1
|
Bai J, Yang SY, Yu SM, Cao Y, Ma CH, Hu XY, Chen X, Song YN, Chen HJ. IGFBP5 mediates the therapeutic effect of isoliquiritigenin in myocardial ischemia-reperfusion injury via AKT/GLUT4 regulated insulin resistance. Front Pharmacol 2025; 16:1544869. [PMID: 40365313 PMCID: PMC12069378 DOI: 10.3389/fphar.2025.1544869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Background Myocardial ischemia/reperfusion injury (MIRI) is a critical problem in cardiovascular medicine, often occurring after coronary revascularization procedures or cardiopulmonary bypass. The characters of MIRI are both energy metabolism disturbances and severe myocardium insulin resistance (IR), which exacerbated myocardial damage and cell death. Isoliquiritigenin (ISL), a flavonoid derived from licorice roots (Glycyrrhiza spp.), has demonstrated protective effects on MIRI. However, the potential cardio-protective effects and mechanism of ISL in MIRI remain unclear. Propose In this study, we aimed to investigate ISL's therapeutic effects on MIRI. Moreover, we elucidate the underlying mechanisms of ISL regulated myocardium insulin resistance in vivo and in vitro. Methods In vivo, SD rats underwent left anterior descending coronary artery ligation/reperfusion to induce MIRI. Chest echocardiography was performed to monitor cardiac function post-reperfusion, followed by measurement of myocardial injury and IR markers. In vitro, H9C2 cardiomyocytes subjected to oxygen-glucose deprivation/reperfusion (OGD/R). Markers associated with myocardial injury and IR were assessed. Then, we identified potential therapeutic targets IGFBP5 for MIRI by network pharmacology and molecular docking analysis. Finally, lentivirus were used to silence or over-express IGFBP5 to elucidate the role of IGFBP5 in regulating the therapeutic effects of ISL on IR in MIRI. Results In the present study, In vivo experiments demonstrated that ISL attenuated myocardial infarct size, decreased serum markers of myocardial injury, improved left ventricular systolic function, and enhanced insulin sensitivity. In vitro data revealed that ISL ameliorated glucose uptake and cell survival rate. Furthermore, ISL increased AKT phosphorylation and upregulated membrane-bound GLUT4 (M-GLUT4) protein expression levels. These effects of ISL are mediated by the induction of IGFBP5, as demonstrated using gene-specific shRNA or overexpression for IGFBP5. Conclusion Our results reveal that ISL protects against myocardial damage caused by MIRI through the regulation of IR via the IGFBP5/AKT/GLUT4 pathway.
Collapse
Affiliation(s)
- Jue Bai
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Si-Yuan Yang
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Shao-Mei Yu
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Ying Cao
- Department of Anesthesiology, The Affliated JinYang Hospital of Guizhou Medical University, The Second People’s Hospital of Guiyang, Guiyang, Guizhou, China
| | - Chang-Han Ma
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xuan-Yi Hu
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Xiong Chen
- Department of Endocrinology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying-Nan Song
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Hong-Jin Chen
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, Guizhou, China
- Division of cardiac surgery, Guizhou Institute of Precision Medicine, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Department of Pharmacology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
2
|
Zhu Q, Lu X, Chen M, Zhang T, Shi M, Yao W, Zhang H, Gao R, Li X, Zhou Y, Liao S. IGFBP5 affects cardiomyocyte survival and functional recovery in mice following myocardial ischemia. Commun Biol 2024; 7:1594. [PMID: 39613849 DOI: 10.1038/s42003-024-07304-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
Insulin-like growth factor-binding protein 5 (IGFBP5) has been shown to be useful for the diagnosis and treatment of multiple tumors and cerebrovascular diseases. However, it is unknown whether IGFBP5 is involved in myocardial repair following myocardial infarction (MI). Here we show high expression of IGFBP5 in multiple models of ischemic and hypoxic injury. IGFBP5 affected the proliferation of neonatal rat cardiomyocytes (NRCMs) and the cardiomyocyte apoptosis induced by oxygen-glucose deprivation (OGD). Subsequently, heart-specific IGFBP5 knockdown inhibited myocardial apoptosis and increased cardiomyocyte proliferation in mice with MI. During the chronic remodeling stage, heart-specific regulation of IGFBP5 ameliorated pathological cardiac remodeling and dysfunction. Mechanistically, IGFBP5 regulated cardiomyocyte survival through the insulin-like growth factor 1 (IGF1) receptor (IGF1R)/protein kinase B (PKB/AKT) pathway. In summary, our results provide mechanistic insights into the effect of IGFBP5 on cardiomyocyte during cardiac repair. IGFBP5 may represent a therapeutic target for myocardial ischemic injury.
Collapse
Affiliation(s)
- Qingqing Zhu
- Division of Cardiac Surgery Intensive Care Unit, Department of Cardiac Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xinyi Lu
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Mengli Chen
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Ting Zhang
- Department of Cardiovascular Medicine, The Air Force Hospital from Eastern Theater, Nanjing, China
| | - Mengsha Shi
- Department of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wenming Yao
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Haifeng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Rongrong Gao
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Xinli Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Yanli Zhou
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China.
| | - Shengen Liao
- National Key Laboratory for Innovation and Transformation of Luobing Theory. Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China.
| |
Collapse
|
3
|
Li Y, Ni SH, Liu X, Sun SN, Ling GC, Deng JP, Ou-Yang XL, Huang YS, Li H, Chen ZX, Huang XF, Xian SX, Yang ZQ, Wang LJ, Wu HY, Lu L. Crosstalk between endothelial cells with a non-canonical EndoMT phenotype and cardiomyocytes/fibroblasts via IGFBP5 aggravates TAC-induced cardiac dysfunction. Eur J Pharmacol 2024; 966:176378. [PMID: 38309679 DOI: 10.1016/j.ejphar.2024.176378] [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: 12/17/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
Heart failure (HF) is a complex chronic condition characterized by structural and functional impairments. The differentiation of endothelial cells into myofibroblasts (EndoMT) in response to cardiac fibrosis is controversial, and the relative contribution of endothelial plasticity remains to be explored. Single-cell RNA sequencing was used to identify endothelial cells undergoing fibrotic differentiation within 2 weeks of transverse aortic constriction (TAC). This subset of endothelial cells transiently expressed fibrotic genes but had low expression of alpha-smooth muscle actin, indicating a non-canonical EndoMT, which we named a transient fibrotic-like phenotype (EndoFP). The role of EndoFP in pathological cardiac remodeling may be correlated with increased levels of osteopontin. Cardiomyocytes and fibroblasts co-cultured with EndoFP exhibited heightened pro-hypertrophic and pro-fibrotic effects. Mechanistically, we found that the upregulated expression of insulin-like growth factor-binding protein 5 may be a key mediator of EndoFP-induced cardiac dysfunction. Furthermore, our findings suggested that Rab5a is a novel regulatory gene involved in the EndoFP process. Our study suggests that the specific endothelial subset identified in TAC-induced pressure overload plays a critical role in the cellular interactions that lead to cardiac fibrosis and hypertrophy. Additionally, our findings provide insight into the mechanisms underlying EndoFP, making it a potential therapeutic target for early heart failure.
Collapse
Affiliation(s)
- Yue Li
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen, 518000, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Shi-Hao Ni
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Xin Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China; School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shu-Ning Sun
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Gui-Chen Ling
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Jian-Ping Deng
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Xiao-Lu Ou-Yang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Yu-Sheng Huang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Huan Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Zi-Xin Chen
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Xiu-Fang Huang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Shao-Xiang Xian
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Zhong-Qi Yang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China
| | - Ling-Jun Wang
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China.
| | - Hong-Yan Wu
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Shenzhen Luohu Hospital of Traditional Chinese Medicine, Shenzhen, 518000, China.
| | - Lu Lu
- Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China; Key Laboratory of Chronic Heart Failure, Guangzhou University of Chinese Medicine, Guangzhou, 510407, China.
| |
Collapse
|
4
|
Liu Y, Chen LJ, Li XW, Yang JZ, Liu JL, Zhang KK, Li JH, Wang Q, Xu JT, Zhi X. Gut microbiota contribute to Methamphetamine-induced cardiotoxicity in mouse model. Chem Biol Interact 2023; 379:110512. [PMID: 37116852 DOI: 10.1016/j.cbi.2023.110512] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
Methamphetamine (METH) is a psychotropic drug known to cause cardiotoxicity. The gut-heart axis is emerging as an important pathway linking gut microbiota to cardiovascular disease, but the precise association between METH-induced cardiotoxicity and gut microbiota has yet to be elucidated. In this study, we established an escalating dose-multiple METH administration model in male BALB/c mice, examined cardiac injury and gut microbiota, and investigated the contribution of gut microbiota to cardiotoxicity induced by METH. Additionally, we treated mice with antibiotics and fecal microbiota transplantation (FMT) to assess the impact of gut microbiota on cardiotoxicity. Our results showed that METH exposure altered the p53 and PI3K/Akt signaling pathways and modulated the apoptosis pathway in heart tissue, accompanied by elevated levels of Bax/BCL-2 expression and cleaved caspase-3 proteins. METH exposure increased the diversity and richness of gut microbiota, and significantly changed the microbial community composition, accompanied by elevated abundance of Lactobacillus, Bifidobacterium, and decreased abundance of Bacteroides, norank_f_Muribaculaceae and Alistipes. Eliminating gut microbiota by antibiotics treatment alleviated METH-induced cardiotoxicity, while FMT treatment transferred similar cardiac injury manifestations from METH-exposed mice to healthy recipient mice. Our study unveils the crucial involvement of gut microbiota in the development of cardiotoxicity induced by METH and provides potential strategies for treating cardiac complications caused by METH.
Collapse
Affiliation(s)
- Yi Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Li-Jian Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Xiu-Wen Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jian-Zheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jia-Li Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Kai-Kai Zhang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jia-Hao Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China.
| | - Jing-Tao Xu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, China.
| | - Xu Zhi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
5
|
Ock J, Suh JK, Hong SS, Kang JH, Yin GN, Ryu JK. IGFBP5 antisense and short hairpin RNA (shRNA) constructs improve erectile function by inducing cavernosum angiogenesis in diabetic mice. Andrology 2023; 11:358-371. [PMID: 35866351 PMCID: PMC10087557 DOI: 10.1111/andr.13234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 05/17/2022] [Accepted: 05/26/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND The incidence of diabetic erectile dysfunction (ED) is rapidly increasing, and due to the severe angiopathy caused by diabetes, current drugs are ineffective at treating ED. Insulin-like growth factor-binding protein 5 (IGFBP5) promotes cell death and induces apoptosis in various cell types. OBJECTIVES To evaluate the effectiveness of IGFBP5 knockdown in improving erectile function in diabetic mice. MATERIALS AND METHODS Diabetes was induced by injecting streptozotocin (STZ) intraperitoneally into male 8-week-old C57BL/6 mice. Eight weeks after diabetes induction, mice were divided into four groups: a nondiabetic control group and three STZ-induced diabetic mice groups, which were administered intracavernous injections of phosphate buffered saline, scrambled control shRNA, or shRNA targeting mouse IGFBP5 (shIGFBP5) lentivirus particles. Two weeks later, we measured erectile function by electrically stimulating the bilateral cavernous nerve. To mimic diabetic angiopathy, primary cavernous endothelial cells (MCECs) from healthy mice were cultured and treated with glucose. RESULTS IGFBP5 expression in MCECs or cavernous tissues were significantly increased under diabetic conditions, and knockdown of IGFBP5 induced MCECs angiogenic activity under high-glucose conditions. STZ-induced diabetic mice had reduced erectile function, but shIGFBP5 treatment resulted in significant improvements (to 90% of the nondiabetic control group level). Furthermore, in diabetic mice, numbers of cavernous endothelial cells, pericytes, and neuronal cells were increased by shIGFBP5 treatment, which also increased eNOS Ser1177 phosphorylation, decreased permeability and apoptosis of cavernous endothelial cells. In addition, IGFBP5 was found to mediate the AKT, ERK, p38 signaling pathways. DISCUSSION AND CONCLUSION Knockdown of IGFBP5 improved erectile function in diabetic mice by promoting cell proliferation and reducing apoptosis and permeability. Local inhibition of IGFBP5 expression may provide a new treatment strategy for diabetic ED and other ischemic vascular or neurological diseases.
Collapse
Affiliation(s)
- Jiyeon Ock
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Jun-Kyu Suh
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Soon-Sun Hong
- Department of Biomedical Sciences, College of Medicine, and Program in Biomedical Science and Engineering, Inha University, Incheon, Republic of Korea
| | - Ju-Hee Kang
- Department of Pharmacology and Medicinal Toxicology Research Center, Inha University College of Medicine, Incheon, Republic of Korea
| | - Guo Nan Yin
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Ji-Kan Ryu
- National Research Center for Sexual Medicine and Department of Urology, Inha University School of Medicine, Incheon, Republic of Korea
| |
Collapse
|
6
|
Tang X, Jiang H, Lin P, Zhang Z, Chen M, Zhang Y, Mo J, Zhu Y, Liu N, Chen X. Insulin-like growth factor binding protein-1 regulates HIF-1α degradation to inhibit apoptosis in hypoxic cardiomyocytes. Cell Death Discov 2021; 7:242. [PMID: 34531382 PMCID: PMC8445926 DOI: 10.1038/s41420-021-00629-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 08/22/2021] [Accepted: 09/01/2021] [Indexed: 12/12/2022] Open
Abstract
Hypoxia is important in ischemic heart disease. Excessive Insulin-like growth factor binding protein-1 (IGFBP-1) amounts are considered to harm cardiomyocytes in acute myocardial infarction. However, the mechanisms by which IGFBP-1 affects cardiomyocytes remain undefined. The present study demonstrated that hypoxia up-regulates IGFBP-1 and HIF-1α protein expression in cardiomyocytes. Subsequent assays showed that IGFBP-1 suppression decreased HIF-1α expression and inhibited hypoxia-induced apoptosis in cardiomyocytes, which was reversed by HIF-1α overexpression, indicating that HIF-1α is essential to IGFBP-1 function in cellular apoptosis. In addition, we showed that IGFBP-1 regulated HIF-1α stabilization through interacting with VHL. The present findings suggest that IGFBP-1–HIF-1α could be targeted for treating ischemic heart disease.
Collapse
Affiliation(s)
- Xiaoyan Tang
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Huilin Jiang
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Peiyi Lin
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Zhenhui Zhang
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Meiting Chen
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Yi Zhang
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Junrong Mo
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Yongcheng Zhu
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China
| | - Ningning Liu
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China. .,Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, China.
| | - Xiaohui Chen
- Department of Emergency, the Second Affiliated Hospital, Guangzhou Medical University, 510260, Guangzhou, Guangdong, China.
| |
Collapse
|
7
|
Seeley SL, D'Souza MS, Stoops TS, Rorabaugh BR. Short term methylphenidate treatment does not increase myocardial injury in the ischemic rat heart. Physiol Res 2020; 69:803-812. [PMID: 32469230 DOI: 10.33549/physiolres.934368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Methylphenidate is commonly used for the treatment of attention deficit hyperactivity disorder. The cardiovascular safety of methylphenidate has been a subject of debate with some studies indicating that methylphenidate increases the likelihood of experiencing a myocardial infarction. However, it is unknown whether methylphenidate worsens the extent of injury during an ischemic insult. The purpose of this study was to determine whether short term exposure to methylphenidate increases the extent of myocardial injury during an ischemic insult. Male and female rats received methylphenidate (5 mg/kg/day) or saline for 10 days by oral gavage. Hearts were subjected to 20 min of ischemia and 2 h of reperfusion on a Langendorff isolated heart apparatus on day 11. Cardiac contractile function was monitored via an intraventricular balloon and myocardial injury was assessed by triphenyltetrazolium chloride staining. Methylphenidate significantly increased locomotor activity in male and female rats, confirming absorption of this psychostimulant into the central nervous system. Male hearts had significantly larger infarcts than female hearts, but methylphenidate had no impact on infarct size or postischemic recovery of contractile function in hearts of either sex. These data indicate that methylphenidate does not increase the extent of injury induced by an ischemic insult.
Collapse
Affiliation(s)
- S L Seeley
- Marshall University School of Pharmacy, Huntington, WV, USA.
| | | | | | | |
Collapse
|
8
|
Lai G, Wang L, Li Z, Zhao Y. Homocysteine downregulates cardiac homeobox transcription factor NKX2.5 via IGFBP5. Am J Physiol Heart Circ Physiol 2020; 319:H1380-H1386. [PMID: 33035436 DOI: 10.1152/ajpheart.00347.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Homocysteine (Hcy) is an independent risk factor of congenital heart disease (CHD), but its exact underlying mechanism is unclear. In this study, we collected amniotic fluid (AF) supernatant samples from pregnant women carrying CHD-affected (n = 16) or normal (n = 16) fetuses. We found that Hcy concentrations were higher in the AF of the CHD group when compared with normal pregnancies. Also, Western blot showed that NK2 homeobox 5 (NKX2.5) was decreased and insulin-like growth factor binding protein 5 (IGFBP5) was increased in the AF of the CHD group. In the H9C2 cell culture experiment, 500 μmol/L Hcy downregulated NKX2.5 and upregulated IGFBP5. Real-time PCR and Western blot showed that NKX2.5 expression was reduced in H9C2 cells treated with IGFBP5. Luciferase reporter gene demonstrated that IGFBP5 decreased the transcription of the NKX2.5 promoter. Chromatin immunoprecipitation and electrophoretic mobility shift assay suggested that IGFBP5 binds to the NKX2.5 promoter region. Thus, the data indicated that one of the possible mechanisms by which Hcy is involved in CHD may be that Hcy inhibits NKX2.5 expression partly through IGFBP5.NEW & NOTEWORTHY We found that Hcy and IGFBP5 were increased, whereas NKX2.5 was decreased, in AF of CHD. Meanwhile, Hcy could upregulate IGFBP5 but downregulate NKX2.5, and IGFBP5 inhibited NKX2.5 expression in vitro. Moreover, IGFBP5 can bind to the NKX2.5 promoter region and reduce NKX2.5 transcriptional activity.
Collapse
Affiliation(s)
- Guangrui Lai
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Leitong Wang
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.,Department of Reproductive Laboratory, Shenyang Jinghua Hospital, Shenyang, Liaoning, People's Republic of China
| | - Zhen Li
- Department of Obstetrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yanyan Zhao
- Department of Clinical Genetics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
9
|
Sancho Santos ME, Grabicová K, Steinbach C, Schmidt-Posthaus H, Šálková E, Kolářová J, Vojs Staňová A, Grabic R, Randák T. Environmental concentration of methamphetamine induces pathological changes in brown trout (Salmo trutta fario). CHEMOSPHERE 2020; 254:126882. [PMID: 32957289 DOI: 10.1016/j.chemosphere.2020.126882] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 04/03/2020] [Accepted: 04/23/2020] [Indexed: 06/11/2023]
Abstract
Methamphetamine, mainly consumed as an illicit drug, is a potent addictive psychostimulant that has been detected in surface water at concentrations ranging from nanograms to micrograms per litre, especially in Middle and East Europe. The aim of this study was to expose brown trout (Salmo trutta fario) to environmental (1 μg L-1) and higher (50 μg L-1) concentrations of methamphetamine for 35 days with a four-day depuration phase to assess the possible negative effects on fish health. Degenerative liver and heart alterations, similar to those described in mammals, were observed at both concentrations, although at different intensities. Apoptotic changes in hepatocytes, revealed by activated caspase-3, were found in exposed fish. The parent compound and a metabolite (amphetamine) were detected in fish tissues in both concentration groups, in the order of kidney > liver > brain > muscle > plasma. Bioconcentration factors ranged from 0.13 to 80. A therapeutic plasma concentration was reached for both compounds in the high-concentration treatment. This study indicates that chronic environmental concentrations of methamphetamine can lead to health issues in aquatic organisms.
Collapse
Affiliation(s)
- Maria Eugenia Sancho Santos
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic.
| | - Kateřina Grabicová
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Christoph Steinbach
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Heike Schmidt-Posthaus
- University of Bern, Centre for Fish and Wildlife Health, Department of Infectious Diseases and Pathobiology, Laenggassstrasse 122, 3001, Bern, Switzerland
| | - Eva Šálková
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Jitka Kolářová
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Andrea Vojs Staňová
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic; Comenius University in Bratislava, Faculty of Natural Sciences, Department of Analytical Chemistry, Ilkovicova 6, SK-842 15, Bratislava, Slovak Republic
| | - Roman Grabic
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| | - Tomáš Randák
- University of South Bohemia in České Budějovice, Faculty of Fisheries and Protection of Waters, South Bohemian Research Center of Aquaculture and Biodiversity of Hydrocenoses, Zátiší 728/II, 389 25, Vodňany, Czech Republic
| |
Collapse
|
10
|
Duan C, Allard JB. Insulin-Like Growth Factor Binding Protein-5 in Physiology and Disease. Front Endocrinol (Lausanne) 2020; 11:100. [PMID: 32194505 PMCID: PMC7063065 DOI: 10.3389/fendo.2020.00100] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 02/17/2020] [Indexed: 12/25/2022] Open
Abstract
Insulin-like growth factor (IGF) signaling is regulated by a conserved family of IGF binding proteins (IGFBPs) in vertebrates. Among the six distinct types of IGFBPs, IGFBP-5 is the most highly conserved across species and has the broadest range of biological activities. IGFBP-5 is expressed in diverse cell types, and its expression level is regulated by a variety of signaling pathways in different contexts. IGFBP-5 can exert a range of biological actions including prolonging the half-life of IGFs in the circulation, inhibition of IGF signaling by competing with the IGF-1 receptor for ligand binding, concentrating IGFs in certain cells and tissues, and potentiation of IGF signaling by delivery of IGFs to the IGF-1 receptor. IGFBP-5 also has IGF-independent activities and is even detected in the nucleus. Its broad biological activities make IGFBP-5 an excellent representative for understanding IGFBP functions. Despite its evolutionary conservation and numerous biological activities, knockout of IGFBP-5 in mice produced only a negligible phenotype. Recent research has begun to explain this paradox by demonstrating cell type-specific and physiological/pathological context-dependent roles for IGFBP-5. In this review, we survey and discuss what is currently known about IGFBP-5 in normal physiology and human disease. Based on recent in vivo genetic evidence, we suggest that IGFBP-5 is a multifunctional protein with the ability to act as a molecular switch to conditionally regulate IGF signaling.
Collapse
|
11
|
Zoubková H, Tomášková A, Nohejlová K, Černá M, Šlamberová R. Prenatal Exposure to Methamphetamine: Up-Regulation of Brain Receptor Genes. Front Neurosci 2019; 13:771. [PMID: 31417344 PMCID: PMC6686742 DOI: 10.3389/fnins.2019.00771] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/09/2019] [Indexed: 01/10/2023] Open
Abstract
Methamphetamine (METH) is a widespread illicit drug. If it is taken by pregnant women, it passes through the placenta and just as it affects the mother, it can impair the development of the offspring. The aim of our study was to identify candidates to investigate for changes in the gene expression in the specific regions of the brain associated with addiction to METH in rats. We examined the various areas of the central nervous system (striatum, hippocampus, prefrontal cortex) for signs of impairment in postnatal day 80 in experimental rats, whose mothers had been administered METH (5 mg/kg/day) during the entire gestation period. Changes in the gene expression at the mRNA level were determined by two techniques, microarray and real-time PCR. Results of two microarray trials were evaluated by LIMMA analysis. The first microarray trial detected either up-regulated or down-regulated expression of 2189 genes in the striatum; the second microarray trial detected either up-regulated or down-regulated expression of 1344 genes in the hippocampus of prenatally METH-exposed rats. We examined the expression of 10 genes using the real-time PCR technique. Differences in the gene expression were counted by the Mann–Whitney U-test. Significant changes were observed in the cocaine- and amphetamine-regulated transcript prepropeptide, tachykinin receptor 3, dopamine receptor D3 gene expression in the striatum regions, in the glucocorticoid nuclear receptor Nr3c1 gene expression in the prefrontal cortex and in the carboxylesterase 2 gene expression in the hippocampus of prenatally METH-exposed rats. The microarray technique also detected up-regulated expression of trace amine-associated receptor 7 h gene in the hippocampus of prenatally METH-exposed rats. We have identified susceptible genes; candidates for the study of an impairment related to methamphetamine addiction in the specific regions of the brain.
Collapse
Affiliation(s)
- Hana Zoubková
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Anežka Tomášková
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Kateryna Nohejlová
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Marie Černá
- Department of Medical Genetics, Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Romana Šlamberová
- Department of Physiology, Third Faculty of Medicine, Charles University, Prague, Czechia
| |
Collapse
|
12
|
Sun X, Wang Y, Xia B, Li Z, Dai J, Qiu P, Ma A, Lin Z, Huang J, Wang J, Xie WB, Wang J. Methamphetamine produces cardiac damage and apoptosis by decreasing melusin. Toxicol Appl Pharmacol 2019; 378:114543. [PMID: 30904475 DOI: 10.1016/j.taap.2019.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/17/2022]
Abstract
Methamphetamine (METH) is an amphetamine-type drug that is highly addictive and widely abused. Many studies have shown that METH exposure causes severe damage not only to the nervous system but also to the cardiovascular system. Melusin protein is a mechanotransducer that plays an important role in maintaining normal heart function. However, the role of melusin in METH-induced cardiotoxicity has not yet been reported. We hypothesized that methamphetamine can produce cardiac damage and apoptosis by decreasing the quantity of melusin. To test this hypothesis, we determined the protein expression of melusin and apoptosis markers in METH-treated rats and primary rat cardiomyocytes. We also established a melusin-overexpressing cell model to assess the importance of melusin in maintaining antiapoptotic pathways. To confirm our findings from the in vitro and animal models, we also evaluated the apoptotic index of cardiomyocytes and the protein expression of apoptotic markers in postmortem heart tissues from deceased METH abusers and age-matched control subjects. The results showed that the apoptosis of cardiomyocytes was increased significantly and that the protein expression of melusin was decreased after exposure to METH in primary rat cardiomyocytes, in rats and in humans. METH treatment also decreased the expression of the downstream proteins FAK, IQGAP1, p-AKT, p-GSK3β, and p-ERK in primary rat cardiomyocytes and in vivo. After overexpression of melusin, the above effects were partially reversed in primary rat cardiomyocytes. We conclude that METH can produce cardiac damage and apoptosis by decreasing melusin, while melusin-activated signaling by phosphorylated AKT, phosphorylated GSK3β, and ERK may be resistant to methamphetamine-induced myocardial apoptosis.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Yu Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Bing Xia
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Zhu Li
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Jialin Dai
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Pingming Qiu
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ande Ma
- Department of Hygiene Inspection & Quarantine Science, Guangdong Province Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine and Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Jiang Huang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Jiawen Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China
| | - Wei-Bing Xie
- School of Forensic Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Jie Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang 550004, China.
| |
Collapse
|
13
|
Rorabaugh BR, Seeley SL, Stoops TS, D’Souza MS. Repeated exposure to methamphetamine induces sex-dependent hypersensitivity to ischemic injury in the adult rat heart. PLoS One 2017; 12:e0179129. [PMID: 28575091 PMCID: PMC5456396 DOI: 10.1371/journal.pone.0179129] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 05/24/2017] [Indexed: 12/23/2022] Open
Abstract
Background We previously reported that adult female, but not male rats that were prenatally exposed to methamphetamine exhibit myocardial hypersensitivity to ischemic injury. However, it is unknown whether hypersensitivity to ischemic injury develops when rats are exposed to methamphetamine during adulthood. The goal of this study was to determine whether methamphetamine exposure during adulthood sensitizes the heart to ischemic injury. Methods Adult male and female rats received daily injections of methamphetamine (5 mg/kg) or saline for 10 days. Their hearts were isolated on day 11 and subjected to a 20 min ischemic insult on a Langendorff isolated heart apparatus. Cardiac contractile function was measured by an intraventricular balloon, and infarct size was measured by triphenyltetrazolium chloride staining. Results Hearts from methamphetamine-treated females exhibited significantly larger infarcts and suppressed postischemic recovery of contractile function compared to hearts from saline-treated females. In contrast, methamphetamine had no effect on infarct size or contractile recovery in male hearts. Subsequent experiments demonstrated that hypersensitivity to ischemic injury persisted in female hearts following a 1 month period of abstinence from methamphetamine. Myocardial protein kinase C-ε expression, Akt phosphorylation, and ERK phosphorylation were unaffected by adult exposure to methamphetamine. Conclusions Exposure of adult rats to methamphetamine sex-dependently increases the extent of myocardial injury following an ischemic insult. These data suggest that women who have a heart attack might be at risk of more extensive myocardial injury if they have a recent history of methamphetamine abuse.
Collapse
Affiliation(s)
- Boyd R. Rorabaugh
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, Ohio Northern University, Ada, Ohio, United States of America
- * E-mail:
| | - Sarah L. Seeley
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, Ohio Northern University, Ada, Ohio, United States of America
| | - Thorne S. Stoops
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, Ohio Northern University, Ada, Ohio, United States of America
| | - Manoranjan S. D’Souza
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, Ohio Northern University, Ada, Ohio, United States of America
| |
Collapse
|
14
|
Reece AS, Norman A, Hulse GK. Acceleration of cardiovascular-biological age by amphetamine exposure is a power function of chronological age. HEART ASIA 2017; 9:30-38. [PMID: 28243315 DOI: 10.1136/heartasia-2016-010832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/05/2016] [Accepted: 11/07/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Amphetamine abuse is becoming more widespread internationally. The possibility that its many cardiovascular complications are associated with a prematurely aged cardiovascular system, and indeed biological organism systemically, has not been addressed. METHODS Radial arterial pulse tonometry was performed using the SphygmoCor system (Sydney). 55 amphetamine exposed patients were compared with 107 tobacco smokers, 483 non-smokers and 68 methadone patients (total=713 patients) from 2006 to 2011. A cardiovascular-biological age (VA) was determined. RESULTS The age of the patient groups was 30.03±0.51-40.45±1.15 years. This was controlled for with linear regression. The sex ratio was the same in all groups. 94% of amphetamine exposed patients had used amphetamine in the previous week. When the (log) VA was regressed against the chronological age (CA) and a substance-type group in both cross-sectional and longitudinal models, models quadratic in CA were superior to linear models (both p<0.02). When log VA/CA was regressed in a mixed effects model against time, body mass index, CA and drug type, the cubic model was superior to the linear model (p=0.001). Interactions between CA, (CA)2 and (CA)3 on the one hand and exposure type were significant from p=0.0120. The effects of amphetamine exposure persisted after adjustment for all known cardiovascular risk factors (p<0.0001). CONCLUSIONS These results show that subacute exposure to amphetamines is associated with an advancement of cardiovascular-organismal age both over age and over time, and is robust to adjustment. That this is associated with power functions of age implies a feed-forward positively reinforcing exacerbation of the underlying ageing process.
Collapse
Affiliation(s)
- Albert Stuart Reece
- School of Psychiatry and Clinical Neurosciences , University of Western Australia , Crawley, Western Australia , Australia
| | - Amanda Norman
- School of Psychiatry and Clinical Neurosciences , University of Western Australia , Crawley, Western Australia , Australia
| | - Gary Kenneth Hulse
- School of Psychiatry and Clinical Neurosciences , University of Western Australia , Crawley, Western Australia , Australia
| |
Collapse
|
15
|
Insulin-like growth factor binding protein 5 (IGFBP5) functions as a tumor suppressor in human melanoma cells. Oncotarget 2016; 6:20636-49. [PMID: 26010068 PMCID: PMC4653031 DOI: 10.18632/oncotarget.4114] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 04/22/2015] [Indexed: 12/12/2022] Open
Abstract
The insulin-like growth factor binding protein 5 (IGFBP5), which is often dysregulated in human cancers, plays a crucial role in carcinogenesis and cancer development. However, the function and underlying mechanism of IGFBP5 in tumor growth and metastasis has been elusive, particularly in malignant human melanoma. Here, we reported that IGFBP5 acts as an important tumor suppressor in melanoma tumorigenicity and metastasis by a series of experiments including transwell assay, xenograft model, in vivo tumor metastasis experiment, and RNA-Seq. Overexpression of IGFBP5 in A375, a typical human melanoma cell line, inhibited cell malignant behaviors significantly, including in vitro proliferation, anchorage-independent growth, migration and invasion, as well as in vivo tumor growth and pulmonary metastasis. In addition, overexpression of IGFBP5 suppressed epithelial-mesenchymal transition (EMT), and decreased the expression of E-cadherin and the key stem cell markers NANOG, SOX2, OCT4, KLF4, and CD133. Furthermore, IGFBP5 exerts its inhibitory activities by reducing the phosphorylation of IGF1R, ERK1/2, and p38-MAPK kinases and abating the expression of HIF1α and its target genes, VEGF and MMP9. All these findings were confirmed by IGFBP5 knockdown in human melanoma cell line A2058. Taken together, these results shed light on the mechanism of IGFBP5 as a potential tumor-suppressor in melanoma progression, indicating that IGFBP5 might be a novel therapeutic target for human melanoma.
Collapse
|
16
|
Chen R, Wang B, Chen L, Cai D, Li B, Chen C, Huang E, Liu C, Lin Z, Xie WB, Wang H. DNA damage-inducible transcript 4 (DDIT4) mediates methamphetamine-induced autophagy and apoptosis through mTOR signaling pathway in cardiomyocytes. Toxicol Appl Pharmacol 2016; 295:1-11. [PMID: 26825372 DOI: 10.1016/j.taap.2016.01.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/20/2016] [Accepted: 01/25/2016] [Indexed: 12/30/2022]
Abstract
Methamphetamine (METH) is an amphetamine-like psychostimulant that is commonly abused. Previous studies have shown that METH can induce damages to the nervous system and recent studies suggest that METH can also cause adverse and potentially lethal effects on the cardiovascular system. Recently, we demonstrated that DNA damage-inducible transcript 4 (DDIT4) regulates METH-induced neurotoxicity. However, the role of DDIT4 in METH-induced cardiotoxicity remains unknown. We hypothesized that DDIT4 may mediate METH-induced autophagy and apoptosis in cardiomyocytes. To test the hypothesis, we examined DDIT4 protein expression in cardiomyocytes and in heart tissues of rats exposed to METH with Western blotting. We also determined the effects on METH-induced autophagy and apoptosis after silencing DDIT4 expression with synthetic siRNA with or without pretreatment of a mTOR inhibitor rapamycin in cardiomyocytes using Western blot analysis, fluorescence microscopy and TUNEL staining. Our results showed that METH exposure increased DDIT4 expression and decreased phosphorylation of mTOR that was accompanied with increased autophagy and apoptosis both in vitro and in vivo. These effects were normalized after silencing DDIT4. On the other hand, rapamycin promoted METH-induced autophagy and apoptosis in DDIT4 knockdown cardiomyocytes. These results suggest that DDIT4 mediates METH-induced autophagy and apoptosis through mTOR signaling pathway in cardiomyocytes.
Collapse
Affiliation(s)
- Rui Chen
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China; Department of Forensic Medicine, Guangdong Medical University, Dongguan 523808, People's Republic of China
| | - Bin Wang
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Ling Chen
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Dunpeng Cai
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Bing Li
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Chuanxiang Chen
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Enping Huang
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou 510030, People's Republic of China
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine and Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Wei-Bing Xie
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China.
| | - Huijun Wang
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China.
| |
Collapse
|
17
|
Zhang X, Sha M, Yao Y, Da J, Jing D. Increased B-type-natriuretic peptide promotes myocardial cell apoptosis via the B-type-natriuretic peptide/long non-coding RNA LSINCT5/caspase-1/interleukin 1β signaling pathway. Mol Med Rep 2015; 12:6761-7. [PMID: 26323562 PMCID: PMC4626192 DOI: 10.3892/mmr.2015.4247] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 08/06/2015] [Indexed: 01/23/2023] Open
Abstract
Chronic heart failure (CHF) is the final stage of various heart diseases, and is increasingly recognized as a major health problem in the elderly. Previous studies demonstrated that B-type-natriuretic peptide (BNP) is an established biomarker of CHF. Furthermore, BNP also regulates cell proliferation, differentiation and apoptosis. Recent evidence has revealed that BNP affects myocardial cell apoptosis during myocardial ischemia-reperfusion injury. Long non-coding RNAs (lncRNAs) are emerging as novel molecular compounds involved in gene regulation, and have important roles in numerous human diseases. However, the mechanism underlying the BNP and lncRNA-induced regulation of myocardial cell apoptosis remains to be elucidated. The present study reported that lncRNA LSINCT5, upregulated by BNP, is able to regulate myocardial cell apoptosis via the activation of the caspase-1/interleukin (IL)-1β signaling pathway. BNP-induced apoptosis of HCM cells was observed using flow cytometry, and involved caspase-1. In addition, expression profiling using a human lncRNA polymerase chain reaction array revealed that LSINCT5 was highly expressed in BNP-treated myocardial cells, as compared with untreated cells. The role of lncRNA LSINCT5 in HCM cell apoptosis was also investigated. The results of the present study indicated that LSINCT5 silencing by small interfering RNA inhibits caspase-1/IL-1β signaling, and suppresses apoptosis in BNP-treated HCM cells. Therefore, high expression levels of BNP promote the apoptosis of myocardial cells through the lncRNA LSINCT5 mediator, which activates the caspase-1/IL-1β signaling pathway. These findings uncovered a novel pathogenic mechanism, and provided a potential therapeutic target for CHF.
Collapse
Affiliation(s)
- Xian Zhang
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Minglei Sha
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Yuting Yao
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Jia Da
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| | - Dadao Jing
- Department of Geriatrics, Shanghai First People's Hospital, Shanghai 200080, P.R. China
| |
Collapse
|