1
|
Abubakar M, Irfan U, Abdelkhalek A, Javed I, Khokhar MI, Shakil F, Raza S, Salim SS, Altaf MM, Habib R, Ahmed S, Ahmed F. Comprehensive Quality Analysis of Conventional and Novel Biomarkers in Diagnosing and Predicting Prognosis of Coronary Artery Disease, Acute Coronary Syndrome, and Heart Failure, a Comprehensive Literature Review. J Cardiovasc Transl Res 2024:10.1007/s12265-024-10540-8. [PMID: 38995611 DOI: 10.1007/s12265-024-10540-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
Coronary artery disease (CAD), acute coronary syndrome (ACS), and heart failure (HF) are major global health issues with high morbidity and mortality rates. Biomarkers like cardiac troponins (cTn) and natriuretic peptides (NPs) are crucial tools in cardiology, but numerous new biomarkers have emerged, proving increasingly valuable in CAD/ACS. These biomarkers are classified based on their mechanisms, such as fibrosis, metabolism, inflammation, and congestion. The integration of established and emerging biomarkers into clinical practice is an ongoing process, and recognizing their strengths and limitations is crucial for their accurate interpretation, incorporation into clinical settings, and improved management of CVD patients. We explored established biomarkers like cTn, NPs, and CRP, alongside newer biomarkers such as Apo-A1, IL-17E, IgA, Gal-3, sST2, GDF-15, MPO, H-FABP, Lp-PLA2, and ncRNAs; provided evidence of their utility in CAD/ACS diagnosis and prognosis; and empowered clinicians to confidently integrate these biomarkers into clinical practice based on solid evidence.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan.
| | - Umema Irfan
- Department of Internal Medicine, Deccan College of Medical Sciences, Hyderabad, India
| | - Ahmad Abdelkhalek
- Department of Internal Medicine, Zhejiang University, Zhejiang, China
| | - Izzah Javed
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan
| | | | - Fraz Shakil
- Department of Emergency Medicine, Mayo Hospital, Lahore, Pakistan
| | - Saud Raza
- Department of Anesthesia, Social Security Teaching Hospital, Lahore, Punjab, Pakistan
| | - Siffat Saima Salim
- Department of Surgery, Holy Family Red Crescent Medical College Hospital, Dhaka, Bangladesh
| | - Muhammad Mahran Altaf
- Department of Internal Medicine, Ameer-Ud-Din Medical College, 6 Birdwood Road, Jinnah Town, Lahore, 54000, Punjab, Pakistan
| | - Rizwan Habib
- Department of Internal Medicine and Emergency, Indus Hospital, Lahore, Pakistan
| | - Simra Ahmed
- Department of Internal Medicine, Ziauddin Medical College, Karachi, Pakistan
| | - Farea Ahmed
- Department of Internal Medicine, Ziauddin Medical College, Karachi, Pakistan
| |
Collapse
|
2
|
Ruan Y, Meng S, Jia R, Cao X, Jin Z. MicroRNA-322-5p protects against myocardial infarction through targeting BTG2. Am J Med Sci 2024; 367:397-405. [PMID: 38437946 DOI: 10.1016/j.amjms.2024.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 01/21/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024]
Abstract
BACKGROUND Numerous studies have explored the therapeutic potential of microRNA (miR) in myocardial infarction (MI) treatment. This study focuses on the role of miR-322-5p in MI, particularly in its regulatory interaction with B-cell translocation gene 2 (BTG2). MATERIALS AND METHODS Expression levels of miR-322-5p and BTG2 were assessed in a rat MI model. Adenovirus altering miR-322-5p or BTG2 expression were administered to MI rats. Evaluation included cardiac function, inflammation, myocardial injury, pathological changes, apoptosis, and NF-κB pathway-related genes in MI rats post-targeted treatment. The miR-322-5p and BTG2 targeting relationship was investigated. RESULTS MI rats exhibited low miR-322-5p and high BTG2 expression in the myocardial tissues. Restoration of miR-322-5p enhanced cardiac function, alleviated inflammation and myocardial injury, mitigated pathological changes and apoptosis, and deactivated the NF-κB pathway in MI rats. BTG2 expression was negatively-regulated by miR-322-5p. Overexpressed BTG2 counteracted miR-322-5p-induced cardioprotection on MI rats. CONCLUSION This study provides evidence that miR-322-5p protects against MI by suppressing BTG2 expression.
Collapse
Affiliation(s)
- Yang Ruan
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shuai Meng
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruofei Jia
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaojing Cao
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zening Jin
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
3
|
Al-Masri A. Apoptosis and long non-coding RNAs: Focus on their roles in Heart diseases. Pathol Res Pract 2023; 251:154889. [PMID: 38238070 DOI: 10.1016/j.prp.2023.154889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 01/23/2024]
Abstract
Heart disease is one of the principal death reasons around the world and there is a growing requirement to discover novel healing targets that have the potential to avert or manage these illnesses. On the other hand, apoptosis is a strongly controlled, cell removal procedure that has a crucial part in numerous cardiac problems, such as reperfusion injury, MI (myocardial infarction), consecutive heart failure, and inflammation of myocardium. Completely comprehending the managing procedures of cell death signaling is critical as it is the primary factor that influences patient mortality and morbidity, owing to cardiomyocyte damage. Indeed, the prevention of heart cell death appears to be a viable treatment approach for heart illnesses. According to current researches, a number of long non-coding RNAs cause the heart cells death via different methods that are embroiled in controlling the activity of transcription elements, the pathways that signals transmission within cells, small miRNAs, and the constancy of proteins. When there is too much cell death in the heart, it can cause problems like reduced blood flow, heart damage after restoring blood flow, heart disease in diabetics, and changes in the heart after reduced blood flow. Therefore, studying how lncRNAs control apoptosis could help us find new treatments for heart diseases. In this review, we present recent discoveries about how lncRNAs are involved in causing cell death in different cardiovascular diseases.
Collapse
Affiliation(s)
- Abeer Al-Masri
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
4
|
Yuan L, Ji HG, Yan XJ, Liu M, Ding YH, Chen XH. Dioscin ameliorates doxorubicin-induced heart failure via inhibiting autophagy and apoptosis by controlling the PDK1-mediated Akt/mTOR signaling pathway. Kaohsiung J Med Sci 2023; 39:1022-1029. [PMID: 37578093 DOI: 10.1002/kjm2.12740] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 08/15/2023] Open
Abstract
Heart failure (HF) is a disease with high mortality and morbidity rate. Autophagy is critically implicated in HF progression. The current research was designed to investigate the function of Dioscin on oxidative stress, autophagy, and apoptosis in HF. In this study, doxorubicin (Dox) was employed to induce HF model and HL-1 cell damage model. Echocardiography implied that Dioscin could dramatically relieve heart function in vivo. Western blotting determined that Dioscin treatment reversed the promotive effect of autophagy caused by Dox through modulating levels of key autophagy-associated molecules, including Atg5 and Beclin1. Dioscin also impaired apoptosis by regulating apoptosis-related protein, including Bcl-2 and cleaved caspase-3 following Dox treatment in vivo and in vitro. Furthermore, the impacts of Dioscin were mediated by upregulation of PDK1-mediated Akt/mTOR signaling. The mTOR inhibitor (rapamycin) could counteract the therapeutic impact of Dioscin in vitro. Taken together, Dioscin could relieve cardiac function through blocking apoptosis and autophagy by activating the PDK1-elicited Akt/mTOR pathway.
Collapse
Affiliation(s)
- Ling Yuan
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
- Department of Cardiology, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Hai-Gang Ji
- Department of Cardiology, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Xiao-Jing Yan
- Changzhou Key Laboratory of Human Use Experience Research & Transformation of Menghe Medical School, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Meng Liu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yu-Han Ding
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Xiao-Hu Chen
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
5
|
Wang K, Wang H, Zhang Q, Liu F. KNOCKDOWN OF CIRC_0001379 ATTENUATES HYPOXIA/REOXYGENATION-INDUCED CARDIOMYOCYTE APOPTOSIS AND INFLAMMATORY RESPONSE BY MIR-98-5P/SOX6 AXIS. Shock 2023; 60:410-418. [PMID: 37493583 DOI: 10.1097/shk.0000000000002178] [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: 07/27/2023]
Abstract
ABSTRACT Background: Aberrant expression of circular RNAs (circRNAs) has been revealed to have crucial roles in the pathological processes of cardiovascular disease. Here, this study aimed to investigate the role and mechanism of circ_0001379 in hypoxia/reoxygenation (H/R)-induced cardiomyocyte injury to explore the potential action of circ_0001379 in acute myocardial infarction (AMI). Methods: Levels of genes and proteins were examined by quantitative real-time polymerase chain reaction and western blot. Cell counting kit-8 assay, 5-ethynyl-2'-deoxyuridine assay, and flow cytometry were used to detect cardiomyocyte proliferation and apoptosis, respectively. The activity of IL-1β, IL-6, and TNF-α was determined by ELISA analysis. The target relationship between miR-98-5p and circ_0001379 or SOX6 (SRY-Box Transcription Factor 6) was verified by dual-luciferase reporter and RNA immunoprecipitation assays. Results: Circ_0001379 was highly expressed in AMI mouse model and H/R-induced cardiomyocytes. Functionally, circ_0001379 silencing attenuated H/R-evoked cardiomyocyte apoptosis and inflammatory response. Mechanistically, circ_0001379 functioned as a sponge for miR-98-5p, which directly targeted SOX6. Moreover, circ_0001379 could regulate SOX6 expression via sponging miR-98-5p. Further rescue experiments showed that inhibition of miR-98-5p reversed the protective effects of circ_0001379 silencing on H/R-induced cardiomyocytes. Besides that, miR-98-5p overexpression abolished H/R-evoked cardiomyocyte apoptosis and inflammatory response, while this condition was abated by SOX6. Conclusion: Circ_0001379 silencing protects cardiomyocytes from H/R-induced apoptosis and inflammatory response by miR-98-5p/SOX6 axis, suggesting a novel therapeutic strategy for AMI prevention.
Collapse
Affiliation(s)
- Kun Wang
- Department of Cardiovascular Medicine, General Hospital of Tisco (Sixth Hospital of Shanxi Medical University), Taiyuan City, China
| | | | | | | |
Collapse
|
6
|
Transcriptomic Analysis of Long Non-Coding RNA during Candida albicans Infection. Genes (Basel) 2023; 14:genes14020251. [PMID: 36833177 PMCID: PMC9956080 DOI: 10.3390/genes14020251] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/07/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Candida albicans is one of the most commonly found species in fungal infections. Due to its clinical importance, molecular aspects of the host immune defense against the fungus are of interest to biomedical sciences. Long non-coding RNAs (lncRNAs) have been investigated in different pathologies and gained widespread attention regarding their role as gene regulators. However, the biological processes in which most lncRNAs perform their function are still unclear. This study investigates the association between lncRNAs with host response to C. albicans using a public RNA-Seq dataset from lung samples of female C57BL/6J wild-type Mus musculus with induced C. albicans infection. The animals were exposed to the fungus for 24 h before sample collection. We selected lncRNAs and protein-coding genes related to the host immune response by combining the results from different computational approaches used for gene selection: differential expression gene analysis, co-expression genes network analysis, and machine learning-based gene selection. Using a guilt by association strategy, we inferred connections between 41 lncRNAs and 25 biological processes. Our results indicated that nine up-regulated lncRNAs were associated with biological processes derived from the response to wounding: 1200007C13Rik, 4833418N02Rik, Gm12840, Gm15832, Gm20186, Gm38037, Gm45774, Gm4610, Mir22hg, and Mirt1. Additionally, 29 lncRNAs were related to genes involved in immune response, while 22 lncRNAs were associated with processes related to reactive species production. These results support the participation of lncRNAs during C. albicans infection, and may contribute to new studies investigating lncRNA functions in the immune response.
Collapse
|
7
|
Li J, Wu X, Ma H, Sun G, Ding P, Lu S, Zhang L, Yang P, Peng Y, Fu J, Wang L. New developments in non-exosomal and exosomal ncRNAs in coronary artery disease. Epigenomics 2022; 14:1355-1372. [PMID: 36514887 DOI: 10.2217/epi-2022-0201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim & methods: Non-exosomal and exosomal ncRNAs have been reported to be involved in the regulation of coronary artery disease (CAD). Therefore, to explore the biological effects of non-exosomal/exosomal ncRNAs in CAD, the authors searched for studies published in the last 3 years on these ncRNAs in CAD and summarized their functions and mechanisms. Results: The authors summarized 120 non-exosomal ncRNAs capable of regulating CAD progression. In clinical studies, 47 non-exosomal and nine exosomal ncRNAs were able to serve as biomarkers for the diagnosis of CAD. Conclusion: Non-exosomal/exosomal ncRNAs are not only able to serve as biomarkers for CAD diagnosis but can also regulate CAD progression through ceRNA mechanisms and are a potential target for early clinical intervention in CAD.
Collapse
Affiliation(s)
- Jingru Li
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Xinyu Wu
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Haocheng Ma
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Guihu Sun
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Peng Ding
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Si Lu
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Lijiao Zhang
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Ping Yang
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Yunzhu Peng
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Jingyun Fu
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| | - Luqiao Wang
- Department of Cardiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, China
| |
Collapse
|
8
|
Xie J, Luo C, Mo B, Lin Y, Liu G, Wang X, Li L. Inflammation and Oxidative Stress Role of S100A12 as a Potential Diagnostic and Therapeutic Biomarker in Acute Myocardial Infarction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2633123. [PMID: 36062187 PMCID: PMC9436632 DOI: 10.1155/2022/2633123] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/20/2022] [Accepted: 08/09/2022] [Indexed: 12/12/2022]
Abstract
Acute myocardial infarction (AMI) is one of the most serious cardiovascular diseases with high morbidity and mortality. Numerous studies have indicated that S100A12 may has an essential role in the occurrence and development of AMI, and in-depth studies are currently lacking. The purpose of this study is to investigate the effect of S100A12 on inflammation and oxidative stress and to determine its clinical applicability in AMI. Here, AMI datasets used to explore the expression pattern of S100A12 in AMI were derived from the Gene Expression Omnibus (GEO) database. The pooled standard average deviation (SMD) was calculated to further determine S100A12 expression. The overlapping differentially expressed genes (DEGs) contained in all included datasets were recognized by the GEO2R tool. Then, functional enrichment analyses, including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses, were carried out to determine the molecular function of overlapping DEGs. Gene set enrichment analysis (GSEA) was conducted to determine unrevealed mechanisms of S100A12. Summary receiver operating characteristic (SROC) curve analysis and receiver operating characteristic (ROC) curve analysis were carried out to identify the diagnostic capabilities of S100A12. Moreover, we screened miRNAs targeting S100A12 using three online databases (miRWalk, TargetScan, and miRDB). In addition, by comprehensively using enzyme-linked immunosorbent assay (ELISA), real-time quantitative PCR (RT-qPCR), Western blotting (WB) methods, etc., we used the AC16 cells to validate the expression and underlying mechanism of S100A12. In our study, five datasets related to AMI, GSE24519, GSE60993, GSE66360, GSE97320, and GSE48060 were included; 412 overlapping DEGs were identified. Protein-protein interaction (PPI) network and functional analyses showed that S100A12 was a pivotal gene related to inflammation and oxidative stress. Then, S100A12 overexpression was identified based on the included datasets. The pooled standard average deviation (SMD) also showed that S100A12 was upregulated in AMI (SMD = 1.36, 95% CI: 0.70-2.03, p = 0.024). The SROC curve analysis result suggested that S100A12 had remarkable diagnostic ability in AMI (AUC = 0.90, 95% CI: 0.87-0.92). And nine miRNAs targeting S100A12 were also identified. Additionally, the overexpression of S100A12 was further confirmed that it maybe promote inflammation and oxidative stress in AMI through comprehensive in vitro experiments. In summary, our study suggests that overexpressed S100A12 may be a latent diagnostic biomarker and therapeutic target of AMI that induces excessive inflammation and oxidative stress. Nine miRNAs targeting S100A12 may play a crucial role in AMI, but further studies are still needed. Our work provides a positive inspiration for the in-depth study of S100A12 in AMI.
Collapse
Affiliation(s)
- Jian Xie
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021 Guangxi, China
| | - Changjun Luo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021 Guangxi, China
| | - Binhai Mo
- Department of Cardiology, The First People Hospital of Nanning & The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, 530016 Guangxi, China
| | - Yunhua Lin
- The First Clinical Medical College, Guangxi Medical University, Nanning 530021, China
| | - Guoqing Liu
- The First Clinical Medical College, Guangxi Medical University, Nanning 530021, China
| | - Xiantao Wang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021 Guangxi, China
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Cardiovascular Institute, Nanning, 530021 Guangxi, China
- Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021 Guangxi, China
| |
Collapse
|
9
|
Dai C, Ma Z, Si J, An G, Zhang W, Li S, Ma Y. Hsa_circ_0007312 Promotes Third-Generation Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor Resistance through Pyroptosis and Apoptosis via the MiR-764/MAPK1 Axis in Lung Adenocarcinoma Cells. J Cancer 2022; 13:2798-2809. [PMID: 35812182 PMCID: PMC9254875 DOI: 10.7150/jca.72066] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/10/2022] [Indexed: 12/24/2022] Open
Abstract
Purposes: Osimertinib is a third-generation epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) used for patients with gefitinib (first-generation EGFR-TKI) resistance, but osimertinib resistance inevitably occurs. Therefore, it is necessary to explore the mechanisms of osimertinib resistance. Materials and Methods: We performed quantitative real-time polymerase chain reaction to detect hsa_circ_0007312 (circ7312), miR-764, and MAPK1 expressions in tissues and cells. Western blotting was used to detect protein levels in cells. Cell Counting Kit-8, apoptotic, and Transwell assays were used to explore biological functions. Luciferase assays were used to identify the interactions between circ7312 and miR-764, MAPK1 and miR-764. A xenograft experiment was performed to clarify the role of circ7312 in vivo. Public datasets were used to identify the relation between circ7312 expression and the cell half maximal inhibitory concentration value of osimertinib in 41 lung adenocarcinoma cell lines. The Student t-test, Kaplan-Meier analysis, and Pearson correlation analysis were used in data analysis. Results: We found that circ7312 knockdown increased miR-764 expression and decreased MAPK1 expression, and circ7312 regulated MAPK1 by sponging miR-764. In addition, high circ7312 expression has significant positive correlation with osimertinib IC50 values, circ7312 knockdown decreased the cell half maximal inhibitory concentration value of osimertinib and increased pyroptosis and apoptosis by sponging the miR-764/MAPK1 axis. We also found that circ7312 and MAPK1 were highly expressed in tumor tissues and related to poor prognosis. Xenograft experiments revealed that circ7312 knockdown decreased osimertinib resistance in vivo. Conclusion: We demonstrated that the inhibition of circ7312 decreased osimertinib resistance by promoting pyroptosis and apoptosis via the miR-764/MAPK1 axis, providing a novel target for osimertinib resistance therapy.
Collapse
Affiliation(s)
- Chenyue Dai
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zeming Ma
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Jiahui Si
- Department of Anesthesiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Guo An
- Department of Laboratory Animals, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Wenlong Zhang
- Department of Laboratory Animals, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Shaolei Li
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yuanyuan Ma
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
10
|
Valacchi G, Pambianchi E, Coco S, Pulliero A, Izzotti A. MicroRNA Alterations Induced in Human Skin by Diesel Fumes, Ozone, and UV Radiation. J Pers Med 2022; 12:176. [PMID: 35207665 PMCID: PMC8880698 DOI: 10.3390/jpm12020176] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 11/17/2022] Open
Abstract
Epigenetic alterations are a driving force of the carcinogenesis process. MicroRNAs play a role in silencing mutated oncogenes, thus defending the cell against the adverse consequences of genotoxic damages induced by environmental pollutants. These processes have been well investigated in lungs; however, although skin is directly exposed to a great variety of environmental pollutants, more research is needed to better understand the effect on cutaneous tissue. Therefore, we investigated microRNA alteration in human skin biopsies exposed to diesel fumes, ozone, and UV light for over 24 h of exposure. UV and ozone-induced microRNA alteration right after exposure, while the peak of their deregulations induced by diesel fumes was reached only at the end of the 24 h. Diesel fumes mainly altered microRNAs involved in the carcinogenesis process, ozone in apoptosis, and UV in DNA repair. Accordingly, each tested pollutant induced a specific pattern of microRNA alteration in skin related to the intrinsic mechanisms activated by the specific pollutant. These alterations, over a short time basis, reflect adaptive events aimed at defending the tissue against damages. Conversely, whenever environmental exposure lasts for a long time, the irreversible alteration of the microRNA machinery results in epigenetic damage contributing to the pathogenesis of inflammation, dysplasia, and cancer induced by environmental pollutants.
Collapse
Affiliation(s)
- Giuseppe Valacchi
- Animal Science Department, Plants for Human Health Institute, North Carolina State University, Research Campus Kannapolis, Kannapolis, NC 28081, USA; (G.V.); (E.P.)
- Department of Environmental Sciences and Prevention, University of Ferrara, 44121 Ferrara, Italy
- Department of Food and Nutrition, Kyung Hee University, Seoul 130-701, Korea
| | - Erika Pambianchi
- Animal Science Department, Plants for Human Health Institute, North Carolina State University, Research Campus Kannapolis, Kannapolis, NC 28081, USA; (G.V.); (E.P.)
| | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | | | - Alberto Izzotti
- Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
- UOC Mutagenesis and Cancer Prevention, IRCCS San Martino Hospital, 16132 Genova, Italy
| |
Collapse
|
11
|
Chen J, Liu Z, Ma L, Gao S, Fu H, Wang C, Lu A, Wang B, Gu X. Targeting Epigenetics and Non-coding RNAs in Myocardial Infarction: From Mechanisms to Therapeutics. Front Genet 2022; 12:780649. [PMID: 34987550 PMCID: PMC8721121 DOI: 10.3389/fgene.2021.780649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) is a complicated pathology triggered by numerous environmental and genetic factors. Understanding the effect of epigenetic regulation mechanisms on the cardiovascular disease would advance the field and promote prophylactic methods targeting epigenetic mechanisms. Genetic screening guides individualised MI therapies and surveillance. The present review reported the latest development on the epigenetic regulation of MI in terms of DNA methylation, histone modifications, and microRNA-dependent MI mechanisms and the novel therapies based on epigenetics.
Collapse
Affiliation(s)
- Jinhong Chen
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Zhichao Liu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Li Ma
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Shengwei Gao
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Huanjie Fu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Can Wang
- Acupuncture Department, The First Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Anmin Lu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Baohe Wang
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Xufang Gu
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| |
Collapse
|
12
|
Liu W, Lin W, Yu L. Long non-coding RNA muscleblind like splicing regulator 1 antisense RNA 1 (LncRNA MBNL1-AS1) promotes the progression of acute myocardial infarction by regulating the microRNA-132-3p/SRY-related high-mobility-group box 4 (SOX4) axis. Bioengineered 2022; 13:1424-1435. [PMID: 34978261 PMCID: PMC8805923 DOI: 10.1080/21655979.2021.2018974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 12/10/2021] [Indexed: 12/18/2022] Open
Abstract
Long non-coding RNA muscleblind like splicing regulator 1 antisense RNA 1 (LncRNA MBNL1-AS1) exerts vital role in various physiological processes. However, its functions in acute myocardial infarction (AMI) are not elucidated. AMI model was constructed using Wistar rats and it was found that LncRNA MBNL1-AS1 was upregulated in AMI model according to the quantitative real-time polymerase chain reaction (qRT-PCR) results. The left ventricular systolic pressure (LVSP), left ventricular end diastolic pressure (LVEDP) and maximum rate of rise/fall of left ventricle pressure (±dp/dt max) were detected through hemodynamics test, which showed that knockdown of MBNL1-AS1 improved cardiac function in AMI model. Next, the myocardial infarction area was estimated by triphenyltetrazole chloride (TTC) staining, and the levels of cardiac troponin I (cTn-I) and creatine kinase-MB (CK-MB) were detected by enzyme-linked immunosorbent assay (ELISA) kit. The results revealed that silencing MBLN1-AS1 alleviated myocardial injury in AMI model. Additionally, MBNL1-AS1 knockdown inhibited apoptosis of myocardial cells and reduced the expression of apoptotic proteins. According to DIANA database and luciferase reporter assay, miR-132-3p was the direct target of MBNL1-AS1 and was negatively regulated by MBNL1-AS1. Furthermore, Targetscan database predicted that SRY-related high-mobility-group box 4 (SOX4) was the direct target of miR-132-3p and was regulated by MBNL1-AS1 through miR-132-3p. Moreover, overexpression of SOX4 partially eliminated effects of MBNL1-AS1 on myocardial cells. In conclusion, this investigation for the first time revealed that LncRNA MBNL1-AS1 was the potential target for treating AMI and expounded the underlying mechanisms of it.
Collapse
Affiliation(s)
- Weifeng Liu
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| | - Wenyuan Lin
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| | - Liangliang Yu
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao Medical College, Qingdao University, Yantai, China
| |
Collapse
|
13
|
Dai C, Liu B, Li S, Hong Y, Si J, Xiong Y, Wu N, Ma Y. Construction of a circRNA-miRNA-mRNA Regulated Pathway Involved in EGFR-TKI Lung Adenocarcinoma Resistance. Technol Cancer Res Treat 2021; 20:15330338211056809. [PMID: 34825849 PMCID: PMC8647233 DOI: 10.1177/15330338211056809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objectives: Epidermal growth factor receptor-tyrosine kinase
inhibitors are widely used for lung epidermal growth factor receptor-positive
lung adenocarcinomas, but acquired resistance is inevitable. Although non-coding
RNAs, such as circular RNA and microRNA, are known to play vital roles in
epidermal growth factor receptor-tyrosine kinase inhibitor resistance,
comprehensive analysis is lacking. Thus, this study aimed to explore the
circular RNA-microRNA-messenger RNA regulatory network involved in epidermal
growth factor receptor-tyrosine kinase inhibitor resistance.
Methods: To identify differentially expressed genes between the
epidermal growth factor receptor-tyrosine kinase inhibitor sensitive cell line
PC9 and resistant cell line PC9/ epidermal growth factor receptor-tyrosine
kinase inhibitor resistance(PC9/ER), circular RNA, microRNA and messenger RNA
microarrays were performed. Candidates were then identified to construct a
circular RNA-microRNA-messenger RNA network using bioinformatics. Additionally,
Gene Oncology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were
conducted to evaluate the network messenger RNA, setting up a protein-protein
interaction network for hub-gene identification. Afterwards, RNA
immunoprecipitation was performed to enrich microRNA, and quantitative real-time
PCR was used to estimated gene expression levels. Results: In
total, 603, 377, and 1863 differentially expressed circular RNA, microRNA,
messenger RNAs, respectively, were identified using microarray analysis,
constructing a circular RNA-microRNA-messenger RNA network containing 18
circular RNAs, 17 microRNAs and 175 messenger RNAs. Moreover, Gene Oncology and
Kyoto Encyclopedia of Genes and Genomes pathway analyses showed that the most
enriched biological process terms and pathways were related to epidermal growth
factor receptor-tyrosine kinase inhibitor resistance, including Wnt and Hippo
signaling pathways. Based on the competing endogenous RNA and protein-protein
interaction network, circ-0007312 was showed to interact with miR-764 and both
circ-0003748 and circ-0001398 were shown to interact with miR-628; both these
microRNAs targeted MAPK1. Furthermore, circ-0007312, circ-0003748, circ-0001398,
and MAPK1 were up-regulated, whereas miR-764 and miR-628 were downregulated in
PC9/ER cells as compared to parental PC9 cells. We also found that circ-0007312
and miR-764 were positively expressed in plasma. Conclusions: Our
original study associated with mechanism of target therapy in lung cancer
provided a systematic and comprehensive regulation of circular RNA, microRNA and
messenger RNA in epidermal growth factor receptor-tyrosine kinase inhibitor
resistance. It was found that circ-0007312- miR-764-MAPK1,
circ-0003748-miR-628-MAPK1, and circ-0001398-miR-628-MAPK1 axis may play key
roles in epidermal growth factor receptor-tyrosine kinase inhibitor
resistance.
Collapse
Affiliation(s)
- Chenyue Dai
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 12519Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Bing Liu
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 12519Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Shaolei Li
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 12519Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Yang Hong
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 12519Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Jiahui Si
- Department of Anesthesiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 12519Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Ying Xiong
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 12519Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Nan Wu
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 12519Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Yuanyuan Ma
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 12519Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| |
Collapse
|