1
|
Feliciano RDS, Manchini MT, Atum ALB, da Silva GA, Antônio EL, Serra AJ, Tucci PJF, Andrade de Mello R, Chavantes MC, Baltatu OC, Silva Júnior JA. Photobiomodulation therapy's effects on cardiac fibrosis activation after experimental myocardial infarction. Lasers Surg Med 2022; 54:883-894. [PMID: 35366381 DOI: 10.1002/lsm.23544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/14/2022] [Accepted: 03/18/2022] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Ischemic heart disease is the leading cause of death worldwide, and interventions to reduce myocardial infarction (MI) complications are widely researched. Photobiomodulation therapy (PBMT) has altered multiple biological processes in tissues and organs, including the heart. OBJECTIVES This study aimed to assess the temporal effects of PBMT on cardiac fibrosis activation after MI in rats. In this proof-of-concept study, we monitored the change in expression patterns over time of genes and microRNAs (miRNAs) involved in the formation of cardiac fibrosis post-MI submitted to PBMT. MATERIALS AND METHODS Experimental MI was induced, and PBMT was applied shortly after coronary artery ligation (laser light of wavelength 660 nm, 15 mW of power, energy density 22.5 J/cm2 , 60 seconds of application, irradiated area 0.785 cm2 , fluence 1.1 J/cm2 ). Ventricular septal samples were collected at 30 minutes, 3, 6, 24 hours, and 3 days post-MI to determine temporal PBMT's effects on messenger RNA (mRNA) expression associated with cardiac fibrosis activation and miRNAs expression. RESULTS PBMT, when applied after ischemia, reversed the changes in mRNA expression of myocardial extracellular matrix genes induced by MI. Surprisingly, PBMT modified cardiac miRNAs expression related to fibrosis replacement in the myocardium. Expression correlations between myocardial mRNAs were assessed. The correlation coefficient between miRNAs and target mRNAs was also determined. A positive correlation was detected among miR-21 and transforming growth factor beta-1 mRNA. The miR-29a expression negatively correlated to Col1a1, Col3a1, and MMP-2 mRNA expressions. In addition, we observed that miR-133 and Col1a1 mRNA were negatively correlated. CONCLUSION The results suggest that PBMT, through the modulation of gene transcription and miRNA expressions, can interfere in cardiac fibrosis activation after MI, mainly reversing the signaling pathway of profibrotic genes.
Collapse
Affiliation(s)
| | - Martha T Manchini
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil.,Department of Cardiovascular Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Allan L B Atum
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | | | - Ednei L Antônio
- Department of Cardiovascular Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Andrey J Serra
- Department of Cardiovascular Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Paulo J F Tucci
- Department of Cardiovascular Physiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ramon Andrade de Mello
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | - Maria C Chavantes
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| | - Ovidiu C Baltatu
- Department of Public Health and Epidemiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates.,Center of Innovation, Technology and Education (CITE), Anhembi Morumbi University-Anima Institute, São José dos Campos, Brazil
| | - José A Silva Júnior
- Postgraduate Program in Medicine, Universidade Nove de Julho, UNINOVE, São Paulo, Brazil
| |
Collapse
|
2
|
Guo S, Tan Y, Huang Z, Li Y, Liu W, Fan X, Zhang J, Stalin A, Fu C, Wu Z, Wang P, Zhou W, Liu X, Wu C, Jia S, Zhang J, Duan X, Wu J. Revealing Calcium Signaling Pathway as Novel Mechanism of Danhong Injection for Treating Acute Myocardial Infarction by Systems Pharmacology and Experiment Validation. Front Pharmacol 2022; 13:839936. [PMID: 35281886 PMCID: PMC8905633 DOI: 10.3389/fphar.2022.839936] [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: 12/20/2021] [Accepted: 02/07/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction: Danhong injection (DHI) is a traditional Chinese medicine preparation commonly used in the clinical treatment of acute myocardial infarction (AMI). In this study, the active components of DHI and its mechanism in the treatment of AMI were investigated. Methods: The chemical components of DHI were detected by the ultra-high-performance liquid chromatography-linear trap quadrupole-orbitrap-tandem mass spectrometry (UHPLC-LTQ-Orbitrap-MS/MS), and the targets and pathways of DHI in the treatment of AMI were analyzed by systems pharmacology, which was verified by molecular docking and animal experiments. Results: A total of 12 active components of DHI were obtained, and 158 common targets of component and disease were identified by systems pharmacology. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis results showed that DHI is closely related to the calcium signaling pathway in the treatment of AMI. Molecular docking showed that the key target protein has good binding affinity to related compounds. The experimental results showed that compared with the model group, LVAWs, EF, and FS significantly (p < 0.05) increased in the DHI group. The percentage of myocardial infarction significantly (p < 0.01) decreased, both in the ventricular and total cardiac regions, and the pathological damage of myocardial tissue also decreased. In addition, the expression of the protein CaMK II decreased (p < 0.01) and the expression of SERCA significantly increased (p < 0.01). Conclusion: This study revealed that ferulic acid, caffeic acid and rosmarinic acid could inhibit AMI by regulating PLB, CaMK II, SERCA, etc. And mechanistically, calcium signaling pathway was critically involved. Combination of systems pharmacology prediction with experimental validation may provide a scientific basis for in-depth clinical investigation of the material basis of DHI.
Collapse
Affiliation(s)
- Siyu Guo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yingying Tan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihong Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yikui Li
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Weiyu Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaotian Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jingyuan Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Antony Stalin
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, China
| | - Changgeng Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhishan Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Penglong Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Zhou
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.,China-Japan Friendship Hospital, Beijing, China
| | - Xinkui Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Shanshan Jia
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jinyan Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaoxia Duan
- Beijing Zest Bridge Medical Technology Inc., Beijing, China
| | - Jiarui Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
3
|
Photobiomodulation therapy preconditioning modifies nitric oxide pathway and oxidative stress in human-induced pluripotent stem cell-derived ventricular cardiomyocytes treated with doxorubicin. Lasers Med Sci 2021; 37:1667-1675. [PMID: 34536182 DOI: 10.1007/s10103-021-03416-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 09/13/2021] [Indexed: 12/16/2022]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic that exhibits high heart toxicity. Human-induced pluripotent stem cell-derived ventricular cardiomyocytes (hiPSC-vCMs) are important in vitro models for testing drug cardiotoxicity. Photobiomodulation therapy (PBMT) is a non-invasive therapy that stimulates cells growth and self-repair using light irradiation. This study aimed to investigate the in vitro effects of PBMT preconditioning on cardiotoxicity induced by DOX. HiPSC-vCMs were treated with PBMT for 500 s, followed by the addition of 2 μM DOX. LED irradiation preconditioning parameters were at 660 nm with an irradiance of 10 mW/cm2, performing 5 J/cm2, followed by 24-h DOX exposure (2 μM). Human iPSC-vCMs treated with 2 μM DOX or irradiated with PBMT composed the second and third groups, respectively. The control group did neither receive PBMT preconditioning nor DOX and was irradiated with a white standard lamp. Cells from all groups were collected to perform mRNA and miRNA expressions quantification. PBMT, when applied before the DOX challenge, restored the viability of hiPSC-vCMs and reduced ROS levels. Although downregulated by DOX, myocardial UCP2 mRNA expression presented marked upregulation after PBMT preconditioning. Expression of eNOS and UCP2 mRNA and NO production were decreased after DOX exposure, and PBMT preconditioning before the DOX challenge reversed these changes. Moreover, our data indicated that PBMT preconditioning lowered the miR-24 expression. Our data suggested that PBMT preconditioning ameliorated in vitro DOX-induced cardiotoxicity on transcription level, restoring NO levels and reducing oxidative stress.
Collapse
|
4
|
Feliciano RDS, Atum ALB, Ruiz ÉGDS, Serra AJ, Antônio EL, Manchini MT, Silva JMA, Tucci PJF, Nathanson L, Morris M, Chavantes MC, Silva Júnior JA. Photobiomodulation Therapy on Myocardial Infarction in Rats: Transcriptional and Posttranscriptional Implications to Cardiac Remodeling. Lasers Surg Med 2021; 53:1247-1257. [PMID: 33846991 DOI: 10.1002/lsm.23407] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND OBJECTIVES Induction of myocardial infarction (MI) in rats by occlusion of the left anterior descending coronary artery is an experimental model used in research to elucidate functional, structural, and molecular modifications associated with ischemic heart disease. Photobiomodulation therapy (PBMT) has become a therapeutic alternative by modulating various biological processes eliciting several effects, including anti-inflammatory and pro-proliferative actions. The main objective of this work was to evaluate the effect of PBMT in the modulation of transcriptional and post-transcriptional changes that occurred in myocardium signal transduction pathways after MI. STUDY DESIGN/MATERIALS AND METHODS Continuous wave (CW) non-thermal laser parameters were: 660 nm wavelength, power 15 mW, with a total energy of 0.9 J, fluence of 1.15 J/cm2 , spot size of 0.785 cm2 , and time of 60 seconds. Using in silico analysis, we selected and then, quantified the expression of messenger RNA (mRNA) of 47 genes of 9 signaling pathways associated with MI (angiogenesis, cell survival, hypertrophy, oxidative stress, apoptosis, extracellular matrix, calcium kinetics, cell metabolism, and inflammation). Messenger RNA expression quantification was performed in myocardial samples by polymerase chain reaction real-time array using TaqMan customized plates. RESULTS Our results evidenced that MI modified mRNA expression of several well-known biomarkers related to detrimental cardiac activity in almost all signaling pathways analyzed. However, PBMT reverted most of these transcriptional changes. More expressively, PBMT provoked a robust decrease in mRNA expression of molecules that participate in post-MI inflammation and ECM composition, such as IL-6, TNF receptor, TGFb1, and collagen I and III. Global microRNA (miRNA) expression analysis revealed that PBMT decreased miR-221, miR-34c, and miR-93 expressions post-MI, which are related to deleterious effects in cardiac remodeling. CONCLUSION Thus, the identification of transcriptional and post-transcriptional changes induced by PBMT may be used to interfere in the molecular dynamics of cardiac remodeling post-MI.
Collapse
Affiliation(s)
| | | | | | - Andrey Jorge Serra
- Universidade Federal de São Paulo, Rua Pedro de Toledo 709, Vila Clementino, São Paulo, SP, 04039-001, Brazil
| | - Ednei Luiz Antônio
- Universidade Federal de São Paulo, Rua Pedro de Toledo 709, Vila Clementino, São Paulo, SP, 04039-001, Brazil
| | | | | | - Paulo José Ferreira Tucci
- Universidade Federal de São Paulo, Rua Pedro de Toledo 709, Vila Clementino, São Paulo, SP, 04039-001, Brazil
| | - Lubov Nathanson
- Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, Florida, 33314
| | - Mariana Morris
- Nova Southeastern University, 3301 College Avenue, Fort Lauderdale, Florida, 33314
| | | | | |
Collapse
|
5
|
Hsu CC, Huang CC, Chien LH, Lin MT, Chang CP, Lin HJ, Chio CC. Ischemia/reperfusion injured intestinal epithelial cells cause cortical neuron death by releasing exosomal microRNAs associated with apoptosis, necroptosis, and pyroptosis. Sci Rep 2020; 10:14409. [PMID: 32873851 PMCID: PMC7462997 DOI: 10.1038/s41598-020-71310-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 08/14/2020] [Indexed: 12/24/2022] Open
Abstract
To date, there is no good evidence that intestine epithelial cells (IEC) affected by ischemia/reperfusion (I/R) injury are able to cause cortical neuron injury directly. Additionally, it remains unclear whether the neuronal damage caused by I/R injured IEC can be affected by therapeutic hypothermia (TH, 32 °C). To address these questions, we performed an oxygen–glucose deprivation (OGD) affected IEC-6-primary cortical neuron coculture system under normothermia (37 °C) or TH (32 °C) conditions. It was found that OGD caused hyperpermeability in IEC-6 cell monolayers. OGD-preconditioned IEC-6 cells caused cortical neuronal death (e.g., decreased cell viability), synaptotoxicity, and neuronal apoptosis (evidenced by increased caspase-3 expression and the number of TUNEL-positive cells), necroptosis (evidenced by increased receptor-interacting serine/threonine-protein kinase-1 [RIPK1], RIPK3 and mixed lineage kinase domain-like pseudokinase [MLKL] expression), and pyroptosis (evidenced by an increase in caspase-1, gasdermin D [GSDMD], IL-1β, IL-18, the apoptosis-associated speck-like protein containing a caspase recruitment domain [ASC], and nucleotide oligomerization domain [NOD]-like receptor [NLRP]-1 expression). TH did not affect the intestinal epithelial hyperpermeability but did attenuate OGD-induced neuronal death and synaptotoxicity. We also performed quantitative real-time PCR to quantify the genes encoding 84 exosomal microRNAs in the medium of the control-IEC-6, the control-neuron, the OGD-IEC-6 at 37 °C, the OGD-IEC-6 at 32 °C, the neuron cocultured with OGD-IEC-6 at 37 °C, and the neurons cocultured with OGD-IEC-6 at 32 °C. We found that the control IEC-6 cell s or cortical neurons are able to secrete a basal level of exosomal miRNAs in their medium. OGD significantly up-regulated the basal level of each parameter for IEC-6 cells. As compared to those of the OGD-IEC-6 cells or the control neurons, the OGD-IEC-6 cocultured neurons had significantly higher levels of 19 exosomal miRNAs related to apoptosis, necroptosis, and/or pyroptosis events. Our results identify that I/R injured intestinal epithelium cells can induce cortical neuron death via releasing paracrine mediators such as exosomal miRNAs associated with apoptosis, necroptosis, and/or pyroptosis, which can be counteracted by TH.
Collapse
Affiliation(s)
- Chien-Chin Hsu
- Department of Biotechnology and Food Technology, Southern Taiwan University of Science and Technology, No. 1, Nan-Tai Street, Yungkang District, Tainan City, 710, Taiwan.,Department of Emergency Medicine, Chi Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan
| | - Chien-Cheng Huang
- Department of Emergency Medicine, Chi Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan.,Department of Senior Services, Southern Taiwan University of Science and Technology, No. 1, Nan-Tai Street, Yungkang District, Tainan City, 710, Taiwan.,Department of Environmental and Occupational Health, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City, 710, Taiwan.,Department of Geriatrics and Gerontology, Chi-Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan.,Department of Occupational Medicine, Chi-Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan
| | - Lan-Hsiang Chien
- Department of Medical Research, Chi Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan
| | - Mao-Tsun Lin
- Department of Medical Research, Chi Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan
| | - Ching-Ping Chang
- Department of Medical Research, Chi Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan.
| | - Hung-Jung Lin
- Department of Emergency Medicine, Chi Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan. .,Department of Medicine, Taipei Medical University, No. 250 Wu-Hsing Street, Taipei City, 110, Taiwan.
| | - Chung-Ching Chio
- Division of Neurosurgery, Department of Surgery, Chi Mei Medical Center, No. 901, Zhonghua Road, Yongkang District, Tainan City, 710, Taiwan.
| |
Collapse
|
6
|
Hu G, Ma L, Dong F, Hu X, Liu S, Sun H. Inhibition of microRNA‑124‑3p protects against acute myocardial infarction by suppressing the apoptosis of cardiomyocytes. Mol Med Rep 2019; 20:3379-3387. [PMID: 31432169 DOI: 10.3892/mmr.2019.10565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 02/19/2019] [Indexed: 11/05/2022] Open
Abstract
The aims of the present study were to investigate the roles and underlying mechanisms of microRNA‑124‑3p (miR‑124‑3p) in the progression of acute myocardial infarction (AMI). The expression of miR‑124‑3p was determined via reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). TargetScan analysis and a luciferase reporter assay were conducted to reveal the association between miR‑124‑3p and nuclear factor κ‑light‑chain‑enhancer of activated B cells (NF‑κB) repressing factor (NKRF). To investigate the role of miR‑124‑3p in AMI, a cell model of myocardial hypoxic/ischemic injury was established by subjecting H9c2 cardiac cells to hypoxia for 48 h. The viability of cells was determined using an MTT assay, and cell apoptosis was analyzed by flow cytometry. Additionally, the expression levels of inflammatory factors [tumor necrosis factor‑α (TNF‑α), interleukin (IL)‑1β and IL‑6] were measured via ELISA. Furthermore, gene and protein expression levels were determined by performing RT‑qPCR and western blot analyses, respectively. It was revealed that the expression of miR‑124‑3p was significantly increased in the blood of patients with AMI and hypoxia‑treated H9c2 cells. Additionally, it was demonstrated that NKRF was a direct target of miR‑124‑3p. The hypoxia‑induced decrease in the viability of H9c2 cells and increase in cell apoptosis were eliminated by the downregulation of miR‑124‑3p. Furthermore, hypoxia significantly increased the levels of TNF‑α, IL‑1β and IL‑6, whereas miR‑124‑3p downregulation eliminated these effects. Downregulated expression of B‑cell lymphoma 2, pro‑caspase 3 and pro‑caspase 9 protein, and upregulated expression of cleaved caspases 3 and 9 was observed in hypoxic H9c2 cells; the altered expression of these proteins was suppressed by miR‑124‑3p inhibitor. Additionally, miR‑124‑3p inhibitor suppressed the hypoxia‑induced activation of the NF‑κB signaling pathway in H9c2 cells. Furthermore, it was demonstrated that the various effects of miR‑124‑3p inhibitor on H9c2 cells were eliminated by the small interfering RNA‑mediated downregulation of NKRF. In conclusion, the results of the present study indicated that miR‑124‑3p downregulation protected against AMI via inhibition of inflammatory responses and the apoptosis of cardiomyocytes by regulating the NKRF/NF‑κB pathway.
Collapse
Affiliation(s)
- Guangrong Hu
- Emergency Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Lingbo Ma
- Emergency Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Fei Dong
- Emergency Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Xiao Hu
- Emergency Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Sida Liu
- Emergency Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Hui Sun
- Emergency Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
7
|
Sun J, Yang J, Chi J, Ding X, Lv N. Identification of drug repurposing candidates based on a miRNA-mediated drug and pathway network for cardiac hypertrophy and acute myocardial infarction. Hum Genomics 2018; 12:52. [PMID: 30514363 PMCID: PMC6280539 DOI: 10.1186/s40246-018-0184-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/25/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Cardiac hypertrophy and acute myocardial infarction (AMI) are two common heart diseases worldwide. However, research is needed into the exact pathogenesis and effective treatment strategies for these diseases. Recently, microRNAs (miRNAs) have been suggested to regulate the pathological pathways of heart disease, indicating a potential role in novel treatments. RESULTS In our study, we constructed a miRNA-gene-drug network and analyzed its topological features. We also identified some significantly dysregulated miRNA-gene-drug triplets (MGDTs) in cardiac hypertrophy and AMI using a computational method. Then, we characterized the activity score profile features for MGDTs in cardiac hypertrophy and AMI. The functional analyses suggested that the genes in the network held special functions. We extracted an insulin-like growth factor 1 receptor-related subnetwork in cardiac hypertrophy and a vascular endothelial growth factor A-related subnetwork in AMI. Finally, we considered insulin-like growth factor 1 receptor and vascular endothelial growth factor A as two candidate drug targets by utilizing the cardiac hypertrophy and AMI pathways. CONCLUSION These results provide novel insights into the mechanisms and treatment of cardiac hypertrophy and AMI.
Collapse
Affiliation(s)
- Jiantao Sun
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang People’s Republic of China
| | - Jiemei Yang
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang People’s Republic of China
| | - Jing Chi
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang People’s Republic of China
| | - Xue Ding
- Department of Cardiology, the First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang People’s Republic of China
| | - Nan Lv
- Department of Obstetrics, the Second Affiliated Hospital, Harbin Medical University, 246 XueFu Road, Harbin, 150086 Heilongjiang People’s Republic of China
| |
Collapse
|
8
|
Zhang Z, Li H, Chen S, Li Y, Cui Z, Ma J. Knockdown of MicroRNA-122 Protects H9c2 Cardiomyocytes from Hypoxia-Induced Apoptosis and Promotes Autophagy. Med Sci Monit 2017; 23:4284-4290. [PMID: 28871076 PMCID: PMC5597036 DOI: 10.12659/msm.902936] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a severe disease causing heart failure and sudden death. Studies indicate that microRNAs (miRNAs) are involved in the pathophysiology of AMI. In the present study, we carefully explored the effects of miR-122 on myocardial hypoxia injury and its possible underlying mechanism. MATERIAL AND METHODS miR-122 expression was analyzed in H9c2 cardiomyocytes after being transfected with miR-122 mimic, ASO-miR-122, or negative control. Cell viability and apoptosis were investigated by CCK-8 assays and flow cytometry analysis, respectively. Cell migration was analyzed using wound-healing assays. Western blotting was performed to analyze the expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN)/phosphatidylinositol 3-hydroxy kinase (PI3K)/AKT and LC3-II/LC3-I. RESULTS Hypoxia exposure significantly inhibited H9c2 cell viability (P<0.01). miR-122 overexpression promoted the hypoxia-induced H9c2 cell proliferation and migration loss (P<0.05), and cell apoptosis was increased (P<0.05). miR-122 knockdown enhanced cell viability and decreased cell apoptosis (P<0.05). Knockdown of miR-122 enhanced PTEN/PI3K/AKT activation and cell autophagy. Overexpression of miR-122 inhibited the PTEN/PI3K/AKT pathway and cell autophagy pathway. CONCLUSIONS The expression of miR-122 is involved in hypoxia-induced H9c2 cardiomyocyte injury. Knockdown of miR-122 protects H9c2 cells from hypoxia-induced apoptosis and enhances cell viability.
Collapse
Affiliation(s)
- Zaiwei Zhang
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Hu Li
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Shasha Chen
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Ying Li
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Zhiyuan Cui
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Jie Ma
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|