiali Liu
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Binwei Duan
- Department of General Surgery CenterBeijing You An Hospital, Clinical Center for Liver Cancer, Capital Medical University, Beijing, China
| | - Nafis A Rahman
- Department of Physiology, Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Sławomir Wołczyński
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland
| | - Guangming Li
- Department of General Surgery CenterBeijing You An Hospital, Clinical Center for Liver Cancer, Capital Medical University, Beijing, China
| | - Xiangdong Li
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, 100193, China.
- Department of Reproduction and Gynecological Endocrinology, Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
6
|
Mameri A, Côté J. JAZF1: A metabolic actor subunit of the NuA4/TIP60 chromatin modifying complex. Front Cell Dev Biol 2023; 11:1134268. [PMID: 37091973 PMCID: PMC10119425 DOI: 10.3389/fcell.2023.1134268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/29/2023] [Indexed: 04/25/2023] Open
Abstract
The multisubunit NuA4/TIP60 complex is a lysine acetyltransferase, chromatin modifying factor and gene co-activator involved in diverse biological processes. The past decade has seen a growing appreciation for its role as a metabolic effector and modulator. However, molecular insights are scarce and often contradictory, underscoring the need for further mechanistic investigation. A particularly exciting route emerged with the recent identification of a novel subunit, JAZF1, which has been extensively linked to metabolic homeostasis. This review summarizes the major findings implicating NuA4/TIP60 in metabolism, especially in light of JAZF1 as part of the complex.
Collapse
|
7
|
Makkos A, Ágg B, Varga ZV, Giricz Z, Gyöngyösi M, Lukovic D, Schulz R, Barteková M, Görbe A, Ferdinandy P. Molecular Network Approach Reveals Rictor as a Central Target of Cardiac ProtectomiRs. Int J Mol Sci 2021; 22:ijms22179539. [PMID: 34502448 PMCID: PMC8430799 DOI: 10.3390/ijms22179539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022] Open
Abstract
Cardioprotective medications are still unmet clinical needs. We have previously identified several cardioprotective microRNAs (termed ProtectomiRs), the mRNA targets of which may reveal new drug targets for cardioprotection. Here we aimed to identify key molecular targets of ProtectomiRs and confirm their association with cardioprotection in a translational pig model of acute myocardial infarction (AMI). By using a network theoretical approach, we identified 882 potential target genes of 18 previously identified protectomiRs. The Rictor gene was the most central and it was ranked first in the protectomiR-target mRNA molecular network with the highest node degree of 5. Therefore, Rictor and its targeting microRNAs were further validated in heart samples obtained from a translational pig model of AMI and cardioprotection induced by pre- or postconditioning. Three out of five Rictor-targeting pig homologue of rat ProtectomiRs showed significant upregulation in postconditioned but not in preconditioned pig hearts. Rictor was downregulated at the mRNA and protein level in ischemic postconditioning but not in ischemic preconditioning. This is the first demonstration that Rictor is the central molecular target of ProtectomiRs and that decreased Rictor expression may regulate ischemic postconditioning-, but not preconditioning-induced acute cardioprotection. We conclude that Rictor is a potential novel drug target for acute cardioprotection.
Collapse
Affiliation(s)
- András Makkos
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (A.M.); (B.Á.); (Z.V.V.); (Z.G.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Bence Ágg
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (A.M.); (B.Á.); (Z.V.V.); (Z.G.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Zoltán V. Varga
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (A.M.); (B.Á.); (Z.V.V.); (Z.G.); (P.F.)
- HCEMM-SU Cardiometabolic Immunology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
| | - Zoltán Giricz
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (A.M.); (B.Á.); (Z.V.V.); (Z.G.); (P.F.)
| | - Mariann Gyöngyösi
- Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (M.G.); (D.L.)
| | - Dominika Lukovic
- Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (M.G.); (D.L.)
| | - Rainer Schulz
- Institute of Physiology, Justus Liebig University Giessen, 35392 Giessen, Germany;
| | - Monika Barteková
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, Dúbravská cesta 9, 84104 Bratislava, Slovakia;
- Institute of Physiology, Comenius University in Bratislava, 81108 Bratislava, Slovakia
| | - Anikó Görbe
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (A.M.); (B.Á.); (Z.V.V.); (Z.G.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
- Correspondence:
| | - Péter Ferdinandy
- Cardiovascular and Metabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (A.M.); (B.Á.); (Z.V.V.); (Z.G.); (P.F.)
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary
- Pharmahungary Group, 6722 Szeged, Hungary
| |
Collapse
|
8
|
Makkos A, Ágg B, Petrovich B, Varga ZV, Görbe A, Ferdinandy P. Systematic review and network analysis of microRNAs involved in cardioprotection against myocardial ischemia/reperfusion injury and infarction: Involvement of redox signalling. Free Radic Biol Med 2021; 172:237-251. [PMID: 33965565 DOI: 10.1016/j.freeradbiomed.2021.04.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/31/2021] [Accepted: 04/27/2021] [Indexed: 01/12/2023]
Abstract
Although myocardial ischemia-reperfusion injury (I/R) and its pathological consequences are the leading cause of morbidity and mortality worldwide, cardioprotective therapeutics are still not on the market. Oxidative stress, a major contributing factor to myocardial I/R, changes transcription of coding and non-coding RNAs, alters post-transcriptional modulations, and regulate protein function. MicroRNA (miRNA) expression can be altered by oxidative stress and microRNAs may also regulate cytoprotective mechanisms and exert cardioprotection againts I/R. Transcriptomic analysis of I/R and oxidative stress-induced alterations followed by microRNA-mRNA target interaction network analysis may reveal microRNAs and their mRNA targets that may play a role in cardioprotection and serve as microRNA therapeutics or novel molecular targets for further drug development. Here we provide a summary of a systematic literature review and in silico molecular network analysis to reveal important cardioprotective microRNAs and their molecular targets that may provide cardioprotection via regulation of redox signalling.
Collapse
Affiliation(s)
- András Makkos
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary.
| | - Bence Ágg
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary; Pharmahungary Group, 6722, Szeged, Hungary.
| | - Balázs Petrovich
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary.
| | - Zoltán V Varga
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, 1089, Budapest, Hungary.
| | - Anikó Görbe
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary; Pharmahungary Group, 6722, Szeged, Hungary.
| | - Péter Ferdinandy
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary; Pharmahungary Group, 6722, Szeged, Hungary.
| |
Collapse
|
9
|
Róka B, Tod P, Kaucsár T, Bukosza ÉN, Vörös I, Varga ZV, Petrovich B, Ágg B, Ferdinandy P, Szénási G, Hamar P. Delayed Contralateral Nephrectomy Halted Post-Ischemic Renal Fibrosis Progression and Inhibited the Ischemia-Induced Fibromir Upregulation in Mice. Biomedicines 2021; 9:biomedicines9070815. [PMID: 34356879 PMCID: PMC8301422 DOI: 10.3390/biomedicines9070815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 07/06/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
(1) Background: Ischemia reperfusion (IR) is the leading cause of acute kidney injury (AKI) and results in predisposition to chronic kidney disease. We demonstrated that delayed contralateral nephrectomy (Nx) greatly improved the function of the IR-injured kidney and decelerated fibrosis progression. Our aim was to identify microRNAs (miRNA/miR) involved in this process. (2) Methods: NMRI mice were subjected to 30 min of renal IR and one week later to Nx/sham surgery. The experiments were conducted for 7-28 days after IR. On day 8, multiplex renal miRNA profiling was performed. Expression of nine miRNAs was determined with qPCR at all time points. Based on the target prediction, plexin-A2 and Cd2AP were measured by Western blot. (3) Results: On day 8 after IR, the expression of 20/1195 miRNAs doubled, and 9/13 selected miRNAs were upregulated at all time points. Nx reduced the expression of several ischemia-induced pro-fibrotic miRNAs (fibromirs), such as miR-142a-duplex, miR-146a-5p, miR-199a-duplex, miR-214-3p and miR-223-3p, in the injured kidneys at various time points. Plexin-A2 was upregulated by IR on day 10, while Cd2AP was unchanged. (4) Conclusion: Nx delayed fibrosis progression and decreased the expression of ischemia-induced fibromirs. The protein expression of plexin-A2 and Cd2AP is mainly regulated by factors other than miRNAs.
Collapse
Affiliation(s)
- Beáta Róka
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (É.N.B.); (G.S.)
| | - Pál Tod
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (É.N.B.); (G.S.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Tamás Kaucsár
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (É.N.B.); (G.S.)
| | - Éva Nóra Bukosza
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (É.N.B.); (G.S.)
| | - Imre Vörös
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (I.V.); (Z.V.V.); (B.P.); (B.Á.); (P.F.)
- HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, 1089 Budapest, Hungary
| | - Zoltán V. Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (I.V.); (Z.V.V.); (B.P.); (B.Á.); (P.F.)
- HCEMM-SU Cardiometabolic Immunology Research Group, Semmelweis University, 1089 Budapest, Hungary
| | - Balázs Petrovich
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (I.V.); (Z.V.V.); (B.P.); (B.Á.); (P.F.)
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (I.V.); (Z.V.V.); (B.P.); (B.Á.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089 Budapest, Hungary; (I.V.); (Z.V.V.); (B.P.); (B.Á.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Gábor Szénási
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (É.N.B.); (G.S.)
| | - Péter Hamar
- Institute of Translational Medicine, Semmelweis University, 1094 Budapest, Hungary; (B.R.); (P.T.); (T.K.); (É.N.B.); (G.S.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
- Correspondence: ; Tel.: +36-20-825-9751
| |
Collapse
|
10
|
Liang Y, Wang M, Wang C, Liu Y, Naruse K, Takahashi K. The Mechanisms of the Development of Atherosclerosis in Prediabetes. Int J Mol Sci 2021; 22:ijms22084108. [PMID: 33921168 PMCID: PMC8071517 DOI: 10.3390/ijms22084108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/15/2022] Open
Abstract
Lifestyle changes, such as overeating and underexercising, can increase the risk of prediabetes. Diabetes is one of the leading causes of atherosclerosis, and recently it became clear that the pathophysiology of atherosclerosis progresses even before the onset of diabetic symptoms. In addition to changes in platelets and leukocytes in the hyperglycemic state and damage to vascular endothelial cells, extracellular vesicles and microRNAs were found to be involved in the progression of prediabetes atherosclerosis. This review discusses the cellular and molecular mechanisms of these processes, with an intention to enable a comprehensive understanding of the pathophysiology of prediabetes and atherosclerosis.
Collapse
|