1
|
Yang K, Zhao Y, Hu J, Gao R, Shi J, Wei X, Chen J, Hu K, Sun A, Ge J. ALKBH5 induces fibroblast-to-myofibroblast transformation during hypoxia to protect against cardiac rupture after myocardial infarction. J Adv Res 2024; 61:193-209. [PMID: 37689242 PMCID: PMC11258655 DOI: 10.1016/j.jare.2023.09.004] [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: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023] Open
Abstract
INTRODUCTION N6-methyladenosine (m6A) methylation produces a marked effect on cardiovascular diseases. The m6A demethylase AlkB homolog 5 (ALKBH5), as an m6A "eraser", is responsible for decreased m6A modification. However, its role in cardiac fibroblasts during the post-myocardial infarction (MI) healing process remains elusive. OBJECTIVES To investigate the effect of ALKBH5 in cardiac fibroblasts during infarct repair. METHODS MI was mimicked by permanent left anterior descending artery ligation in global ALKBH5-knockout, ALKBH5-knockin, and fibroblast-specific ALKBH5-knockout mice to study the function of ALKBH5 during post-MI collagen repair. Methylated RNA immunoprecipitation sequencing was performed to explore potential ALKBH5 targets. RESULTS Dramatic alterations in ALKBH5 expression were observed during the early stages post-MI and in hypoxic fibroblasts. Global ALKBH5 knockin reduced infarct size and ameliorated cardiac function after MI. The global and fibroblast-specific ALKBH5-knockout mice both exhibited low survival rates along with poor collagen repair, impaired cardiac function, and cardiac rupture. Both in vivo and in vitro ALKBH5 loss resulted in impaired fibroblast activation and decreased collagen deposition. Additionally, hypoxia, but not TGF-β1 or Ang II, upregulated ALKBH5 expression in myofibroblasts by HIF-1α-dependent transcriptional regulation. Mechanistically, ALKBH5 promoted the stability of ErbB4 mRNA and the degradation of ST14 mRNA via m6A demethylation. Fibroblast-specific ErbB4 overexpression ameliorated the impaired fibroblast-to-myofibroblast transformation and poor post-MI repair due to ALKBH5 knockout. CONCLUSION Fibroblast ALKBH5 positively regulates post-MI healing by stabilization of ErbB4 mRNA in an m6A-dependent manner. ALKBH5/ErbB4 might be potential therapeutic targets for post-MI cardiac rupture.
Collapse
Affiliation(s)
- Kun Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China
| | - Yongchao Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Department of Cardiology, Affiliated Hospital of Zunyi Medical University, Guizhou Province, China
| | - Jingjing Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, China
| | - Rifeng Gao
- The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jiaran Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang Province, China
| | - Xiang Wei
- The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Juntao Chen
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China; Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| |
Collapse
|
2
|
Benak D, Kolar F, Hlavackova M. Epitranscriptomic Regulations in the Heart. Physiol Res 2024; 73:S185-S198. [PMID: 38634649 PMCID: PMC11412340 DOI: 10.33549/physiolres.935265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
RNA modifications affect key stages of the RNA life cycle, including splicing, export, decay, and translation. Epitranscriptomic regulations therefore significantly influence cellular physiology and pathophysiology. Here, we selected some of the most abundant modifications and reviewed their roles in the heart and in cardiovascular diseases: N6-methyladenosine (m6A), N6,2'-O-dimethyladenosine (m6Am), N1-methyladenosine (m1A), pseudouridine (?), 5 methylcytidine (m5C), and inosine (I). Dysregulation of epitranscriptomic machinery affecting these modifications vastly changes the cardiac phenotype and is linked with many cardiovascular diseases such as myocardial infarction, cardiomyopathies, or heart failure. Thus, a deeper understanding of these epitranscriptomic changes and their regulatory mechanisms can enhance our knowledge of the molecular underpinnings of prevalent cardiac diseases, potentially paving the way for novel therapeutic strategies. Keywords: Epitranscriptomics, RNA modifications, Epigenetics, m6A, RNA, Heart.
Collapse
Affiliation(s)
- D Benak
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic.
| | | | | |
Collapse
|
3
|
Ma B, Xiu L, Ding L. The m6 RNA methylation regulator KIAA1429 is associated with autophagy-mediated drug resistance in lung cancer. FASEB Bioadv 2024; 6:105-117. [PMID: 38585432 PMCID: PMC10995705 DOI: 10.1096/fba.2023-00083] [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: 08/17/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 04/09/2024] Open
Abstract
N6-methyladenosine (m6A) modification plays a crucial role in cancer progression. However, the role of m6A modification-mediated autophagy underlying non-small cell lung cancer (NSCLC) gefitinib resistance remains unknown. Here, we discovered that m6A methyltransferase KIAA1429 was highly expressed in NSCLC gefitinib-resistant cells (PC9-GR) as well as tissues, and KIAA1429 high expression was associated with poor survival. In addition, silent KIAA1429 repressed gefitinib resistance in NSCLC and reduced tumor growth in vivo. Mechanistically, KIAA1429 stabilized WTAP, a significant player in autophagy, by binding to the 3' untranslated regions (3'-UTR) of WTAP. In a word, our findings indicated that KIAA1429 could elevate NSCLC gefitinib resistance, which may provide a promising targeted therapy for NSCLC patients.
Collapse
Affiliation(s)
- Bo Ma
- Department of General Thoracic SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuanChina
| | - Lei Xiu
- Department of Thoracic and Cardiac SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuanChina
| | - Lili Ding
- Department of Obstetrics and Gynecology ExaminationGeneral Hospital of Ningxia Medical UniversityYinchuanChina
| |
Collapse
|
4
|
Benak D, Holzerova K, Hrdlicka J, Kolar F, Olsen M, Karelson M, Hlavackova M. Epitranscriptomic regulation in fasting hearts: implications for cardiac health. RNA Biol 2024; 21:1-14. [PMID: 38326277 PMCID: PMC10854364 DOI: 10.1080/15476286.2024.2307732] [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] [Accepted: 01/16/2024] [Indexed: 02/09/2024] Open
Abstract
Cardiac tolerance to ischaemia can be increased by dietary interventions such as fasting, which is associated with significant changes in myocardial gene expression. Among the possible mechanisms of how gene expression may be altered are epigenetic modifications of RNA - epitranscriptomics. N6-methyladenosine (m6A) and N6,2'-O-dimethyladenosine (m6Am) are two of the most prevalent modifications in mRNA. These methylations are reversible and regulated by proteins called writers, erasers, readers, and m6A-repelled proteins. We analysed 33 of these epitranscriptomic regulators in rat hearts after cardioprotective 3-day fasting using RT-qPCR, Western blot, and targeted proteomic analysis. We found that the most of these regulators were changed on mRNA or protein levels in fasting hearts, including up-regulation of both demethylases - FTO and ALKBH5. In accordance, decreased methylation (m6A+m6Am) levels were detected in cardiac total RNA after fasting. We also identified altered methylation levels in Nox4 and Hdac1 transcripts, both of which play a role in the cytoprotective action of ketone bodies produced during fasting. Furthermore, we investigated the impact of inhibiting demethylases ALKBH5 and FTO in adult rat primary cardiomyocytes (AVCMs). Our findings indicate that inhibiting these demethylases reduced the hypoxic tolerance of AVCMs isolated from fasting rats. This study showed that the complex epitranscriptomic machinery around m6A and m6Am modifications is regulated in the fasting hearts and might play an important role in cardiac adaptation to fasting, a well-known cardioprotective intervention.
Collapse
Affiliation(s)
- Daniel Benak
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Kristyna Holzerova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jaroslav Hrdlicka
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Frantisek Kolar
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Mark Olsen
- Department of Pharmaceutical Sciences, College of Pharmacy-Glendale, Midwestern University, Glendale, Arizona, USA
| | - Mati Karelson
- Institute of Chemistry, University of Tartu, Tartu, Estonia
| | - Marketa Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
5
|
Benak D, Kolar F, Zhang L, Devaux Y, Hlavackova M. RNA modification m 6Am: the role in cardiac biology. Epigenetics 2023; 18:2218771. [PMID: 37331009 DOI: 10.1080/15592294.2023.2218771] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/20/2023] Open
Abstract
Epitranscriptomic modifications have recently emerged into the spotlight of researchers due to their vast regulatory effects on gene expression and thereby cellular physiology and pathophysiology. N6,2'-O-dimethyladenosine (m6Am) is one of the most prevalent chemical marks on RNA and is dynamically regulated by writers (PCIF1, METTL4) and erasers (FTO). The presence or absence of m6Am in RNA affects mRNA stability, regulates transcription, and modulates pre-mRNA splicing. Nevertheless, its functions in the heart are poorly known. This review summarizes the current knowledge and gaps about m6Am modification and its regulators in cardiac biology. It also points out technical challenges and lists the currently available techniques to measure m6Am. A better understanding of epitranscriptomic modifications is needed to improve our knowledge of the molecular regulations in the heart which may lead to novel cardioprotective strategies.
Collapse
Affiliation(s)
- Daniel Benak
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
- Department of Physiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Frantisek Kolar
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lu Zhang
- Bioinformatics Platform, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Marketa Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
6
|
Komal S, Gohar A, Althobaiti S, Ahmad Khan I, Cui LG, Zhang LR, Han SN, Shakeel M. ALKBH5 inhibitors as a potential treatment strategy in heart failure-inferences from gene expression profiling. Front Cardiovasc Med 2023; 10:1194311. [PMID: 37583580 PMCID: PMC10425272 DOI: 10.3389/fcvm.2023.1194311] [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: 03/28/2023] [Accepted: 07/18/2023] [Indexed: 08/17/2023] Open
Abstract
Heart Failure (HF) is a complex clinical syndrome in which the heart is unable to provide enough blood flow to meet metabolic needs and lacks efficient venous return. HF is a major risk factor for morbidity and mortality with cardiovascular diseases globally. Despite enormous research, the molecular markers relevant to disease prognosis and management remain not well understood. Here, we analyzed the whole transcriptomes of 18 failing hearts and 15 non-failing hearts (predominantly of Caucasian origin), by applying the standard in silico tools. The analyses revealed novel gene-markers including ALKBH5 of mRNA demethylation and KMT2E of histone modification processes, significantly over-expressed in the HF compared with the non-failing hearts (FDR < 0.05). To validate the over-expression of ALKBH5, we determined the global m6A level in hypoxic H9c2 cells using a dot blot assay. The global m6A level was found markedly lower in the hypoxic H9c2 cells than in the control cells. Additionally, the expression of ALKBH5 in the H9c2 cells was quantified by the qPCR and found to be 1.18 times higher at 12 h (p < 0.05), and 1.67 times higher at 24 h of hypoxia (p < 0.01) compared with the control cells, indicating a likely role of ALKBH5 in the failing cardiac cells. Furthermore, we identified several compounds through the virtual screening of 11,272 drug-like molecules of the ZINC15 database to inhibit the ALKBH5 in a molecular docking process. Collectively, the study revealed novel markers potentially involved in the pathophysiology of HF and suggested plausible therapeutic molecules for the management of the disease.
Collapse
Affiliation(s)
- Sumra Komal
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Atia Gohar
- Dow Institute for Advanced Biological and Animal Research, Dow University of Health Sciences, Karachi, Pakistan
| | | | - Ishtiaq Ahmad Khan
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Liu-Gen Cui
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Sheng-Na Han
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Muhammad Shakeel
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
7
|
Benak D, Benakova S, Plecita-Hlavata L, Hlavackova M. The role of m 6A and m 6Am RNA modifications in the pathogenesis of diabetes mellitus. Front Endocrinol (Lausanne) 2023; 14:1223583. [PMID: 37484960 PMCID: PMC10360938 DOI: 10.3389/fendo.2023.1223583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 06/26/2023] [Indexed: 07/25/2023] Open
Abstract
The rapidly developing research field of epitranscriptomics has recently emerged into the spotlight of researchers due to its vast regulatory effects on gene expression and thereby cellular physiology and pathophysiology. N6-methyladenosine (m6A) and N6,2'-O-dimethyladenosine (m6Am) are among the most prevalent and well-characterized modified nucleosides in eukaryotic RNA. Both of these modifications are dynamically regulated by a complex set of epitranscriptomic regulators called writers, readers, and erasers. Altered levels of m6A and also several regulatory proteins were already associated with diabetic tissues. This review summarizes the current knowledge and gaps about m6A and m6Am modifications and their respective regulators in the pathophysiology of diabetes mellitus. It focuses mainly on the more prevalent type 2 diabetes mellitus (T2DM) and its treatment by metformin, the first-line antidiabetic agent. A better understanding of epitranscriptomic modifications in this highly prevalent disease deserves further investigation and might reveal clinically relevant discoveries in the future.
Collapse
Affiliation(s)
- Daniel Benak
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Stepanka Benakova
- Laboratory of Pancreatic Islet Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- First Faculty of Medicine, Charles University, Prague, Czechia
| | - Lydie Plecita-Hlavata
- Laboratory of Pancreatic Islet Research, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Marketa Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
8
|
Liu S, Wang T, Cheng Z, Liu J. N6-methyladenosine (m6A) RNA modification in the pathophysiology of heart failure: a narrative review. Cardiovasc Diagn Ther 2022; 12:908-925. [PMID: 36605077 PMCID: PMC9808110 DOI: 10.21037/cdt-22-277] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022]
Abstract
Background and Objective Heart failure is the end-stage of various cardiovascular diseases. Recent progress in molecular biology has facilitated the understanding of the mechanisms of heart failure development at the molecular level. N6-adenosine methylation (m6A) is a post-transcriptional modification of RNA. Recent research work reported that m6A regulates gene expression and subsequently affects the activation of cell signaling pathways related to heart failure. Moreover, m6A regulators like methyltransferase-like 3 (METTL3) were reported to participate in myocardium hypertrophy. However, the current research work related to the role of m6A participating in the occurrence of heart failure is rare in some aspects like immune cell infiltration and diabetic heart diseases. Thus, it is reasonable to review the current achievements and provide further study orientation. Methods We searched related literature using the keywords: m6A AND heart failure in PubMed, Web of Science and Medline. The language was confined to English. The published year of searched literature ranged from 2012 to 2022. The searched results were put into Endnote software for management. Two authors investigated the searching terms and reviewed the full text of selected terms. Key Content and Findings m6A and its regulators are involved in the metabolism of various types of RNAs. m6A modification can regulate various types of cell signaling pathways related to the heart failure via interaction with m6A regulators. m6A and its regulators broadly participate in the myocardium fibrosis, myocardium hypertrophy, myocardial cell apoptosis, and ischemic reperfusion injury. Specifically, m6A participates in the cell apoptosis via regulation of autophagy flux. However, the current research work does not have enough evidence to prove that m6A regulator played its specific effect on the target transcript via regulating the m6A level. Conclusions m6A and its regulators participates in the progression of heart failure via modifying the RNA level. Future investigation of m6A should focus on the interaction between the m6A regulators and targeted transcript. Besides, the regulation role of m6A in immune cell infiltration and diabetic heart diseases should also be focused.
Collapse
Affiliation(s)
- Sihan Liu
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Tongyu Wang
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zeyi Cheng
- Department of Cardiac Surgery, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Liu
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
9
|
Targeting Epigenetic Regulation of Cardiomyocytes through Development for Therapeutic Cardiac Regeneration after Heart Failure. Int J Mol Sci 2022; 23:ijms231911878. [PMID: 36233177 PMCID: PMC9569953 DOI: 10.3390/ijms231911878] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death globally, with no cure currently. Therefore, there is a dire need to further understand the mechanisms that arise during heart failure. Notoriously, the adult mammalian heart has a very limited ability to regenerate its functional cardiac cells, cardiomyocytes, after injury. However, the neonatal mammalian heart has a window of regeneration that allows for the repair and renewal of cardiomyocytes after injury. This specific timeline has been of interest in the field of cardiovascular and regenerative biology as a potential target for adult cardiomyocyte repair. Recently, many of the neonatal cardiomyocyte regeneration mechanisms have been associated with epigenetic regulation within the heart. This review summarizes the current and most promising epigenetic mechanisms in neonatal cardiomyocyte regeneration, with a specific emphasis on the potential for targeting these mechanisms in adult cardiac models for repair after injury.
Collapse
|
10
|
Geng X, Li Z, Yang Y. Emerging Role of Epitranscriptomics in Diabetes Mellitus and Its Complications. Front Endocrinol (Lausanne) 2022; 13:907060. [PMID: 35692393 PMCID: PMC9184717 DOI: 10.3389/fendo.2022.907060] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/14/2022] [Indexed: 01/13/2023] Open
Abstract
Diabetes mellitus (DM) and its related complications are among the leading causes of disability and mortality worldwide. Substantial studies have explored epigenetic regulation that is involved in the modifications of DNA and proteins, but RNA modifications in diabetes are still poorly investigated. In recent years, posttranscriptional epigenetic modification of RNA (the so-called 'epitranscriptome') has emerged as an interesting field of research. Numerous modifications, mainly N6 -methyladenosine (m6A), have been identified in nearly all types of RNAs and have been demonstrated to have an indispensable effect in a variety of human diseases, such as cancer, obesity, and diabetes. Therefore, it is particularly important to understand the molecular basis of RNA modifications, which might provide a new perspective for the pathogenesis of diabetes mellitus and the discovery of new therapeutic targets. In this review, we aim to summarize the recent progress in the epitranscriptomics involved in diabetes and diabetes-related complications. We hope to provide some insights for enriching the understanding of the epitranscriptomic regulatory mechanisms of this disease as well as the development of novel therapeutic targets for future clinical benefit.
Collapse
Affiliation(s)
- Xinqian Geng
- Department of Endocrinology, The Affiliated Hospital of Yunnan University and the Second People’s Hospital of Yunnan Province, Kunming, China
| | - Zheng Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ying Yang
- Department of Endocrinology, The Affiliated Hospital of Yunnan University and the Second People’s Hospital of Yunnan Province, Kunming, China
| |
Collapse
|
11
|
Nilsson EE, Ben Maamar M, Skinner MK. Role of epigenetic transgenerational inheritance in generational toxicology. ENVIRONMENTAL EPIGENETICS 2022; 8:dvac001. [PMID: 35186326 PMCID: PMC8848501 DOI: 10.1093/eep/dvac001] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/04/2021] [Accepted: 02/03/2022] [Indexed: 05/27/2023]
Abstract
Many environmental toxicants have been shown to be associated with the transgenerational inheritance of increased disease susceptibility. This review describes the generational toxicity of some of these chemicals and their role in the induction of epigenetic transgenerational inheritance of disease. Epigenetic factors include DNA methylation, histone modifications, retention of histones in sperm, changes to chromatin structure, and expression of non-coding RNAs. For toxicant-induced epigenetic transgenerational inheritance to occur, exposure to a toxicant must result in epigenetic changes to germ cells (sperm or eggs) since it is the germ cells that carry molecular information to subsequent generations. In addition, the epigenetic changes induced in transgenerational generation animals must cause alterations in gene expression in these animals' somatic cells. In some cases of generational toxicology, negligible changes are seen in the directly exposed generations, but increased disease rates are seen in transgenerational descendants. Governmental policies regulating toxicant exposure should take generational effects into account. A new approach that takes into consideration generational toxicity will be needed to protect our future populations.
Collapse
Affiliation(s)
- Eric E Nilsson
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA 99164-4236, USA
| | - Millissia Ben Maamar
- Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA 99164-4236, USA
| | - Michael K Skinner
- **Correspondence address. Center for Reproductive Biology, School of Biological Sciences, Washington State University, Pullman, WA 99164-4236, USA. Tel: +509-335-1524; E-mail:
| |
Collapse
|
12
|
Leptidis S, Papakonstantinou E, Diakou KI, Pierouli K, Mitsis T, Dragoumani K, Bacopoulou F, Sanoudou D, Chrousos GP, Vlachakis D. Epitranscriptomics of cardiovascular diseases (Review). Int J Mol Med 2022; 49:9. [PMID: 34791505 PMCID: PMC8651226 DOI: 10.3892/ijmm.2021.5064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/20/2021] [Indexed: 11/09/2022] Open
Abstract
RNA modifications have recently become the focus of attention due to their extensive regulatory effects in a vast array of cellular networks and signaling pathways. Just as epigenetics is responsible for the imprinting of environmental conditions on a genetic level, epitranscriptomics follows the same principle at the RNA level, but in a more dynamic and sensitive manner. Nevertheless, its impact in the field of cardiovascular disease (CVD) remains largely unexplored. CVD and its associated pathologies remain the leading cause of death in Western populations due to the limited regenerative capacity of the heart. As such, maintenance of cardiac homeostasis is paramount for its physiological function and its capacity to respond to environmental stimuli. In this context, epitranscriptomic modifications offer a novel and promising therapeutic avenue, based on the fine‑tuning of regulatory cascades, necessary for cardiac function. This review aimed to provide an overview of the most recent findings of key epitranscriptomic modifications in both coding and non‑coding RNAs. Additionally, the methods used for their detection and important associations with genetic variations in the context of CVD were summarized. Current knowledge on cardiac epitranscriptomics, albeit limited still, indicates that the impact of epitranscriptomic editing in the heart, in both physiological and pathological conditions, holds untapped potential for the development of novel targeted therapeutic approaches in a dynamic manner.
Collapse
Affiliation(s)
- Stefanos Leptidis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Eleni Papakonstantinou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Kalliopi Io Diakou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Katerina Pierouli
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Thanasis Mitsis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Konstantina Dragoumani
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
| | - Flora Bacopoulou
- Laboratory of Molecular Endocrinology, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- First Department of Pediatrics, Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, Medical School, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Despina Sanoudou
- Fourth Department of Internal Medicine, Clinical Genomics and Pharmacogenomics Unit, Medical School, 'Attikon' Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Molecular Biology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P. Chrousos
- Laboratory of Molecular Endocrinology, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- First Department of Pediatrics, Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, Medical School, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, 11855 Athens, Greece
- Laboratory of Molecular Endocrinology, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
- First Department of Pediatrics, Center for Adolescent Medicine and UNESCO Chair on Adolescent Health Care, Medical School, Aghia Sophia Children's Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- School of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, London WC2R 2LS, UK
| |
Collapse
|
13
|
Deng W, Jin Q, Li L. Protective mechanism of demethylase fat mass and obesity-associated protein in energy metabolism disorder of hypoxia-reoxygenation-induced cardiomyocytes. Exp Physiol 2021; 106:2423-2433. [PMID: 34713923 DOI: 10.1113/ep089901] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/26/2021] [Indexed: 01/02/2023]
Abstract
NEW FINDINGS What is the central question of this study? What is the effect of fat mass and obesity-associated protein (FTO) on energy metabolism in hypoxia-reoxygenation (H/R)-induced cardiomyocytes? What is the main finding and its importance? FTO modification of N6 -methyladenosine (m6 A) is associated with myocardial cell energy metabolism disorder. FTO reduced the m6 A level of sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a) mRNA through demethylation, thus promoting SERCA2a expression, maintaining calcium homeostasis, and improving energy metabolism of H/R cardiomyocytes. ABSTRACT Energy metabolism disorder is the initial physiological link of myocardial ischaemia-reperfusion injury. Fat mass and obesity-associated protein (FTO) is an N6 -methyladenosine (m6 A) demethylase implicated in several cardiac defects. This study sought to investigate the effect of FTO on energy metabolism in hypoxia-reoxygenation (H/R)-induced cardiomyocytes. FTO and sarcoplasmic/endoplasmic reticulum calcium ATPase 2a (SERCA2a) expression in H/R-induced cardiomyocytes were determined. Cardiomyocyte viability, cytotoxicity and apoptosis were measured. The total RNA and polyA+ RNA contents were isolated from cells. The m6 A level of RNA and the enrichment of m6 A of SERCA2a mRNA were calculated. Several indices such as the glycolytic potential, reactive oxygen species (ROS), mitochondrial activity and ATP content were evaluated. The concentration of calcium in cardiomyocytes was determined. FTO and SERCA2a were poorly expressed in H/R-induced cardiomyocytes. There was an elevated m6 A level in total RNA and enrichment of m6 A in SERCA2a mRNA. H/R treatment reduced the cell viability, mitochondrial membrane potential and ATP content in cardiomyocytes, but increased the cytotoxicity, apoptosis, ROS content and calcium concentration. Upregulation of FTO reversed the preceding findings with downregulation of the m6 A level of SERCA2a mRNA. Downregulation of SERCA2a annulled the promoting effect of FTO on calcium homeostasis and energy metabolism in H/R-induced cardiomyocytes. Collectively, the current study demonstrated that FTO reduced the m6 A level on SERCA2a mRNA through demethylation, thus promoting SERCA2a expression, maintaining calcium homeostasis and improving the energy metabolism of H/R cardiomyocytes.
Collapse
Affiliation(s)
- Wenzheng Deng
- Department of Cardiology, Chenzhou First People's Hospital, Chenzhou, Hunan, China
| | - Qiao Jin
- Department of Cardiovascular Medicine, Nanhua University affiliated Changsha Central Hospital, Changsha, Hunan, China
| | - Liang Li
- Department of Cardiovascular Medicine, Nanhua University affiliated Changsha Central Hospital, Changsha, Hunan, China
| |
Collapse
|
14
|
Sikorski V, Karjalainen P, Blokhina D, Oksaharju K, Khan J, Katayama S, Rajala H, Suihko S, Tuohinen S, Teittinen K, Nummi A, Nykänen A, Eskin A, Stark C, Biancari F, Kiss J, Simpanen J, Ropponen J, Lemström K, Savinainen K, Lalowski M, Kaarne M, Jormalainen M, Elomaa O, Koivisto P, Raivio P, Bäckström P, Dahlbacka S, Syrjälä S, Vainikka T, Vähäsilta T, Tuncbag N, Karelson M, Mervaala E, Juvonen T, Laine M, Laurikka J, Vento A, Kankuri E. Epitranscriptomics of Ischemic Heart Disease-The IHD-EPITRAN Study Design and Objectives. Int J Mol Sci 2021; 22:6630. [PMID: 34205699 PMCID: PMC8235045 DOI: 10.3390/ijms22126630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 12/11/2022] Open
Abstract
Epitranscriptomic modifications in RNA can dramatically alter the way our genetic code is deciphered. Cells utilize these modifications not only to maintain physiological processes, but also to respond to extracellular cues and various stressors. Most often, adenosine residues in RNA are targeted, and result in modifications including methylation and deamination. Such modified residues as N-6-methyl-adenosine (m6A) and inosine, respectively, have been associated with cardiovascular diseases, and contribute to disease pathologies. The Ischemic Heart Disease Epitranscriptomics and Biomarkers (IHD-EPITRAN) study aims to provide a more comprehensive understanding to their nature and role in cardiovascular pathology. The study hypothesis is that pathological features of IHD are mirrored in the blood epitranscriptome. The IHD-EPITRAN study focuses on m6A and A-to-I modifications of RNA. Patients are recruited from four cohorts: (I) patients with IHD and myocardial infarction undergoing urgent revascularization; (II) patients with stable IHD undergoing coronary artery bypass grafting; (III) controls without coronary obstructions undergoing valve replacement due to aortic stenosis and (IV) controls with healthy coronaries verified by computed tomography. The abundance and distribution of m6A and A-to-I modifications in blood RNA are charted by quantitative and qualitative methods. Selected other modified nucleosides as well as IHD candidate protein and metabolic biomarkers are measured for reference. The results of the IHD-EPITRAN study can be expected to enable identification of epitranscriptomic IHD biomarker candidates and potential drug targets.
Collapse
Affiliation(s)
- Vilbert Sikorski
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (V.S.); (D.B.); (E.M.)
| | - Pasi Karjalainen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Daria Blokhina
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (V.S.); (D.B.); (E.M.)
| | - Kati Oksaharju
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Jahangir Khan
- Tampere Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland; (J.K.); (J.L.)
| | | | - Helena Rajala
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Satu Suihko
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Suvi Tuohinen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Kari Teittinen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Annu Nummi
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Antti Nykänen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Arda Eskin
- Graduate School of Informatics, Department of Health Informatics, Middle East Technical University, 06800 Ankara, Turkey;
| | - Christoffer Stark
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Fausto Biancari
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
- Heart Center, Turku University Hospital and Department of Surgery, University of Turku, 20521 Turku, Finland
- Research Unit of Surgery, Anesthesiology and Critical Care, University of Oulu, 90014 Oulu, Finland
| | - Jan Kiss
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Jarmo Simpanen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Jussi Ropponen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Karl Lemström
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Kimmo Savinainen
- Clinical Biobank Tampere, Tampere University Hospital, 33520 Tampere, Finland;
| | - Maciej Lalowski
- Helsinki Institute of Life Science (HiLIFE), Meilahti Clinical Proteomics Core Facility, Department of Biochemistry and Developmental Biology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland;
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Department of Biomedical Proteomics, 61-704 Poznan, Poland
| | - Markku Kaarne
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Mikko Jormalainen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Outi Elomaa
- Folkhälsan Research Center, 00250 Helsinki, Finland; (S.K.); (O.E.)
| | - Pertti Koivisto
- Chemistry Unit, Finnish Food Authority, 00790 Helsinki, Finland;
| | - Peter Raivio
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Pia Bäckström
- Helsinki Biobank, Hospital District of Helsinki and Uusimaa, 00029 Helsinki, Finland;
| | - Sebastian Dahlbacka
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Simo Syrjälä
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Tiina Vainikka
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Tommi Vähäsilta
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Nurcan Tuncbag
- Department of Chemical and Biological Engineering, College of Engineering, Koç University, 34450 Istanbul, Turkey;
- School of Medicine, Koç University, 34450 Istanbul, Turkey
| | - Mati Karelson
- Institute of Chemistry, University of Tartu, 50411 Tartu, Estonia;
| | - Eero Mervaala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (V.S.); (D.B.); (E.M.)
| | - Tatu Juvonen
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
- Research Unit of Surgery, Anesthesiology and Critical Care, University of Oulu, 90014 Oulu, Finland
| | - Mika Laine
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Jari Laurikka
- Tampere Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland; (J.K.); (J.L.)
| | - Antti Vento
- Heart and Lung Center, Helsinki University Hospital, 00029 Helsinki, Finland; (P.K.); (K.O.); (H.R.); (S.S.); (S.T.); (K.T.); (A.N.); (A.N.); (C.S.); (F.B.); (J.K.); (J.S.); (J.R.); (K.L.); (M.K.); (M.J.); (P.R.); (S.D.); (S.S.); (T.V.); (T.V.); (T.J.); (M.L.); (A.V.)
| | - Esko Kankuri
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (V.S.); (D.B.); (E.M.)
| |
Collapse
|
15
|
Tang J, Han T, Tong W, Zhao J, Wang W. N 6-methyladenosine (m 6A) methyltransferase KIAA1429 accelerates the gefitinib resistance of non-small-cell lung cancer. Cell Death Discov 2021; 7:108. [PMID: 34001850 PMCID: PMC8128911 DOI: 10.1038/s41420-021-00488-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/19/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
N6-methyladenosine (m6A) modification has been convincingly identified to be a critical regulator in human cancer. However, the contribution of m6A to NSCLC gefitinib resistance is still largely unknown. Here, we screened and identified that m6A methyltransferase KIAA1429 was highly expressed in gefitinib-resistant NSCLC cells (PC9-GR), tissues, and closely related to unfavorable survival. Functionally, KIAA1429 accelerated the gefitinib resistance of NSCLC in vitro. Depletion of KIAA1429 repressed the tumor growth of PC9-GR cells in vivo. Mechanistically, KIAA1429 enhanced the mRNA stability of HOXA1 through targeting its 3'-untranslated regions (3'-UTR). Overall, our findings indicate that KIAA1429 plays essential oncogenic roles in NSCLC gefitinib resistance, which may provide a feasible therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Jun Tang
- No. 1 Department of Tuberculosis, Chest Hospital, Shenyang, Liaoning Province, 110044, China
| | - Tianci Han
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, 110042, China
- Department of Thoracic Surgery, Liaoning Cancer Hospital, Shenyang, Liaoning Province, 110042, China
| | - Wei Tong
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, 110042, China
- Department of Thoracic Surgery, Liaoning Cancer Hospital, Shenyang, Liaoning Province, 110042, China
| | - Jian Zhao
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, 110042, China
- Department of Thoracic Surgery, Liaoning Cancer Hospital, Shenyang, Liaoning Province, 110042, China
| | - Wei Wang
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, 110042, China.
- Department of Thoracic Surgery, Liaoning Cancer Hospital, Shenyang, Liaoning Province, 110042, China.
| |
Collapse
|