151
|
Prevalence of Moderate to Severe Anxiety Symptoms among Patients with Myocardial Infarction: a Meta-Analysis. Psychiatr Q 2022; 93:161-180. [PMID: 34013389 DOI: 10.1007/s11126-021-09921-y] [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] [Accepted: 04/25/2021] [Indexed: 10/21/2022]
Abstract
This study attempted to synthesize the evidence on the prevalence of moderate to severe anxiety symptoms among myocardial infarction (MI) patients to offer a reliable and accurate estimate on the number of MI patients suffering from moderate to severe anxiety symptoms. Comprehensive electronic searches (PubMed, Embase and Web of Science) were performed from their inception to February 2021. Between-study heterogeneity was analyzed using the Cochran's Q test and [Formula: see text] statistic, and if it was high across the eligible studies, meta-regression and subgroup analyses were conducted to examine the source of heterogeneity. Publication bias and the robustness of the pooled results were also examined. A total of 18 eligible studies covering 8,532 MI patients were included, of which 3,443 were identified with moderate to severe anxiety symptoms. Between-study heterogeneity was high ([Formula: see text]=98.8%) with the reported prevalence ranging from 9.6% to 69.17%, and the pooled prevalence was 38.08% (95% confidence interval: 28.82-47.81%) by a random-effects model. Meta-regression analyses indicated that publication year (β = -0.014) was significant moderators contributing 16.11% to the heterogeneity. Subgroup analyses indicated that studies using the anxiety subscale of Brief Symptom Inventory to assess anxiety were homogenous ([Formula: see text]=0.0). Furthermore, the pooled prevalence of moderate to severe anxiety symptoms varied significantly by geographic region, instrument used to assess anxiety, methodological quality, sex, education level, a history of previous MI and hypercholesterolemia. Additionally, the results of Egger's linear test (t = -0.630) and Begg's rank test (z = -0.190) indicated no evidence of publication bias, and the sensitivity of the pooled results was low. Nearly two fifth of MI patients suffered from moderate to severe anxiety symptoms, which emphasizes the importance of early identification of anxiety symptoms after MI, as well as the need of implementing psychological interventions for those with elevated anxiety symptoms.
Collapse
|
152
|
Veldhuizen J, Chavan R, Moghadas B, Park JG, Kodibagkar VD, Migrino RQ, Nikkhah M. Cardiac ischemia on-a-chip to investigate cellular and molecular response of myocardial tissue under hypoxia. Biomaterials 2022; 281:121336. [PMID: 35026670 PMCID: PMC10440189 DOI: 10.1016/j.biomaterials.2021.121336] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 12/18/2021] [Accepted: 12/24/2021] [Indexed: 12/31/2022]
Abstract
Tissue engineering has enabled the development of advanced and physiologically relevant models of cardiovascular diseases, with advantages over conventional 2D in vitro assays. We have previously demonstrated development of a heart on-a-chip microfluidic model with mature 3D anisotropic tissue formation that incorporates both stem cell-derived cardiomyocytes and cardiac fibroblasts within a collagen-based hydrogel. Using this platform, we herein present a model of myocardial ischemia on-a-chip, that recapitulates ischemic insult through exposure of mature 3D cardiac tissues to hypoxic environments. We report extensive validation and molecular-level analyses of the model in its ability to recapitulate myocardial ischemia in response to hypoxia, demonstrating the 1) induction of tissue fibrosis through upregulation of contractile fibers, 2) dysregulation in tissue contraction through functional assessment, 3) upregulation of hypoxia-response genes and downregulation of contractile-specific genes through targeted qPCR, and 4) transcriptomic pathway regulation of hypoxic tissues. Further, we investigated the complex response of ischemic myocardial tissues to reperfusion, identifying 5) cell toxicity, 6) sustained contractile irregularities, as well as 7) re-establishment of lactate levels and 8) gene expression, in hypoxic tissues in response to ischemia reperfusion injury.
Collapse
Affiliation(s)
- Jaimeson Veldhuizen
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | - Ramani Chavan
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Babak Moghadas
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | - Jin G Park
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Vikram D Kodibagkar
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, 85012, USA; University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering (SBHSE), Arizona State University, Tempe, AZ, 85287, USA; Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
153
|
Zhang Y, Hua W, Dang Y, Cheng Y, Wang J, Zhang X, Teng M, Wang S, Zhang M, Kong Z, Lu X, Zheng Y. Validated Impacts of N6-Methyladenosine Methylated mRNAs on Apoptosis and Angiogenesis in Myocardial Infarction Based on MeRIP-Seq Analysis. Front Mol Biosci 2022; 8:789923. [PMID: 35155564 PMCID: PMC8831860 DOI: 10.3389/fmolb.2021.789923] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/26/2021] [Indexed: 12/13/2022] Open
Abstract
Objectives: N6-methyladenosine (m6A) is hypothesized to play a role in the regulation of pathogenesis of myocardial infarction (MI). This study was designed to compare m6A-tagged transcript profiles to identify mRNA-specific changes on pathophysiological variations after MI. Methods: N6-methyladenosine methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were interacted to select m6A-modified mRNAs with samples collected from sham operated and MI rat models. m6A methylation regulated mRNAs were interacted with apoptosis/angiogenesis related genes in GeneCards. Afterwards, MeRIP-quantitative real-time PCR (MeRIP-qRT-PCR) was performed to measure m6A methylation level of hub mRNAs. m6A methylation variation was tested under different oxygen concentration or hypoxic duration in H9c2 cells and HUVECs. In addition, Western blot and qRT-PCR were employed to detect expression of hub mRNAs and relevant protein level. Flow cytometry and Tunel assay were conducted to assess apoptotic level. CCK-8, EdU, and tube formation assay were performed to measure cell proliferation and tube formation ability. Results: Upregulation of Mettl3 was firstly observed in vivo and in vitro, followed by upregulation of m6A methylation level. A total of 567 significantly changed m6A methylation peaks were identified, including 276 upregulated and 291 downregulated peaks. A total of 576 mRNAs were upregulated and 78 were downregulated. According to combined analysis of MeRIP-seq and RNA-seq, we identified 26 significantly hypermethylated and downregulated mRNAs. Based on qRT-PCR and interactive analysis, Hadh, Kcnn1, and Tet1 were preliminarily identified as hub mRNAs associated with apoptosis/angiogenesis. MeRIP-qRT-PCR assay confirmed the results from MeRIP-seq. With the inhibition of Mettl3 in H9c2 cells and HUVECs, downregulated m6A methylation level of total RNA and upregulated expression of hub mRNAs were observed. Increased m6A level was verified in the gradient context in terms of prolonged hypoxic duration and decreased oxygen concentration. Under simulated hypoxia, roles of Kcnn1 and Tet1 in angiogenesis and Hadh, Tet1, and Kcnn1 in apoptosis were further confirmed with our validation experiments. Conclusion: Roles of m6A-modified mRNA transcripts in the context of MI were preliminarily verified. In the context of m6A methylation, three hub mRNAs were validated to impact the process of apoptosis/angiogenesis. Our study provided theoretical basis and innovative targets for treatment of MI and paved the way for future investigations aiming at exploring upstream epigenetic mechanisms of pathogenesis after MI.
Collapse
Affiliation(s)
- Yingjie Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenjie Hua
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yini Dang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yihui Cheng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiayue Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiu Zhang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meiling Teng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shenrui Wang
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Min Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zihao Kong
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao Lu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Yu Zheng, ; Xiao Lu,
| | - Yu Zheng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Yu Zheng, ; Xiao Lu,
| |
Collapse
|
154
|
PCSK9 Promotes Cardiovascular Diseases: Recent Evidence about Its Association with Platelet Activation-Induced Myocardial Infarction. Life (Basel) 2022; 12:life12020190. [PMID: 35207479 PMCID: PMC8875594 DOI: 10.3390/life12020190] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/22/2022] [Accepted: 01/26/2022] [Indexed: 12/22/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, with the majority of the cases being heart failure due to myocardial infarction. Research on cardiovascular diseases is currently underway, particularly on atherosclerosis prevention, to reduce the risk of myocardial infarction. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has been reported to play a role in lipid metabolism, by enhancing low-density lipoprotein (LDL) receptor degradation. Therefore, PCSK9 inhibitors have been developed and found to successfully decrease LDL plasma levels. Recent experimental studies have also implicated PCSK9 in platelet activation, having a key role during atherosclerosis progression. Although numerous studies have addressed the role of PCSK9 role in controlling hypercholesterolemia, studies and discussions exploring its involvement in platelet activation are still limited. Hence, here, we address our current understanding of the pathophysiological process involved in atherosclerosis-induced myocardial infarction (MI) through platelet activation and highlight the molecular mechanisms used by PCSK9 in regulating platelet activation. Undoubtedly, a deeper understanding of the relationship between platelet activation and the underlying molecular mechanisms of PCSK9 in the context of MI progression will provide a new strategy for developing drugs that selectively inhibit the most relevant pathways in cardiovascular disease progression.
Collapse
|
155
|
GAO J, MENG C, GUAN L, ZHANG H, ZHANG W. Astragaloside IV promotes cardiac remodeling after myocardial infarction by inhibiting DNMT3B-mediated Runx3 methylation via downregulating LncRNA MIRT1 expression. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.44721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Jing GAO
- The First Affiliated Hospital of Kangda College of Nanjing Medical University, China
| | - Chunming MENG
- The First Affiliated Hospital of Kangda College of Nanjing Medical University, China
| | - Li GUAN
- The First Affiliated Hospital of Kangda College of Nanjing Medical University, China
| | | | - Wei ZHANG
- Navy Qingdao Special Service Convalescent Center,, China
| |
Collapse
|
156
|
Wang Y, Shen Y. Exosomal miR-455-3p from BMMSCs prevents cardiac ischemia-reperfusion injury. Hum Exp Toxicol 2022; 41:9603271221102508. [PMID: 35577544 DOI: 10.1177/09603271221102508] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Bone marrow mesenchymal stem cells (BMMSCs) exert protective effects against myocardial infarction (MI). Here, we focused on the function and mechanism of miR-455-3p from BMMSCs-derived exosomes (BMMSCs-Exo) in myocardial infarction. MATERIALS AND METHODS BMMSCs were isolated from rat bone marrow, and the exosomes from the culture medium of BMMSCs were separated, and administered to H9C2 cells under hypoxia-reperfusion (H/R) stimulation. MTT and TUNEL staining analyzed cell viability and apoptosis, respectively. RT-qPCR determined miR-455-3p expression. Apoptosis-related proteins, autophagy-associated proteins, and the MEKK1-MKK4-JNK signaling pathway were detected. The interaction between miR-455-3p and MEKK1 was confirmed through dual luciferase activity and RIP assay. An in vivo ischemia reperfusion (I/R) model was established in rats. 2, 3, 5 triphenyltetrazolium chloride (TTC) staining, hematoxylin-eosin (H&E) staining, Masson staining, and TUNEL staining evaluated the infarct volume and histopathological changes. RESULTS miR-455-3p's expression was down-regulated in BMMSCs-derived exosomes, I/R myocardial tissues, and H/R myocardial cells. miR-455-3p enriched by BMMSC exosomes reduced H/R-mediated cardiomyocyte damage and death-related autophagy. miR-455-3p upregulation suppressed MEKK1-MKK4-JNK. MEKK1 overexpression notably mitigated cell apoptosis, cramped cell viability, suppressed autophagy expansion, and attenuated Exo-miR-455-3p's protection on H/R myocardial cells. In-vivo trials reflected that BMMSC exosomes enriched with miR-455-3p repressed ischemia reperfusion-induced myocardial damage and myocardial cell function. CONCLUSION miR-455-3p, shuttled by exosomes from MSCs, targets the MEKK1-MKK4-JNK signaling pathway to guard against myocardial ischemia-reperfusion damage.
Collapse
Affiliation(s)
- Yue Wang
- Department of Cardiology, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, China
| | - Yusi Shen
- Second Department of Orthopedic Rehabilitation, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
157
|
Peres Valgas da Silva C, Shettigar VK, Baer LA, Abay E, Madaris KL, Mehling MR, Hernandez-Saavedra D, Pinckard KM, Seculov NP, Ziolo MT, Stanford KI. Brown adipose tissue prevents glucose intolerance and cardiac remodeling in high-fat-fed mice after a mild myocardial infarction. Int J Obes (Lond) 2022; 46:350-358. [PMID: 34716427 PMCID: PMC8794788 DOI: 10.1038/s41366-021-00999-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Obesity increases the risk of developing impaired glucose tolerance (IGT) and type 2 diabetes (T2D) after myocardial infarction (MI). Brown adipose tissue (BAT) is important to combat obesity and T2D, and increasing BAT mass by transplantation improves glucose metabolism and cardiac function. The objective of this study was to determine if BAT had a protective effect on glucose tolerance and cardiac function in high-fat diet (HFD) fed mice subjected to a mild MI. METHODS Male C57BL/6 mice were fed a HFD for eight weeks and then divided into Sham (Sham-operated) and +BAT (mice receiving 0.1 g BAT into their visceral cavity). Sixteen weeks post-transplantation, mice were further subdivided into ±MI (Sham; Sham-MI; +BAT; +BAT-MI) and maintained on a HFD. Cardiac (echocardiography) and metabolic function (glucose and insulin tolerance tests, body composition and exercise tolerance) were assessed throughout 22 weeks post-MI. Quantitative PCR (qPCR) was performed to determine the expression of genes related to metabolic function of perigonadal adipose tissue (pgWAT), subcutaneous white adipose tissue (scWAT), liver, heart, tibialis anterior skeletal muscle (TA); and BAT. RESULTS +BAT prevented the increase in left ventricle mass (LVM) and exercise intolerance in response to MI. Similar to what is observed in humans, Sham-MI mice developed IGT post-MI, but this was negated in +BAT-MI mice. IGT was independent of changes in body composition. Genes involved in inflammation, insulin resistance, and metabolism were significantly altered in pgWAT, scWAT, and liver in Sham-MI mice compared to all other groups. CONCLUSIONS BAT transplantation prevents IGT, the increase in LVM, and exercise intolerance following MI. MI alters the expression of several metabolic-related genes in WAT and liver in Sham-MI mice, suggesting that these tissues may contribute to the impaired metabolic response. Increasing BAT may be an important intervention to prevent the development of IGT or T2D and cardiac remodeling in obese patients post-MI.
Collapse
Affiliation(s)
- Carmem Peres Valgas da Silva
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Vikram K. Shettigar
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Lisa A. Baer
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Eaman Abay
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Kendra L. Madaris
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Mikayla R. Mehling
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Diego Hernandez-Saavedra
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Kelsey M. Pinckard
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Nickolai P. Seculov
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA
| | - Mark T. Ziolo
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH USA
| | - Kristin I. Stanford
- grid.412332.50000 0001 1545 0811Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH USA
| |
Collapse
|
158
|
Sane R, Mandole R, Amin G. Use of reverse diet kit as a treatment to regress atheroma in a known CAD patient: A case report. J Ayurveda Integr Med 2021; 13:100511. [PMID: 34973885 PMCID: PMC8814396 DOI: 10.1016/j.jaim.2021.08.004] [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: 12/03/2020] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 11/28/2022] Open
Abstract
The way people eat have changed rapidly across the globe and is one of the reasons for developing coronary artery disease (CAD). Atheroma is caused due to the accumulation of fatty deposits and scar tissue leading to the degeneration of the walls of the arteries, restriction of the circulation and a risk of thrombosis. This single case experimental study shows that regression in atheroma may be achieved by replacing the normal diet with a low calories diet used in the reverse diet kit. The patient complained of angina on minor exertion. Post admission the patient underwent a computed tomography angiogram (CT-angiogram) to measure the plaque volumes of left anterior descending artery (LAD), left circumflex artery (LXC) and right coronary artery (RCA) of the heart. The patient was monitored for 12 weeks with the administration of reverse diet as a treatment method. The change in weight, blood pressure and heart rate was monitored every week. After 12 weeks, CT-angiogram was performed again. There was a 7.3 kg decrease in weight along with normalization of blood pressure (BP). A 92.8 mm3 regression of the total atheroma volume was observed. The diet provided may help in reducing atheroma burden in this CAD patient.
Collapse
Affiliation(s)
- Rohit Sane
- Madhavbaug Cardiac Hospitals and Clinics, India
| | - Rahul Mandole
- Department of Research Development, Madhavbaug Cardiac Clinic and Hospital, Thane, Maharashtra, India.
| | - Gurudatta Amin
- Madhavbaug Cardiac Clinic and Hospital, Thane, Maharashtra, India
| |
Collapse
|
159
|
Yang Y, Shi X, Du Z, Zhou G, Zhang X. Associations between genetic variations in microRNA and myocardial infarction susceptibility: a meta-analysis and systematic review. Herz 2021; 47:524-535. [PMID: 34878577 DOI: 10.1007/s00059-021-05086-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/01/2021] [Accepted: 11/07/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Current genetic association studies have reported conflicting results regarding the association between miRNA polymorphisms and myocardial infarction (MI) risk METHODS: Relevant studies were retrieved from the PubMed, EMBASE, ISI Web of Science, and Scopus databases. Eligible studies determining the association between miRNA polymorphisms and MI susceptibility were included and a meta-analysis was performed to quantify the associations between miRNA polymorphisms and MI risk. RESULTS A total of eight studies with 2507 MI patients and 3796 healthy controls were included, dealing with nine miRNA genes containing 11 different loci, including miR-149 (rs71428439 and rs2292832), miR-126 (rs4636297 and rs1140713), miR-146a (rs2910164), miR-218 (rs11134527), miR-196a2 (rs11614913), miR-499 (rs3746444), miR-27a (rs895819), miR-26a‑1 (rs7372209), and miR-100 (rs1834306). miR-146a rs2910164 and miR-499 rs3746444 were determined to have a significant association with MI susceptibility, a finding that was supported by the meta-analysis (rs2910164: GG/CC, odds ratio [OR]: 1.40, 95% confidence interval [95% CI]: 1.05-1.74, p < 0.001; rs3746444: AA + AG/GG, OR = 2.04, 95% CI: 1.37-2.70, p < 0.001). Limited or conflicting data were found for the relationship between the other miRNA polymorphisms (rs71428439, rs4636297, rs1140713, rs11134527, rs11614913, rs895819, rs7372209, rs1834306, rs2292832) and MI risk. CONCLUSION There was a significant association between rs2910164 and rs3746444 and MI susceptibility. Further studies are required to investigate the role of miRNA polymorphisms in MI risk.
Collapse
Affiliation(s)
- Yang Yang
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University, 230061, Hefei, Anhui, China
| | - Xiajun Shi
- Department of Cardiology, Tongling People's Hospital, 244002, Tongling, Anhui, China
| | - Zhengxun Du
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University, 230061, Hefei, Anhui, China
| | - Gendong Zhou
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University, 230061, Hefei, Anhui, China
| | - Xiaohong Zhang
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University, 230061, Hefei, Anhui, China.
| |
Collapse
|
160
|
Li Y, Qu M, Xing F, Li H, Cheng D, Xing N, Zhang W. The Protective Mechanism of Dexmedetomidine in Regulating Atg14L-Beclin1-Vps34 Complex Against Myocardial Ischemia-Reperfusion Injury. J Cardiovasc Transl Res 2021; 14:1063-1074. [PMID: 33914271 DOI: 10.1007/s12265-021-10125-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/28/2021] [Indexed: 02/06/2023]
Abstract
The blood flow restoration of ischemic tissues causes myocardial injury. Dexmedetomidine (Dex) protects multi-organs against ischemia/reperfusion (I/R) injury. This study investigated the protective mechanism of Dex post-treatment in myocardial I/R injury. The rat model of myocardial I/R was established. The effects of Dex post-treatment on cardiac function and autophagy flow were observed. Dex attenuated myocardial I/R injury and reduced I/R-induced autophagy in rats. Dex weakened the interactions between Beclin1 and Vps34 and Beclin1 and Atg14L, thus downregulating Vps34 kinase activity. In vitro, the cardiomyocytes subjected to oxygen glucose deprivation/reoxygenation were treated with Dex and PI3K inhibitor LY294002. LY294002 attenuated the myocardial protective effect of DEX, indicating that Dex protected against cardiac I/R by activating the PI3K/Akt pathway. In conclusion, Dex upregulated the phosphorylation of Beclin1 at S295 site by activating the PI3K/Akt pathway and reduced the interactions of Atg14L-Beclin1-Vps34 complex, thus inhibiting autophagy and protecting against myocardial I/R injury.
Collapse
Affiliation(s)
- Yanna Li
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Mingcui Qu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Fei Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Huixin Li
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Dan Cheng
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China
| | - Na Xing
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China.
| | - Wei Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou, 450000, China.
| |
Collapse
|
161
|
Sayed-Pathan NI, Kumar P, Paknikar KM, Gajbhiye V. MicroRNAs: A Neoteric Approach to Understand Pathogenesis, Diagnose, and Treat Myocardial Infarction. J Cardiovasc Pharmacol 2021; 78:773-781. [PMID: 34882110 DOI: 10.1097/fjc.0000000000001141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 09/12/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Myocardial infarction is a substantial contributor to ischemic heart diseases, affecting a large number of people leading to fatal conditions worldwide. MicroRNAs (miRNAs) are explicitly emerging as excellent modulators of pathways involved in maintaining cardiomyocyte survival, repair, and regeneration. Altered expression of genes in cardiomyocytes postinfarction can lead to the disordered state of the myocardium, such as cardiac hypertrophy, ischemia-reperfusion injury, left ventricular remodeling, and cardiac fibrosis. Therapeutic targeting of miRNAs in cardiomyocytes can potentially reverse the adverse effects in the heart postinfarction. This review aims to understand the role of several miRNAs involved in the regeneration and repair of cardiomyocytes postmyocardial infarction and presents comprehensive information on the subject.
Collapse
Affiliation(s)
- Nida Irfan Sayed-Pathan
- Nanobioscience Group, Agharkar Research Institute, Pune, India; and
- Savitribai Phule Pune University, Ganeshkhind, Pune, India
| | - Pramod Kumar
- Nanobioscience Group, Agharkar Research Institute, Pune, India; and
- Savitribai Phule Pune University, Ganeshkhind, Pune, India
| | - Kishore M Paknikar
- Nanobioscience Group, Agharkar Research Institute, Pune, India; and
- Savitribai Phule Pune University, Ganeshkhind, Pune, India
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, Pune, India; and
- Savitribai Phule Pune University, Ganeshkhind, Pune, India
| |
Collapse
|
162
|
Shi X, Cao Y, Zhang X, Gu C, Liang F, Xue J, Ni HW, Wang Z, Li Y, Wang X, Cai Z, Hocher B, Shen LH, He B. Comprehensive Analysis of N6-Methyladenosine RNA Methylation Regulators Expression Identify Distinct Molecular Subtypes of Myocardial Infarction. Front Cell Dev Biol 2021; 9:756483. [PMID: 34778266 PMCID: PMC8578940 DOI: 10.3389/fcell.2021.756483] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/13/2021] [Indexed: 01/17/2023] Open
Abstract
Background: Myocardial infarction (MI) is one of the leading threats to human health. N6-methyladenosine (m6A) modification, as a pivotal regulator of messenger RNA stability, protein expression, and cellular processes, exhibits important roles in the development of cardiac remodeling and cardiomyocyte contractile function. Methods: The expression levels of m6A regulators were analyzed using the GSE5406 database. We analyzed genome-wide association study data and single-cell sequencing data to confirm the functional importance of m6A regulators in MI. Three molecular subtypes with different clinical characteristics were established to tailor treatment strategies for patients with MI. We applied pathway analysis and differentially expressed gene (DEG) analysis to study the changes in gene expression and identified four common DEGs. Furthermore, we constructed the protein–protein interaction network and confirmed several hub genes in three clusters of MI. To lucubrate the potential functions, we performed a ClueGO analysis of these hub networks. Results: In this study, we identified that the levels of FTO, YTHDF3, ZC3H13, and WTAP were dramatically differently expressed in MI tissues compared with controls. Bioinformatics analysis showed that DEGs in MI were significantly related to modulating calcium signaling and chemokine signaling, and m6A regulators were related to regulating glucose measurement and elevated blood glucose levels. Furthermore, genome-wide association study data analysis showed that WTAP single-nucleotide polymorphism was significantly related to the progression of MI. In addition, single-cell sequencing found that WTAP is widely expressed in the heart tissues. Moreover, we conducted consensus clustering for MI in view of the dysregulated m6A regulators’ expression in MI. According to the expression levels, we found MI patients could be clustered into three subtypes. Pathway analysis showed the DEGs among different clusters in MI were assigned to HIF-1, IL-17, MAPK, PI3K-Akt signaling pathways, etc. The module analysis detected several genes, including BAG2, BAG3, MMP2, etc. We also found that MI-related network was significantly related to positive and negative regulation of angiogenesis and response to heat. The hub networks in MI clusters were significantly related to antigen processing and ubiquitin-mediated proteolysis, RNA splicing, and stability, indicating that these processes may contribute to the development of MI. Conclusion: Collectively, our study could provide more information for understanding the roles of m6A in MI, which may provide a novel insight into identifying biomarkers for MI treatment and diagnosis.
Collapse
Affiliation(s)
- Xin Shi
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yaochen Cao
- Department of Nephrology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Xiaobin Zhang
- Department of Cardiovascular Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chang Gu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Liang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jieyuan Xue
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Han-Wen Ni
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zi Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Li
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xia Wang
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhaohua Cai
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Berthold Hocher
- 5th Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Rheumatology), University Hospital Mannheim, University of Heidelberg, Mannheim, Germany
| | - Ling-Hong Shen
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ben He
- Department of Cardiology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
163
|
Chen Z, Yan Y, Qi C, Liu J, Li L, Wang J. The Role of Ferroptosis in Cardiovascular Disease and Its Therapeutic Significance. Front Cardiovasc Med 2021; 8:733229. [PMID: 34765653 PMCID: PMC8576275 DOI: 10.3389/fcvm.2021.733229] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/17/2021] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of deaths worldwide with regulated cell death playing an important role in cardiac pathophysiology. However, the classical mode of cell death cannot fully explain the occurrence and development of heart disease. In recent years, much research has been performed on ferroptosis, a new type of cell death that causes cell damage and contributes to the development of atherosclerosis, myocardial infarction, heart failure, and other diseases. In this review, we discuss the role of different organelles in ferroptosis and also focus on the relationship between autophagy and ferroptosis. Additionally, we describe the specific mechanism by which ferroptosis contributes to the development of CVD. Finally, we summarize the current research on ferroptosis-related pathway inhibitors and the applications of clinically beneficial cardiovascular drugs.
Collapse
Affiliation(s)
- Zhenzhen Chen
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Youyou Yan
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Chao Qi
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Jia Liu
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Longbo Li
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
164
|
Dundas JA, Hassanabad AF, Zarzycki AN, Fedak PW, Deniset JF. Ischemic heart disease: Cellular and molecular immune contributions of the pericardium. Int J Biochem Cell Biol 2021; 140:106076. [PMID: 34543760 DOI: 10.1016/j.biocel.2021.106076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/30/2021] [Accepted: 09/07/2021] [Indexed: 11/20/2022]
Abstract
Ischemic heart disease promotes complex inflammatory and remodeling pathways which contribute to the development of chronic heart failure. Although blood-derived and local cardiac mediators have traditionally been linked with these processes, the pericardial space has more recently been noted as alternative contributor to the injury response in the heart. The pericardial space contains fluid rich in physiologically active mediators, and immunologically active adipose tissue, which are altered during myocardial infarction. Key immune cells in the pericardial fluid and adipose tissue have been identified which act as mediators for cell recruitment and function after myocardial infarction have been identified in experimental models. Here, we provide an overview of the current understanding of the inflammatory mechanisms of the pericardial space and their role in post-myocardial infarction remodeling and the potential for the use of the pericardial space as a delivery vehicle for treatments to modulate heart healing.
Collapse
Affiliation(s)
- Jameson A Dundas
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Ali Fatehi Hassanabad
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Anna N Zarzycki
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Paul Wm Fedak
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, Alberta, Canada
| | - Justin F Deniset
- Section of Cardiac Surgery, Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, Calgary, Alberta, Canada; Department of Pharmacology and Physiology, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
165
|
Liu P, Huang J, Mei W, Zeng X, Wang C, Wen C, Xu J. Epigallocatechin-3-gallate protects cardiomyocytes from hypoxia-reoxygenation damage via raising autophagy related 4C expression. Bioengineered 2021; 12:9496-9506. [PMID: 34699312 PMCID: PMC8810140 DOI: 10.1080/21655979.2021.1996018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Myocardial ischemia/reperfusion (I/R) injury is a serious issue during the therapy of myocardial infarction. Herein, we explored the beneficial influence of Epigallocatechin-3-gallate (EGCG) on hypoxia/reoxygenation (H/R)-stimulated cardiomyocyte H9c2 cells damage, along with possible internal molecular mechanism related autophagy related 4C (ATG4C). H9c2 cells were subjected to H/R stimulation and/or EGCG treatment. ATG4C mRNA expression was measured via q-PCR assay. ATG4C overexpression plasmid (OE-ATG4C) was transfected to arise ATG4C level. Cell viability, apoptosis, reactive oxygen species (ROS) production, ATP level were tested via CCK-8 assay, Annexin V-FITC/PI staining, DCFH-DA staining and ATP Assay Kit, respectively. Western blotting was performed to test Cleaved-caspase 3, Cleaved-caspase 9, cytochrome C, and LC3B protein levels. H/R stimulation resulted in H9c2 cell viability loss, promoted cell apoptosis, and ROS overproduction, as well as lowered ATP level in cells. EGCG treatment alleviated H/R-resulted H9c2 cell viability loss, cell apoptosis, ROS overproduction, and reduction of ATP level. Moreover, H/R stimulation reduced the ATG4C expression in H9c2 cells, while EGCG raised the ATG4C expression. Overexpression of ATG4C strengthened the beneficial influence of EGCG on H/R-stimulated H9c2 cell viability, apoptosis and ROS production. Besides, ATG4C overexpression weakened the H/R-stimulated H9c2 cell autophagy via reducing LC3B II/I expression. EGCG exerted beneficial influence on H/R-stimulated cardiomyocytes, which protected cardiomyocytes from H/R-stimulated viability loss, apoptosis, and ROS overproduction via enhancing ATG4C expression.
Collapse
Affiliation(s)
- Ping Liu
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatric Neurology and Cardiovasology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jin Huang
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wanzhen Mei
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatric Neurology and Cardiovasology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xingfang Zeng
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatric Neurology and Cardiovasology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Cheng Wang
- Department of Pediatric Neurology and Cardiovasology, Children's Medical Center, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chuan Wen
- Department of Pediatric Hematology and Oncology, Children's Medical Center, the Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Xu
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.,Department of Pediatric Hematology and Oncology, Children's Medical Center, the Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
166
|
Engineering Cardiac Small Extracellular Vesicle-Derived Vehicles with Thin-Film Hydration for Customized microRNA Loading. J Cardiovasc Dev Dis 2021; 8:jcdd8110135. [PMID: 34821688 PMCID: PMC8626043 DOI: 10.3390/jcdd8110135] [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: 08/31/2021] [Revised: 09/27/2021] [Accepted: 10/19/2021] [Indexed: 11/17/2022] Open
Abstract
Cell therapies for myocardial infarction, including cardiac ckit+ progenitor cell (CPC) therapies, have been promising, with clinical trials underway. Recently, paracrine signaling, specifically through small extracellular vesicle (sEV) release, was implicated in cell-based cardiac repair. sEVs carry cardioprotective cargo, including microRNA (miRNA), within a complex membrane and improve cardiac outcomes similar to that of their parent cells. However, miRNA loading efficiency is low, and sEV yield and cargo composition vary with parent cell conditions, minimizing sEV potency. Synthetic mimics allow for cargo-loading control but consist of much simpler membranes, often suffering from high immunogenicity and poor stability. Here, we aim to combine the benefits of sEVs and synthetic mimics to develop sEV-like vesicles (ELVs) with customized cargo loading. We developed a modified thin-film hydration (TFH) mechanism to engineer ELVs from CPC-derived sEVs with pro-angiogenic miR-126 encapsulated. Characterization shows miR-126+ ELVs are similar in size and structure to sEVs. Upon administration to cardiac endothelial cells (CECs), ELV uptake is similar to sEVs too. Further, when functionally validated with a CEC tube formation assay, ELVs significantly improve tube formation parameters compared to sEVs. This study shows TFH-ELVs synthesized from sEVs allow for select miRNA loading and can improve in vitro cardiac outcomes.
Collapse
|
167
|
Zhang L, Wang Y, Yu F, Li X, Gao H, Li P. CircHIPK3 Plays Vital Roles in Cardiovascular Disease. Front Cardiovasc Med 2021; 8:733248. [PMID: 34660735 PMCID: PMC8511503 DOI: 10.3389/fcvm.2021.733248] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/03/2021] [Indexed: 12/20/2022] Open
Abstract
Circular RNAs (circRNAs) are covalently closed RNAs that function in various physiological and pathological processes. CircRNAs are widely involved in the development of cardiovascular disease (CVD), one of the leading causes of morbidity and mortality worldwide. CircHIPK3 is generated from the second exon of the HIPK3 gene, a corepressor of homeodomain transcription factors. As an exonic circRNA (ecRNA), circHIPK3 is produced through intron-pairing driven circularization facilitated by Alu elements. In the past 5 years, a growing number of studies have revealed the multifunctional roles of circHIPK3 in different diseases, such as cancer and CVD. CircHIPK3 mainly participates in CVD pathogenesis through interacting with miRNAs. This paper summarizes the current literature on the biogenesis and functions of circHIPK3, elucidates the role of circHIPK3 in different CVD patterns, and explores future perspectives.
Collapse
Affiliation(s)
- Lei Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Fei Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xin Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Huijuan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|
168
|
Wang J, Zhang S, Di L. Acute myocardial infarction therapy: in vitro and in vivo evaluation of atrial natriuretic peptide and triphenylphosphonium dual ligands modified, baicalin-loaded nanoparticulate system. Drug Deliv 2021; 28:2198-2204. [PMID: 34662253 PMCID: PMC8525923 DOI: 10.1080/10717544.2021.1989086] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Background Myocardial infarction (MI) is one of the most common ischemic heart diseases. It is very essential to explore new types of cardioprotective drugs delivery systems in this area. Objective The aim of the present study was to investigate the protective effect of baicalin (BA) and puerarin (PU) against acute MI rat models. BA and PU co-loaded nanoparticulate system were developed to improve bioavailability of the drugs, to prolong retention time in vivo and to enhance the protective effect. Methods In the present study, ANP and TPP contained ligands were synthesized. ANP/TPP-BN-LPNs were prepared and its physico-chemical properties were evaluated. The MI therapy efficiency of ANP/TPP-BN-LPNs was assessed in rats after intravenous injection. Single ligand contained LPNs, no ligand contained LPNs, and BN solution formulations were also prepared and used for the comparison. Results ANP/TPP-BN-LPNs were uniform and spheroidal particles. The size of ANP/TPP-BN-LPNs was 98.5 ± 2.9 nm, with a zeta potential of –19.5 ± 1.9 mV. The dual ligands modified LPNs exhibited significantly improved therapeutic efficiency compared with the single ligand modified LPNs and other systems. In vivo infarct therapy studies in rats proved that ANP/TPP-BN-LPNs were a promising system for efficient delivery of cardiovascular drugs for the treatment of cardiovascular diseases. Conclusions ANP/TPP-BN-LPNs could be used as a long-circulating and heart-targeting drug delivery system.
Collapse
Affiliation(s)
- Jie Wang
- Intervention Center, Linyi People's Hospital Beicheng New District Hospital, Linyi, PR China
| | - Shouwen Zhang
- Cardiology Pacing and Electrophysiology Ward, Linyi People's Hospital Beicheng New District Hospital, Linyi, PR China
| | - Lizhe Di
- Oral Cavity Clinic, Linyi People's Hospital, Linyi, PR China
| |
Collapse
|
169
|
Huang LH, Liu LP, Chen FG, Hui YS. Two new Cd(II) coordination polymers: Luminescent properties and protective activity on ischemic myocardial infarction. J SOLID STATE CHEM 2021. [DOI: 10.1016/j.jssc.2021.122374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
170
|
Sun B, Zhao C, Mao Y. MiR-218-5p Mediates Myocardial Fibrosis after Myocardial Infarction by Targeting CX43. Curr Pharm Des 2021; 27:4504-4512. [PMID: 34587879 DOI: 10.2174/1381612827666210929111622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/21/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND Myocardial fibrosis after myocardial infarction (MI) has been considered a core factor in the deterioration of cardiac function. Previous studies have shown that miRNA plays an important role in various pathophysiological processes of the heart. However, the role of miRNA in myocardial fibrosis regulation after MI remains unclear. In the present study, we documented that miR-218-5p was significantly decreased in myocardial fibroblasts after MI. METHODS The miRNA expression profiles of MI were downloaded from GEO Datasets. The expression of a fibrosis-related gene in vivo and in vitro was analyzed by RT-PCR, western blotting, and immunohistochemical staining. RESULTS Total 7 up- and 9 downregulated common miRNAs were found in the two profiles. Among these common genes, miR-218-5p was downregulated in the MI mice. MiR-218-5p mediated the myocardial fibrosis in vivo and in vitro. Mechanistically, we found that GJA1 (CX43) may be the target of miR218-5p, and overexpressed CX43 can partly block the function of miR-218-5p in fibrosis inhibition. CONCLUSION Our results suggested that miR-218-5p plays an important role in myocardial fibrosis after MI by targeting CX43. Thus, miR-218-5p promises to be a potential diagnosis and treatment of myocardial fibrosis after MI.
Collapse
Affiliation(s)
- Bing Sun
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Cuimei Zhao
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| | - Yu Mao
- Department of Cardiology, Tongji Hospital Affiliated to Tongji University, Shanghai, China
| |
Collapse
|
171
|
Mayfield J, Gill M, Zhang HJ, Ganti L. Sex-Based Differences in the Presentation of Myocardial Infarction. Cureus 2021; 13:e16906. [PMID: 34513479 PMCID: PMC8418226 DOI: 10.7759/cureus.16906] [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] [Accepted: 08/05/2021] [Indexed: 11/22/2022] Open
Abstract
The authors report a case of a middle-aged female presenting with a chief complaint of shoulder pain. Workup revealed a non-ST-segment elevation myocardial infarction. The typical symptoms of myocardial infarction and the evaluation of a non-ST-segment elevation myocardial infarction are discussed. The authors highlight the sex-based differences in the presentation of myocardial infarction and remind us to keep a broad differential and consider atypical presentations.
Collapse
Affiliation(s)
- Jeremy Mayfield
- Emergency Medicine, HCA Healthcare Graduate Medical Education Consortium Emergency Medicine Residency Program of Greater Orlando, Orlando, USA.,Emergency Medicine, Osceola Regional Medical Center, Kissimmee, USA
| | - Muneet Gill
- Emergency Medicine, Brown University, Providence, USA
| | - Helen J Zhang
- Emergency Medicine, Brown University, Providence, USA
| | - Latha Ganti
- Emergency Medicine, Envision Physician Services, Plantation, USA.,Emergency Medicine, University of Central Florida College of Medicine, Orlando, USA.,Emergency Medicine, Osceola Regional Medical Center, Kissimmee, USA.,Emergency Medicine, HCA Healthcare Graduate Medical Education Consortium Emergency Medicine Residency Program of Greater Orlando, Orlando, USA
| |
Collapse
|
172
|
Trajković N, Đorđević D, Stanković M, Petrušič T, Bogataj Š, Peršič V. Exercise-Based Interventions in Middle-Aged and Older Adults after Myocardial Infarction: A Systematic Review. Life (Basel) 2021; 11:life11090928. [PMID: 34575077 PMCID: PMC8466422 DOI: 10.3390/life11090928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/30/2021] [Accepted: 09/04/2021] [Indexed: 11/16/2022] Open
Abstract
This study summarized the relevant literature and aimed to determine the effect of exercise-based interventions after myocardial infarction in middle-aged and older adults. Studies were identified and analyzed according to the PRISMA guidelines. The following electronic databases were used: Google Scholar, PubMed, Mendeley, Science Direct, and Scopus. The identified studies had to be longitudinal, be published in English, have taken place between 2010 and 2020, involve participants who had suffered myocardial infarction, and address different types of exercise-based interventions to be included. Of the 592 relevant studies identified, 20 were included in the qualitative analysis. After analyzing the results obtained, it could be concluded that different types of exercise-based interventions or their combination have a positive effect after myocardial infarction in middle-aged and elderly adults. It can also be suggested that the combination of a cycle ergometer and a treadmill may be the most effective exercise-based intervention. An adequate choice of intensity and volume is crucial, with the optimal duration of the experimental program and the type(s) of exercises adapted to participants after myocardial infarction.
Collapse
Affiliation(s)
- Nebojša Trajković
- Faculty of Sport and Physical Education, University of Niš, 18000 Niš, Serbia; (N.T.); (D.Đ.); (M.S.)
| | - Dušan Đorđević
- Faculty of Sport and Physical Education, University of Niš, 18000 Niš, Serbia; (N.T.); (D.Đ.); (M.S.)
| | - Mima Stanković
- Faculty of Sport and Physical Education, University of Niš, 18000 Niš, Serbia; (N.T.); (D.Đ.); (M.S.)
| | - Tanja Petrušič
- Faculty of Education, University of Ljubljana, 1000 Ljubljana, Slovenia;
| | - Špela Bogataj
- Department of Nephrology, University Medical Centre, 1000 Ljubljana, Slovenia;
- Faculty of Sport, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Vanja Peršič
- Department of Nephrology, University Medical Centre, 1000 Ljubljana, Slovenia;
- Correspondence:
| |
Collapse
|
173
|
Ji Z, Wang C, Tong Q. Role of miRNA-324-5p-Modified Adipose-Derived Stem Cells in Post-Myocardial Infarction Repair. Int J Stem Cells 2021; 14:298-309. [PMID: 34158416 PMCID: PMC8429947 DOI: 10.15283/ijsc21025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/26/2021] [Accepted: 04/30/2021] [Indexed: 11/09/2022] Open
Abstract
Background and Objectives To seek out the role of mircoRNA (miR)-324-5p-modified adipose-derived stem cells (ADSCs) in post-myocardial infarction (MI) myocardial repair. Methods and Results Rat ADSCs were cultivated and then identified by morphologic observation, osteogenesis and adipogenesis induction assays and flow cytometry. Afterwards, ADSCs were modified by miR-324-5p lentiviral vector, with ADSC proliferation and migration measured. Then, rat MI model was established, which was treated by ADSCs or miR-324-5p-modified ADSCs. Subsequently, the function of miR-324-5p-modified ADSCs in myocardial repair of MI rats was assessed through functional assays. Next, the binding relation of miR-324-5p and Toll-interacting protein (TOLLIP) was validated. Eventually, functional rescue assay of TOLLIP was performed to verify the role of TOLLIP in MI. First, rat ADSCs were harvested. Overexpressed miR-324-5p improved ADSC viability. ADSC transplantation moderately enhanced cardiac function of MI rats, reduced enzyme levels and decreased infarct size and apoptosis; while miR-324-5p-modified ADSCs could better promote post-MI repair. Mechanically, miR-324-5p targeted TOLLIP in myocardial tissues. Moreover, TOLLIP overexpression debilitated the promotive role of miR-324-5p-modified ADSCs in post-MI repair in rats. Conclusions miR-324-5p-modified ADSCs evidently strengthened post-MI myocardial repair by targeting TOLLIP in myocardial tissues.
Collapse
Affiliation(s)
- Zhou Ji
- Department of Cardiovascular Medicine, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chan Wang
- Jinzhou Hospital of Traditional Chinese Medicine, Jinzhou, China
| | - Qing Tong
- Office of Academic Research, The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
174
|
Liu XJ, Lv YF, Cui WZ, Li Y, Liu Y, Xue YT, Dong F. Icariin inhibits hypoxia/reoxygenation-induced ferroptosis of cardiomyocytes via regulation of the Nrf2/HO-1 signaling pathway. FEBS Open Bio 2021; 11:2966-2976. [PMID: 34407320 PMCID: PMC8564343 DOI: 10.1002/2211-5463.13276] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/25/2021] [Accepted: 08/17/2021] [Indexed: 02/06/2023] Open
Abstract
Myocardial infarction (MI) is caused by the formation of plaques in the arterial walls, leading to a decrease of blood flow to the heart and myocardium injury as a result of hypoxia. Ferroptosis is a crucial event in myocardial injury, and icariin (ICA) exerts protective effects against myocardial injury. Here, we investigated the protective mechanism of ICA in hypoxia/reoxygenation (H/R)-induced ferroptosis of cardiomyocytes. H9C2 cells were subjected to H/R induction. The content of lactate dehydrogenase and the levels of oxidative stress and intracellular ferrous ion Fe2+ were measured. The levels of ferroptosis markers (ACSL4 and GPX4) were detected. H/R-induced H9C2 cells were cultured with ICA in the presence or absence of ferroptosis inducer (erastin). Znpp (an HO-1 inhibitor) was added to ICA-treated H/R cells to verify the role of the Nrf2/HO-1 pathway. H/R-induced H9C2 cells showed reduced viability, enhanced oxidative stress and lactate dehydrogenase content, increased levels of Fe2+ and ACSL4, and decreased levels of GPX4. ICA inhibited H/R-induced ferroptosis and oxidative stress in cardiomyocytes. Erastin treatment reversed the inhibitory effect of ICA on ferroptosis in H/R cells. The expression of Nrf2 and HO-1 in H/R-induced H9C2 cells was reduced, whereas ICA treatment reversed this trend. Inhibition of the Nrf2/HO-1 pathway reversed the protective effect of ICA on H/R-induced ferroptosis. Collectively, our results suggest that ICA attenuates H/R-induced ferroptosis of cardiomyocytes by activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Xiu-Juan Liu
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Yan-Fei Lv
- Department of Rehabilitation Medicine, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, China
| | - Wen-Zhu Cui
- Department of cardiovascular diseases, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Yang Liu
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Yi-Tao Xue
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| | - Feng Dong
- Department of cardiovascular diseases, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, China
| |
Collapse
|
175
|
Li L, Huang J, Zhao Z, Wen Z, Li K, Ma T, Zhang L, Zheng J, Liang S. Decreased Spp1 Expression in Acute Myocardial Infarction after Ischemia and Reperfusion Injury. Cardiol Res Pract 2021; 2021:3925136. [PMID: 34426769 PMCID: PMC8380156 DOI: 10.1155/2021/3925136] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 07/04/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND With the progress of shock therapy and the establishment and promotion of methods such as thrombolytic therapy and percutaneous coronary intervention (PCI), many tissues and organs have been reperfused after ischemia which may cause even worse disorder called ischemia-reperfusion injury (IRI). mRNAs have been found to have significant impacts on ischemia-reperfusion through various mechanisms. In view of the accessibility of mRNAs from blood, we aimed to find the association between mRNA and ischemia-reperfusion. METHODS We used the GSE83472 dataset from the Gene Expression Omnibus (GEO) database to find differential RNA expression between ischemia-reperfusion tissue and control samples. In addition, Gene Ontology (GO) annotation and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed to find the biological property of 449 RNAs from GSE83472 via the Database for Annotation, Visualization, and Integrated Discovery (DAVID). Besides, Gene Set Enrichment Analysis (GSEA), a tool to find the pathway orientation of a gene set, was used for further study in the four most significant KEGG pathways. Furthermore, we constructed a protein-protein interaction (PPI) network. In the end, we used quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and western blotting to measure and compare the expression of Spp1 in patients who accepted percutaneous coronary intervention. RESULTS The bioinformatics analyses suggested that Spp1 was a hub gene in reperfusion after ischemia. The qRT-PCR result showed that the Spp1 expression was significantly downregulated in ischemia-reperfusion cells after PCI compared with normal samples and so as the western blotting. CONCLUSION Spp1 might play an essential role in acute myocardial infarction after ischemia and reperfusion injury.
Collapse
Affiliation(s)
- Ling Li
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jungang Huang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zongkai Zhao
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Zhuzhi Wen
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Kang Li
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Tianjiao Ma
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Lisui Zhang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Junmeng Zheng
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shi Liang
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| |
Collapse
|
176
|
Chen L, Qu J, Mei Q, Chen X, Fang Y, Chen L, Li Y, Xiang C. Small extracellular vesicles from menstrual blood-derived mesenchymal stem cells (MenSCs) as a novel therapeutic impetus in regenerative medicine. Stem Cell Res Ther 2021; 12:433. [PMID: 34344458 PMCID: PMC8330084 DOI: 10.1186/s13287-021-02511-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/11/2021] [Indexed: 02/07/2023] Open
Abstract
Menstrual blood-derived mesenchymal stem cells (MenSCs) have great potential in regenerative medicine. MenSC has received increasing attention owing to its impressive therapeutic effects in both preclinical and clinical trials. However, the study of MenSC-derived small extracellular vesicles (EVs) is still in its initial stages, in contrast to some common MSC sources (e.g., bone marrow, umbilical cord, and adipose tissue). We describe the basic characteristics and biological functions of MenSC-derived small EVs. We also demonstrate the therapeutic potential of small EVs in fulminant hepatic failure, myocardial infarction, pulmonary fibrosis, prostate cancer, cutaneous wound, type-1 diabetes mellitus, aged fertility, and potential diseases. Subsequently, novel hotspots with respect to MenSC EV-based therapy are proposed to overcome current challenges. While complexities regarding the therapeutic potential of MenSC EVs continue to be unraveled, advances are rapidly emerging in both basic science and clinical medicine. MenSC EV-based treatment has great potential for treating a series of diseases as a novel therapeutic strategy in regenerative medicine.
Collapse
Affiliation(s)
- Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Quanhui Mei
- Department of Intensive Care Unit, The First People's Hospital of Changde City, Changde, Hunan, 415000, People's Republic of China
| | - Xin Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Yangxin Fang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Lu Chen
- Innovative Precision Medicine (IPM) Group, Hangzhou, Zhejiang, 311215, People's Republic of China
| | - Yifei Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China
| | - Charlie Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, People's Republic of China.
| |
Collapse
|
177
|
Jiang W, Song J, Zhang S, Ye Y, Wang J, Zhang Y. CTRP13 Protects H9c2 Cells Against Hypoxia/Reoxygenation (H/R)-Induced Injury Via Regulating the AMPK/Nrf2/ARE Signaling Pathway. Cell Transplant 2021; 30:9636897211033275. [PMID: 34338573 PMCID: PMC8335840 DOI: 10.1177/09636897211033275] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Myocardial infarction (MI) is identified as the myocardial necrosis due to myocardial ischemia/reperfusion (I/R) injury and remains a leading cause of mortality. C1q/TNF-related protein 13 (CTRP13) is a member of CTRP family that has been found to be involved in coronary artery disease (CAD). However, the role of CTRP13 in MI remains unclear. We aimed to explore the functional role of CTRP13 in H9c2 cells exposed to hypoxia/reoxygenation (H/R). Our results demonstrated that H/R stimulation significantly decreased the expression of CTRP13 in H9c2 cells. H/R-induced an increase in ROS production and reductions in activities of SOD and CAT were prevented by CTRP13 overexpression but were aggravated by CTRP13 silencing. Moreover, CTRP13 overexpression could reverse the inductive effect of H/R on caspase-3 activity and bax expression, as well as the inhibitory effect of H/R on bcl-2 expression in H9c2 cells. However, CTRP13 silencing presented opposite effects with CTRP13 overexpression. Furthermore, CTRP13 overexpression enhanced the H/R-stimulated the expression levels of p-AMPK and nuclear Nrf2, and Nrf2 transcriptional activity. However, inhibition of AMPK reversed the CTRP13-mediated activation of Nrf2/ARE signaling and the cardiac-protective effect in H/R-exposed H9c2 cells. Additionally, silencing of Nrf2 reversed the protective effects of CTRP13 against H/R-stimulated oxidative stress and apoptosis in H9c2 cells. Finally, recombinant CTRP13 protein attenuated myocardial I/R-induced injury in rats. Taken together, these findings indicated that CTRP13 protected H9c2 cells from H/R-stimulated oxidative stress and apoptosis via regulating the AMPK/Nrf2/ARE signaling pathway. Our results provided evidence for the therapeutic potential of CTRP13 in myocardial I/R injury.
Collapse
Affiliation(s)
- Weifeng Jiang
- Department of Cardiology, Kaifeng People's Hospital, Kaifeng 475000, China
| | - Jungang Song
- Department of Cardiology, Kaifeng People's Hospital, Kaifeng 475000, China
| | - Suitao Zhang
- Department of Cardiology, Kaifeng People's Hospital, Kaifeng 475000, China
| | - Yanyan Ye
- Department of Cardiology, Kaifeng People's Hospital, Kaifeng 475000, China
| | - Jun Wang
- Teaching and Research Office of Human Anatomy, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yilin Zhang
- Department of Cardiology, Kaifeng People's Hospital, Kaifeng 475000, China
| |
Collapse
|
178
|
Tan J, Pan W, Chen H, Du Y, Jiang P, Zeng D, Wu J, Peng K. Circ_0124644 Serves as a ceRNA for miR-590-3p to Promote Hypoxia-Induced Cardiomyocytes Injury via Regulating SOX4. Front Genet 2021; 12:667724. [PMID: 34249089 PMCID: PMC8267871 DOI: 10.3389/fgene.2021.667724] [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] [Received: 02/14/2021] [Accepted: 04/16/2021] [Indexed: 11/13/2022] Open
Abstract
Circular RNA (circRNA) is an important factor for regulating the progression of many cardiovascular diseases, including acute myocardial infarction (AMI). However, the role of circ_0124644 in AMI progression remains unclear. Hypoxia was used to induce cardiomyocytes injury. The expression of circ_0124644, microRNA (miR)-590-3p, and SRY-box transcription factor 4 (SOX4) mRNA was measured by qRT-PCR. Cell counting kit 8 (CCK8) assay and flow cytometry were utilized to detect cell viability, cell cycle progression, and apoptosis. The protein levels of apoptosis markers and SOX4 were determined by western blot (WB) analysis, and the levels of oxidative stress markers were assessed using commercial Assay Kits. Dual-luciferase reporter assay, RIP assay, and RNA pull-down assay were employed to confirm the interaction between miR-590-3p and circ_0124644 or SOX4. Circ_0124644 was upregulated in AMI patients and hypoxia-induced cardiomyocytes. Hypoxia could inhibit cardiomyocytes viability, cell cycle process, and promote apoptosis and oxidative stress, while silencing circ_0124644 could alleviate hypoxia-induced cardiomyocytes injury. In terms of mechanism, circ_0124644 could target miR-590-3p. MiR-590-3p overexpression could relieve hypoxia-induced cardiomyocytes injury. Also, the suppressive effect of circ_0124644 knockdown on hypoxia-induced cardiomyocytes injury could be reversed by miR-590-3p inhibitor. Moreover, SOX4 was found to be a target of miR-590-3p, and its overexpression also could reverse the regulation of miR-590-3p on hypoxia-induced cardiomyocytes injury. Circ_0124644 silencing could alleviate hypoxia-induced cardiomyocytes injury by regulating the miR-590-3p/SOX4 axis, suggesting that it might be a target for alleviating AMI.
Collapse
Affiliation(s)
- Juan Tan
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Weinan Pan
- College of Pharmacy, Hunan Food and Drug Vocational College, Changsha, China
| | - Huilin Chen
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Yafang Du
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Peiyong Jiang
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Dianmei Zeng
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jie Wu
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Kuang Peng
- Department of Cardiology, The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
179
|
Bu S, Singh KK. Epigenetic Regulation of Autophagy in Cardiovascular Pathobiology. Int J Mol Sci 2021; 22:ijms22126544. [PMID: 34207151 PMCID: PMC8235464 DOI: 10.3390/ijms22126544] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/12/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the number one cause of debilitation and mortality worldwide, with a need for cost-effective therapeutics. Autophagy is a highly conserved catabolic recycling pathway triggered by various intra- or extracellular stimuli to play an essential role in development and pathologies, including CVDs. Accordingly, there is great interest in identifying mechanisms that govern autophagic regulation. Autophagic regulation is very complex and multifactorial that includes epigenetic pathways, such as histone modifications to regulate autophagy-related gene expression, decapping-associated mRNA degradation, microRNAs, and long non-coding RNAs; pathways are also known to play roles in CVDs. Molecular understanding of epigenetic-based pathways involved in autophagy and CVDs not only will enhance the understanding of CVDs, but may also provide novel therapeutic targets and biomarkers for CVDs.
Collapse
Affiliation(s)
| | - Krishna K. Singh
- Correspondence: ; Tel.: +1-519-661-2111 (ext. 80542) (Office) or (ext. 85683) (Lab)
| |
Collapse
|
180
|
Luo XY, Zhong Z, Chong AG, Zhang WW, Wu XD. Function and Mechanism of Trimetazidine in Myocardial Infarction-Induced Myocardial Energy Metabolism Disorder Through the SIRT1-AMPK Pathway. Front Physiol 2021; 12:645041. [PMID: 34220528 PMCID: PMC8248253 DOI: 10.3389/fphys.2021.645041] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/14/2021] [Indexed: 02/04/2023] Open
Abstract
Myocardial energy metabolism (MEM) is an important factor of myocardial injury. Trimetazidine (TMZ) provides protection against myocardial ischemia/reperfusion injury. The current study set out to evaluate the effect and mechanism of TMZ on MEM disorder induced by myocardial infarction (MI). Firstly, a MI mouse model was established by coronary artery ligation, which was then treated with different concentrations of TMZ (5, 10, and 20 mg kg-1 day-1). The results suggested that TMZ reduced the heart/weight ratio in a concentration-dependent manner. TMZ also reduced the levels of Bax and cleaved caspase-3 and promoted Bcl-2 expression. In addition, TMZ augmented adenosine triphosphate (ATP) production and superoxide dismutase (SOD) activity induced by MI and decreased the levels of lipid peroxide (LPO), free fatty acids (FFA), and nitric oxide (NO) in a concentration-dependent manner (all P < 0.05). Furthermore, an H2O2-induced cell injury model was established and treated with different concentrations of TMZ (1, 5, and 10 μM). The results showed that SIRT1 overexpression promoted ATP production and reactive oxygen species (ROS) activity and reduced the levels of LPO, FFA, and NO in H9C2 cardiomyocytes treated with H2O2 and TMZ. Silencing SIRT1 suppressed ATP production and ROS activity and increased the levels of LPO, FFA, and NO (all P < 0.05). TMZ activated the SIRT1-AMPK pathway by increasing SIRT1 expression and AMPK phosphorylation. In conclusion, TMZ inhibited MI-induced myocardial apoptosis and MEM disorder by activating the SIRT1-AMPK pathway.
Collapse
Affiliation(s)
- Xiu-Ying Luo
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| | - Ze Zhong
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| | - Ai-Guo Chong
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| | - Wei-Wei Zhang
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| | - Xin-Dong Wu
- Department of Cardiology, The Second Affiliated Hospital (Jiande Branch), Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
181
|
Zhao K, Yang CX, Li P, Sun W, Kong XQ. Epigenetic role of N6-methyladenosine (m6A) RNA methylation in the cardiovascular system. J Zhejiang Univ Sci B 2021; 21:509-523. [PMID: 32633106 DOI: 10.1631/jzus.b1900680] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As the most prevalent and abundant transcriptional modification in the eukaryotic genome, the continuous and dynamic regulation of N6-methyladenosine (m6A) has been shown to play a vital role in physiological and pathological processes of cardiovascular diseases (CVDs), such as ischemic heart failure (HF), myocardial hypertrophy, myocardial infarction (MI), and cardiomyogenesis. Regulation is achieved by modulating the expression of m6A enzymes and their downstream cardiac genes. In addition, this process has a major impact on different aspects of internal biological metabolism and several other external environmental effects associated with the development of CVDs. However, the exact molecular mechanism of m6A epigenetic regulation has not been fully elucidated. In this review, we outline recent advances and discuss potential therapeutic strategies for managing m6A in relation to several common CVD-related metabolic disorders and external environmental factors. Note that an appropriate understanding of the biological function of m6A in the cardiovascular system will pave the way towards exploring the mechanisms responsible for the development of other CVDs and their associated symptoms. Finally, it can provide new insights for the development of novel therapeutic agents for use in clinical practice.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chuan-Xi Yang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Peng Li
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei Sun
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiang-Qing Kong
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
182
|
Myocardial Infarction-Associated Extracellular Vesicle-Delivered miR-208b Affects the Growth of Human Umbilical Vein Endothelial Cells via Regulating CDKN1A. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9965639. [PMID: 34195287 PMCID: PMC8203352 DOI: 10.1155/2021/9965639] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/24/2021] [Indexed: 01/08/2023]
Abstract
This study was aimed at investigating the effects of myocardial infarction- (MI-) associated extracellular vesicle- (EV-) delivered miR-208b on human umbilical vein endothelial cells (HUVECs). EVs were isolated and subsequently stained with PHK67. A dual-luciferase reporter gene assay was used to determine the target of miR-208b. Afterwards, HUVECs were transfected with either MI-associated EVs or miR-208b mimics, and cell viability, migration, and apoptosis were subsequently measured. Real-time quantitative polymerase chain reaction (RT-qPCR) was applied to determine the expressions of the tested genes. NanoSight, transmission electron microscopy, and western blotting showed that EVs were successfully isolated. Among the potential microRNA biomarkers for MI, miR-208b was chosen for subsequent experiments. We found that MI-associated EVs could be taken up by HUVECs and confirmed that CDKN1A was a direct target of miR-208b. Additionally, miR-208b mimics and MI-associated EVs significantly inhibited the viability and migration of HUVECs (P < 0.05) and promoted cell apoptosis, as well as reduced S phase and increased G2/M phase cell distribution. RT-qPCR revealed that both miR-208b mimics and MI-associated EVs upregulated the expressions of CDKN1A, FAK, Raf-1, MAPK1, and Bax but downregulated the expression of Bcl2 and reduced the Bcl2/Bax ratio. Our study concludes that MI-associated EVs delivered miR-208b to HUVECs, and EV-delivered miR-208b could affect the growth of HUVECs by regulating the miR-208b/CDKN1A pathway; thus, miR-208b can be therefore served as important therapeutic targets for MI treatment.
Collapse
|
183
|
Yang X, Chen Y, Wang H, Fu X, Kural KC, Cao H, Li Y. Schizophrenia Plays a Negative Role in the Pathological Development of Myocardial Infarction at Multiple Biological Levels. Front Genet 2021; 12:607690. [PMID: 34149793 PMCID: PMC8211423 DOI: 10.3389/fgene.2021.607690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
It has shown that schizophrenia (SCZ) is associated with a higher chance of myocardial infarction (MI) and increased mortality. However, the underlying mechanism is largely unknown. Here, we first constructed a literature-based genetic pathway linking SCZ and MI, and then we tested the expression levels of the genes involved in the pathway by a meta-analysis using nine gene expression datasets of MI. In addition, a literature-based data mining process was conducted to explore the connection between SCZ at different levels: small molecules, complex molecules, and functional classes. The genetic pathway revealed nine genes connecting SCZ and MI. Specifically, SCZ activates two promoters of MI (IL6 and CRP) and deactivates seven inhibitors of MI (ADIPOQ, SOD2, TXN, NGF, ADORA1, NOS1, and CTNNB1), suggesting that no protective role of SCZ in MI was detected. Meta-analysis showed that one promoter of MI (CRP) presented no significant increase, and six out of seven genetic inhibitors of MI demonstrated minor to moderately increased expression. Therefore, the elevation of CRP and inhibition of the six inhibitors of MI by SCZ could be critical pathways to promote MI. Nine other regulators of MI were influenced by SCZ, including two gene families (inflammatory cytokine and IL1 family), five small molecules (lipid peroxide, superoxide, ATP, ascorbic acid, melatonin, arachidonic acid), and two complexes (CaM kinase 2 and IL23). Our results suggested that SCZ promotes the development and progression of MI at different levels, including genes, small molecules, complex molecules, and functional classes.
Collapse
Affiliation(s)
- Xiaorong Yang
- Department of Outpatient, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yao Chen
- Department of Outpatient, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Huiyao Wang
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, China
| | - Xia Fu
- Department of Outpatient, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Kamil Can Kural
- School of Systems Biology, George Mason University (GMU), Fairfax, VA, United States
| | - Hongbao Cao
- School of Systems Biology, George Mason University (GMU), Fairfax, VA, United States.,Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Ying Li
- The Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
184
|
Xu Y, Patino Gaillez M, Rothe R, Hauser S, Voigt D, Pietzsch J, Zhang Y. Conductive Hydrogels with Dynamic Reversible Networks for Biomedical Applications. Adv Healthc Mater 2021; 10:e2100012. [PMID: 33930246 PMCID: PMC11468162 DOI: 10.1002/adhm.202100012] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/21/2021] [Indexed: 12/30/2022]
Abstract
Conductive hydrogels (CHs) are emerging as a promising and well-utilized platform for 3D cell culture and tissue engineering to incorporate electron signals as biorelevant physical cues. In conventional covalently crosslinked conductive hydrogels, the network dynamics (e.g., stress relaxation, shear shining, and self-healing) required for complex cellular functions and many biomedical utilities (e.g., injection) cannot be easily realized. In contrast, dynamic conductive hydrogels (DCHs) are fabricated by dynamic and reversible crosslinks. By allowing for the breaking and reforming of the reversible linkages, DCHs can provide dynamic environments for cellular functions while maintaining matrix integrity. These dynamic materials can mimic some properties of native tissues, making them well-suited for several biotechnological and medical applications. An overview of the design, synthesis, and engineering of DCHs is presented in this review, focusing on the different dynamic crosslinking mechanisms of DCHs and their biomedical applications.
Collapse
Affiliation(s)
- Yong Xu
- Technische Universität DresdenB CUBE Center for Molecular BioengineeringDresden01307Germany
| | | | - Rebecca Rothe
- Helmholtz‐Zentrum Dresden‐Rossendorf (HZDR)Institute of Radiopharmaceutical Cancer ResearchDepartment of Radiopharmaceutical and Chemical BiologyDresden01328Germany
- Technische Universität DresdenSchool of ScienceFaculty of Chemistry and Food ChemistryDresden01062Germany
| | - Sandra Hauser
- Helmholtz‐Zentrum Dresden‐Rossendorf (HZDR)Institute of Radiopharmaceutical Cancer ResearchDepartment of Radiopharmaceutical and Chemical BiologyDresden01328Germany
| | - Dagmar Voigt
- Technische Universität Dresden, School of ScienceFaculty of BiologyInstitute of BotanyDresden01062Germany
| | - Jens Pietzsch
- Helmholtz‐Zentrum Dresden‐Rossendorf (HZDR)Institute of Radiopharmaceutical Cancer ResearchDepartment of Radiopharmaceutical and Chemical BiologyDresden01328Germany
- Technische Universität DresdenSchool of ScienceFaculty of Chemistry and Food ChemistryDresden01062Germany
| | - Yixin Zhang
- Technische Universität DresdenB CUBE Center for Molecular BioengineeringDresden01307Germany
- Cluster of Excellence Physics of LifeTechnische Universität DresdenDresden01062Germany
| |
Collapse
|
185
|
Babaei M, Rezaie J. Application of stem cell-derived exosomes in ischemic diseases: opportunity and limitations. J Transl Med 2021; 19:196. [PMID: 33964940 PMCID: PMC8106139 DOI: 10.1186/s12967-021-02863-w] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic diseases characterized by an insufficient blood flow that leads to a decrease in oxygen and nutrient uptake by cells have emerged as an important contributor to both disability and death worldwide. Up-regulation of angiogenesis may be a key factor for the improvement of ischemic diseases. This article searched articles in PubMed with the following keywords: stem cells, exosomes, angiogenesis, ischemic diseases either alone or in grouping form. The most relevant selected items were stem cell-derived exosomes and ischemic diseases. A growing body of evidence indicates that stem cells produce exosomes, which is the novel emerging approach to cell-to-cell communication and offers a new standpoint on known therapeutic strategies of ischemic diseases. Exosomes transport biological molecules such as many types of proteins, RNAs, DNA fragments, signaling molecules, and lipids between cells. Different stem cells release exosomes representing beneficial effects on ischemic diseases as they promote angiogenesis both in vitro and in vivo experiments. Application of exosomes for therapeutic angiogenesis opened new opportunities in the regenerative medicine, however, some limitations regarding exosomes isolation and application remain concerned. In addition, most of the experiments were conducted in preclinical and therefore translation of these results from bench to bed requires more effort in this field. Exosomes from stem cells are a promising tool for the treatment of ischemic diseases. In addition, translation of pre-clinic results into clinic needs further studies in this field.
Collapse
Affiliation(s)
- Majid Babaei
- Social Determinants of Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, P.O. Box: 1138, 57147, Urmia, Iran.
| |
Collapse
|
186
|
Photosynthetic biomaterials: applications of photosynthesis in algae as oxygenerator in biomedical therapies. Biodes Manuf 2021. [DOI: 10.1007/s42242-021-00129-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
187
|
Health-Related Quality of Life and Associated Factors among Myocardial Infarction Patients at Cardiac Center, Ethiopia. BIOMED RESEARCH INTERNATIONAL 2021. [DOI: 10.1155/2021/6675267] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Introduction. Myocardial infarction is the most frequent manifestation of coronary heart disease and one of the leading causes of death worldwide. The sudden and often profound physiological and psychological effect of the acute onset of myocardial infarction hurts the health-related quality of life. Objective. To assess health-related quality of life and associated factors among myocardial infarction patients at Cardiac Center, Ethiopia, Addis Ababa, Ethiopia, 2020. Method. Institution-based cross-sectional study was conducted from April 10 to June 25, 2020, at the Cardiac Center-Ethiopia, Addis Ababa, Ethiopia. The sample consisted of 421 myocardial infarction patients. Data were collected through an interviewer-administered structured questionnaire by using the World Health Organization Quality of Life Questionnaire. Samples were selected using a consecutive sampling technique. The linear regression analysis model was fitted using SPSS 26 and STATA 14, and the unstandardized beta (𝛽) coefficient with a 95% confidence interval was used. A
value <0.05 was considered statistically significant for all analyses. Results. The mean score of the overall health-related quality of life was found to be
, and the mean score for the physical domain was
; for the psychological domain, it was
, and for environmental and social relationship domains, it was
and
, respectively. In the multiple regression analysis, increased age, living in a rural area, heart failure, and hypertension were inversely associated with overall health-related quality of life, while secondary and higher education were associated with better overall health-related quality of life. Conclusion. Patients with myocardial infarction at Cardiac Center, Ethiopia, had lower health-related quality of life. Hence, the finding of this study suggests the implementation of a cardiac rehabilitation program and comprehensive service given by healthcare providers.
Collapse
|
188
|
Pala R, Pattnaik S, Busi S, Nauli SM. Nanomaterials as Novel Cardiovascular Theranostics. Pharmaceutics 2021; 13:pharmaceutics13030348. [PMID: 33799932 PMCID: PMC7998597 DOI: 10.3390/pharmaceutics13030348] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/16/2022] Open
Abstract
Cardiovascular diseases (CVDs) are a group of conditions associated with heart and blood vessels and are considered the leading cause of death globally. Coronary heart disease, atherosclerosis, myocardial infarction represents the CVDs. Since CVDs are associated with a series of pathophysiological conditions with an alarming mortality and morbidity rate, early diagnosis and appropriate therapeutic approaches are critical for saving patients’ lives. Conventionally, diagnostic tools are employed to detect disease conditions, whereas therapeutic drug candidates are administered to mitigate diseases. However, the advent of nanotechnological platforms has revolutionized the current understanding of pathophysiology and therapeutic measures. The concept of combinatorial therapy using both diagnosis and therapeutics through a single platform is known as theranostics. Nano-based theranostics are widely used in cancer detection and treatment, as evident from pre-clinical and clinical studies. Nanotheranostics have gained considerable attention for the efficient management of CVDs. The differential physicochemical properties of engineered nanoparticles have been exploited for early diagnosis and therapy of atherosclerosis, myocardial infarction and aneurysms. Herein, we provided the information on the evolution of nano-based theranostics to detect and treat CVDs such as atherosclerosis, myocardial infarction, and angiogenesis. The review also aims to provide novel avenues on how nanotherapeutics’ trending concept could transform our conventional diagnostic and therapeutic tools in the near future.
Collapse
Affiliation(s)
- Rajasekharreddy Pala
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA 92618, USA
- Department of Medicine, University of California Irvine, Irvine, CA 92868, USA
- Correspondence: (R.P.); (S.M.N.); Tel.: +1-714-516-5462 (R.P.); +1-714-516-5480 (S.M.N.); Fax: +1-714-516-5481 (R.P. & S.M.N.)
| | - Subhaswaraj Pattnaik
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India; (S.P.); (S.B.)
| | - Siddhardha Busi
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India; (S.P.); (S.B.)
| | - Surya M. Nauli
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University, Irvine, CA 92618, USA
- Department of Medicine, University of California Irvine, Irvine, CA 92868, USA
- Correspondence: (R.P.); (S.M.N.); Tel.: +1-714-516-5462 (R.P.); +1-714-516-5480 (S.M.N.); Fax: +1-714-516-5481 (R.P. & S.M.N.)
| |
Collapse
|
189
|
Zhang X, Chen Z, Zang J, Yao C, Shi J, Nie R, Wu G. LncRNA-mRNA co-expression analysis discovered the diagnostic and prognostic biomarkers and potential therapeutic agents for myocardial infarction. Aging (Albany NY) 2021; 13:8944-8959. [PMID: 33668039 PMCID: PMC8034908 DOI: 10.18632/aging.202713] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 02/09/2021] [Indexed: 12/14/2022]
Abstract
Currently, the role of lncRNA in myocardial infarction (MI) is poorly understood. 17 co-expression modules were determined, specifically, the greenyellow, saddlebrown, grey60, royalblue, lightgreen, white, and pink modules were specifically expressed in the acute phase of MI, and brown, darkred, and royalblue, while greenyellow modules were specifically expressed in MI compared with CAD. 12 time-dependent of lncRNA/mRNA clusters with consistent expression trends were also identified. MI-associated modules were mainly enriched to immune, cell cycle, and metabolic pathways. We further obtained a network of 1816 lncRNA-mRNAs with higher expression correlations among these lncRNAs by analyzing the topological properties of the network. Herein, lncRNA RP11-847H18.2 and KLHL28, SPRTN, and EPM2AIP1 were determined as gene markers specifically expressed in MI, and they demonstrated a high predictive performance for MI diagnosis and prognosis. Three drugs, namely, Calcium citrate, Calcium Phosphate, and Calcium phosphate dihydrate, were identified as potential precursors of MI. Finally, gene and lncRNA diagnostic models were developed based on these genes and lncRNAs, with their AUCs averaged above 0.89 in both training and validation datasets. The findings of this study improve the diagnosis and prognosis of MI and personalized treatment of MI.
Collapse
Affiliation(s)
- Xiaocong Zhang
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China.,Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Ziqi Chen
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jiabin Zang
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Chun Yao
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Jian Shi
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China
| | - Ruqiong Nie
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Guifu Wu
- Department of Cardiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.,Guangdong Innovative Engineering and Technology Research Center for Assisted Circulation, Shenzhen, China.,NHC Key Laboratory of Assisted Circulation, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
190
|
Sundararaman SS, van der Vorst EPC. Calcium-Sensing Receptor (CaSR), Its Impact on Inflammation and the Consequences on Cardiovascular Health. Int J Mol Sci 2021; 22:2478. [PMID: 33804544 PMCID: PMC7957814 DOI: 10.3390/ijms22052478] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/11/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022] Open
Abstract
The calcium Sensing Receptor (CaSR) is a cell surface receptor belonging to the family of G-protein coupled receptors. CaSR is mainly expressed by parathyroid glands, kidneys, bone, skin, adipose tissue, the gut, the nervous system, and the cardiovascular system. The receptor, as its name implies is involved in sensing calcium fluctuations in the extracellular matrix of cells, thereby having a major impact on the mineral homeostasis in humans. Besides calcium ions, the receptor is also activated by other di- and tri-valent cations, polypeptides, polyamines, antibiotics, calcilytics and calcimimetics, which upon binding induce intracellular signaling pathways. Recent studies have demonstrated that CaSR influences a wide variety of cells and processes that are involved in inflammation, the cardiovascular system, such as vascular calcification, atherosclerosis, myocardial infarction, hypertension, and obesity. Therefore, in this review, the current understanding of the role that CaSR plays in inflammation and its consequences on the cardiovascular system will be highlighted.
Collapse
Affiliation(s)
- Sai Sahana Sundararaman
- Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany;
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany;
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, 6229 ER Maastricht, The Netherlands
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany
- German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
191
|
Ma C, Ding H, Deng Y, Liu H, Xiong X, Yang Y. Irisin: A New Code Uncover the Relationship of Skeletal Muscle and Cardiovascular Health During Exercise. Front Physiol 2021; 12:620608. [PMID: 33597894 PMCID: PMC7882619 DOI: 10.3389/fphys.2021.620608] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/11/2021] [Indexed: 12/12/2022] Open
Abstract
Exercise not only produces beneficial effects on muscle itself via various molecular pathways, but also mediates the interaction between muscles and other organs in an autocrine/paracrine manner through myokines, which plays a positive role in maintaining overall health. Irisin, an exercise-derived myokine, has been found involved in the regulation of some cardiovascular diseases. However, the relationship between irisin and cardiovascular health is not fully elucidated and there are some divergences on the regulation of irisin by exercise. In this review, we present the current knowledge on the origin and physiology of irisin, describe the regulation of irisin by acute and chronic exercises, and discuss the divergences of the related research results. Importantly, we discuss the role of irisin as a biomarker in the diagnosis of cardiovascular diseases and describe its treatment and molecular mechanism in some cardiovascular diseases. It is expected that irisin will be used as a therapeutic agent to combat cardiovascular diseases or other disorders caused by inactivity in the near future.
Collapse
Affiliation(s)
- Chunlian Ma
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Haichao Ding
- Graduate School, Wuhan Sports University, Wuhan, China
| | - Yuting Deng
- Graduate School, Wuhan Sports University, Wuhan, China
| | - Hua Liu
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Xiaoling Xiong
- College of Health Science, Wuhan Sports University, Wuhan, China
| | - Yi Yang
- College of Health Science, Wuhan Sports University, Wuhan, China
| |
Collapse
|
192
|
Chen HY, Xiao ZZ, Ling X, Xu RN, Zhu P, Zheng SY. ELAVL1 is transcriptionally activated by FOXC1 and promotes ferroptosis in myocardial ischemia/reperfusion injury by regulating autophagy. Mol Med 2021; 27:14. [PMID: 33568052 PMCID: PMC7874472 DOI: 10.1186/s10020-021-00271-w] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Aims Myocardial ischemia is the most common form of cardiovascular disease and the leading cause of morbidity and mortality. Understanding the mechanisms is very crucial for the development of effective therapy. Therefore, this study aimed to investigate the functional roles and mechanisms by which ELAVL1 regulates myocardial ischemia and reperfusion (I/R) injury. Methods Mouse myocardial I/R model and cultured myocardial cells exposed to hypoxia/reperfusion (H/R) were used in this study. Features of ferroptosis were evidenced by LDH activity, GPx4 activity, cellular iron, ROS, LPO, and GSH levels. The expression levels of autophagy markers (Beclin-1, p62, LC3), ELAVL1 and FOXC1 were measured by qRT-PCR, immunostaining and western blot. RIP assay, biotin-pull down, ChIP and dual luciferase activity assay were employed to examine the interactions of ELAVL1/Beclin-1 mRNA and FOXC1/ELAVL1 promoter. CCK-8 assay was used to examine viability of cells. TTC staining was performed to assess the myocardial I/R injury. Results Myocardial I/R surgery induced ferroptosis and up-regulated ELAVL1 level. Knockdown of ELAVL1 decreased ferroptosis and ameliorated I/R injury. Si-ELAVL1 repressed autophagy and inhibition of autophagy by inhibitor suppressed ferroptosis and I/R injury in myocardial cells. Increase of autophagy could reverse the effects of ELAVL1 knockdown on ferroptosis and I/R injury. ELAVL1 directly bound with and stabilized Beclin-1 mRNA. Furthermore, FOXC1 bound to ELAVL1 promoter region and activated its transcription upon H/R exposure. Conclusion FOXC1 transcriptionally activated ELAVL1 may promote ferroptosis during myocardial I/R by modulating autophagy, leading to myocardial injury. Inhibition of ELAVL1-mediated autophagic ferroptosis would be a new viewpoint in the treatment of myocardial I/R injury.
Collapse
Affiliation(s)
- Hui-Yong Chen
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, No.1838 North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, Guangdong, People's Republic of China.,Department of Thoracic Surgery, Yuebei People's Hospital, Shantou University, Shaoguan, 512026, Guangdong, People's Republic of China
| | - Ze-Zhou Xiao
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, No.1838 North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Xiao Ling
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, No.1838 North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Rong-Ning Xu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, No.1838 North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Peng Zhu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, No.1838 North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, Guangdong, People's Republic of China.
| | - Shao-Yi Zheng
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, No.1838 North Guangzhou Avenue, Baiyun District, Guangzhou, 510515, Guangdong, People's Republic of China.
| |
Collapse
|
193
|
Wang Q, Wang K, Zhao X. Monocytes recruitment blocking synergizes with mesenchymal stem cell transplantation for treating myocardial infarction. Regen Med 2021; 16:9-17. [PMID: 33560157 DOI: 10.2217/rme-2020-0047] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Mesenchymal stem cell (MSC) transplantation is a promising therapeutic approach for acute myocardial infarction (AMI), however, research to date has demonstrated unsatisfactory results. Materials & methods: An AMI mouse model was established via left coronary artery ligation. AMI mice were treated with MSCs, anti-CCR2 or MSCs + anti-CCR2 and the effects of each treatment group were compared. Macrophage infiltration was analyzed by immunofluorescence staining and flow cytometry. Results: Implantation of MSCs + anti-CCR2 yielded a greater improvement in cardiac function and significantly reduced macrophage accumulation in the infarct site of AMI mice compared with the injection of MSCs or anti-CCR2 alone. Moreover, reduced macrophage infiltration was accompanied by reduced pro-inflammatory cytokine secretion in the injury sites and the low inflammatory response favored tissue regeneration. Conclusion: Treatment with MSCs and anti-CCR2 in combination may be a promising therapeutic strategy for AMI.
Collapse
Affiliation(s)
- Qian Wang
- Department of Cardiology, Changhai Hospital, Naval Medical University, No.168 Changhai Road, Shanghai 200433, China
| | - Ke Wang
- Department of Cardiology, Changhai Hospital, Naval Medical University, No.168 Changhai Road, Shanghai 200433, China
| | - Xianxian Zhao
- Department of Cardiology, Changhai Hospital, Naval Medical University, No.168 Changhai Road, Shanghai 200433, China
| |
Collapse
|
194
|
Abstract
Isofraxidin is a well-known coumarin compound refined from traditional Chinese medicines. It has been previously demonstrated to play an anti-inflammatory role in various inflammatory conditions. However, the effect of isofraxidin on myocardial infarction (MI) remains uncovered. In this study, we aimed to investigate the effect of isofraxidin on MI. MI mice was created and triphenyltetrazolium chloride (TTC) staining as well as echocardiographic evaluation were conducted to analyze the severity of MI. Oxygen-glucose deprivation (OGD) was used for the mimics of ischemic stress in murine cardiomyocytes, and Cell Counting Kit-8 (CCK-8), Annexin V, and lactate dehydrogenase (LDH) release assays were conducted for cell viability. Western blot was used for the detection of NOD-like receptor family, pyrin domain containing 3 (NLRP3), and adapter protein apoptosis-associated speck-like protein (ASC) in heart tissues and cardiomyocytes. Real-time polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA) were applied for the detection of proinflammatory cytokines. We found that isofraxidin alleviated the severity of MI and produced a cardio-protective effect against OGD damage. Isofraxidin also decreased the overall and local inflammatory reaction in MI. Those effects were through the inhibition of the NLRP3 inflammasome. Taken together, we initially reported the cardio-protective and alleviative effect of isofraxidin on MI and uncovered its underlying mechanism related to the NLRP3 inflammasome inhibition.
Collapse
|
195
|
Huang Q, Wang J, Li D, Zhao J, Feng X, Zhou N. Exercise electrocardiography combined with stress echocardiography for predicting myocardial ischemia in adults. Exp Ther Med 2021; 21:130. [PMID: 33376512 PMCID: PMC7751467 DOI: 10.3892/etm.2020.9562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Myocardial ischemia (MI) has the highest mortality rate in the world. Traditional noninvasive MI examinations include exercise electrocardiography tests (EETs) and stress echocardiography (SE). Treadmill and dobutamine tests are commonly used as stress protocols. In the present study, 278 patients with suspected MI were examined, 66 of whom were diagnosed with MI and 212 did not show evidence of MI by coronary angiography (CAG)/coronary CT angiography (CCTA). All patients underwent clinical EET and SE evaluations prior to CAG/CCTA. All groups were compared based on specific clinical parameters including age, sex, blood pressure, heart rate, blood oxygen saturation, underlying conditions and ejection fraction/fraction shortening. The data indicated superior diagnostic efficiency of the combined EET+SE method for the diagnosis of suspected MI compared with either EET or SE alone. The sensitivity/specificity/positive predictive value and negative predictive value for detecting MI were excellent compared with those of traditional examinations. The diagnostic efficiency of the combination analysis may reduce the prevalence of MI and medical costs. The present study provided novel insight for the development of methods that may be used for MI detection and prediction.
Collapse
Affiliation(s)
- Qiong Huang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Junhong Wang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Dianfu Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Jihong Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| | - Xiangjun Feng
- Department of Geriatric General Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650000, P.R. China
| | - Ningtian Zhou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
196
|
Liu CY, Zhou Y, Chen T, Lei JC, Jiang XJ. AMPK/SIRT1 Pathway is Involved in Arctigenin-Mediated Protective Effects Against Myocardial Ischemia-Reperfusion Injury. Front Pharmacol 2021; 11:616813. [PMID: 33574759 PMCID: PMC7870703 DOI: 10.3389/fphar.2020.616813] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022] Open
Abstract
Arctigenin, one of the active ingredients extracted from Great Burdock (Arctium lappa) Achene, has been found to relieve myocardial infarction injury. However, the specific mechanism of Arctigenin against myocardial infarction remains largely unknown. Here, both acute myocardial ischemia-reperfusion injury (AMI/R) rat model and oxygen glucose deprivation (OGD)-induced myocardial cell injury model were constructed to explore the underlying role of AMPK/SIRT1 pathway in Arctigenin-mediated effects. The experimental data in our study demonstrated that Arctigenin ameliorated OGD-mediated cardiomyocytes apoptosis, inflammation and oxidative stress in a dose-dependent manner. Besides, Arctigenin activated AMPK/SIRT1 pathway and downregulated NF-κB phosphorylation in OGD-treated cardiomyocytes, while inhibiting AMPK or SIRT1 by the Compound C (an AMPK inhibitor) or SIRT1-IN-1 (a SIRT1 inhibitor) significantly attenuated Arctigenin-exerted protective effects on cardiomyocytes. In the animal experiments, Arctigenin improved the heart functions and decreased infarct size of the AMI/R-rats, accompanied with downregulated oxidative stress, inflammation and apoptotic levels in the heart tissues. What's more, Arctigenin enhanced the AMPK/SIRT1 pathway and repressed NF-κB pathway activation. Taken together, our data indicated that Arctigenin reduced cardiomyocytes apoptosis against AMI/R-induced oxidative stress and inflammation at least via AMPK/SIRT1 pathway.
Collapse
Affiliation(s)
- Cheng-Yin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Yi Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Tao Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Jing-Chao Lei
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Xue-Jun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, China.,Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
197
|
Wang W, Yang F, Lin X, Zhong Q, Li Z, Chen X, Wang J, He K. The Preference, Effect, and Prognosis of Intra-Aortic Balloon Counterpulsation in Acute Myocardial Infarction Complicated by Cardiogenic Shock Patients: A Retrospective Cohort Study. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6656926. [PMID: 33542922 PMCID: PMC7840249 DOI: 10.1155/2021/6656926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/04/2021] [Accepted: 01/08/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUNDS Intra-aortic balloon counterpulsation is increasingly used in acute myocardial infarction complicated by cardiogenic shock. The aim of this study was to explore the preference, effect, and prognosis of intra-aortic balloon counterpulsation in acute myocardial infarction complicated by cardiogenic shock patients. METHODS Data of acute myocardial infarction complicated by cardiogenic shock patients at the Fourth Medical Center of PLA General Hospital were collected retrospectively. A propensity score was calculated with a logistic regression which contained clinically meaningful variables and variables selected by Lasso and then used to match the control group. The cumulative incidence curve and Gray's test were employed to analyse the effect and prognosis of intra-aortic balloon counterpulsation on mortality. RESULTS A total of 1962 acute myocardial infarction cases admitted between May 2015 and November 2018 were identified, and 223 cases with acute myocardial infarction complicated by cardiogenic shock were included as the study cohort, which contained 34 cases that received IABP and 189 cases that did not receive IABP. Patients with higher alanine aminotransferase (OR = 1.93, 95% CI 1.29-2.98), higher triglyceride (OR = 3.71, 95% CI 1.87-7.95), and higher blood glucose (OR = 1.08, 95% CI 0.99-1.18) had a higher probability of receiving intra-aortic balloon counterpulsation. In the propensity score matching analysis, 34 cases received intra-aortic balloon counterpulsation and 102 matched controls were included in the comparison. By comparing the cumulative incidence of in-hospital mortality, there was no statistically significant difference between the intra-aortic balloon counterpulsation group and matched control group (P = 0.454). CONCLUSION The use of intra-aortic balloon counterpulsation may not improve the prognosis of the acute myocardial infarction complicated by cardiogenic shock patients.
Collapse
Affiliation(s)
- Wenjun Wang
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese PLA General Hospital, Beijing 100853, China
- Translational Medical Research Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical Artificial Intelligence Research Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Feifei Yang
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese PLA General Hospital, Beijing 100853, China
- Translational Medical Research Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical Artificial Intelligence Research Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Xixiang Lin
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese PLA General Hospital, Beijing 100853, China
- Translational Medical Research Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical Artificial Intelligence Research Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Qin Zhong
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese PLA General Hospital, Beijing 100853, China
- Translational Medical Research Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical Artificial Intelligence Research Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Zongren Li
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese PLA General Hospital, Beijing 100853, China
- Translational Medical Research Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical Artificial Intelligence Research Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Xu Chen
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese PLA General Hospital, Beijing 100853, China
- Translational Medical Research Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical Artificial Intelligence Research Center, Chinese PLA General Hospital, Beijing 100853, China
| | - Junfeng Wang
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, 3584 CG Utrecht, Netherlands
| | - Kunlun He
- Key Laboratory of Ministry of Industry and Information Technology of Biomedical Engineering and Translational Medicine, Chinese PLA General Hospital, Beijing 100853, China
- Translational Medical Research Center, Chinese PLA General Hospital, Beijing 100853, China
- Medical Artificial Intelligence Research Center, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
198
|
Jin Z, Wu Q, Chen S, Gao J, Li X, Zhang X, Zhou Y, He D, Cheng Z, Zhu Y, Wu S. The Associations of Two Novel Inflammation Indexes, SII and SIRI with the Risks for Cardiovascular Diseases and All-Cause Mortality: A Ten-Year Follow-Up Study in 85,154 Individuals. J Inflamm Res 2021; 14:131-140. [PMID: 33500649 PMCID: PMC7822090 DOI: 10.2147/jir.s283835] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/31/2020] [Indexed: 01/12/2023] Open
Abstract
Background SII and SIRI are two novel systemic inflammation indexes that were suggested in predicting poor outcomes in cancers. However, no studies have examined their effect on cardiovascular diseases (CVDs) and all-cause mortality. Thus, this study aims to investigate associations between SII, SIRI, and the risks for CVDs and all-cause mortality. Methods A total of 85,154 participants from the Kailuan cohort were included and followed up for incidents of CVDs (including MI, stroke) and all-cause death for 10 years. Multiple Cox regression was used to calculate the adjusted hazard ratios (HRs). Results During the follow-up period, 4262 stroke events, 1233 MI events, and 7225 all-cause deaths were identified, respectively. Compared with the lowest quantile (Q1) of SII or SIRI, after adjusted for most cardiovascular risk factors, both indexes showed positive associations with the risk for stroke (adjusted HRs in Q4 were 1.264 (95% CI: 1.157,1.382) for SII, 1.194 (95% CI: 1.087,1.313) for SIRI), and all-cause death (adjusted HRs in Q4 were 1.246 (95% CI: 1.165,1.331) for SII, 1.393 (95% CI: 1.296,1.498) for SIRI). Additionally, higher SII and SIRI are also associated with increased risk of hemorrhagic stroke and ischemic stroke. Higher SIRI but not SII exhibited a higher MI risk, the adjusted HR in Q4 was 1.204 (1.013,1.431). The significant association remained after additional adjustment for CRP. Subgroup analysis and sensitivity analysis displayed consistent results except for SIRI with MI, where the association did not arrive at significance in subjects aged ≥60. Conclusion Elevated SII and SIRI increased the risk of stroke, two stroke subtypes, and all-cause death. Higher SIRI, but not SII associated with increased MI incidence, and the association of SIRI was only significant in subjects aged <60.
Collapse
Affiliation(s)
- Ziqi Jin
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Qiong Wu
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Shuohua Chen
- Health Care Center, Kailuan Group, Tangshan 063000, People's Republic of China
| | - Jingli Gao
- Department of Intensive Medicine, Kailuan General Hospital, Tangshan 063000, People's Republic of China
| | - Xiaolan Li
- Department of Intensive Medicine, Kailuan General Hospital, Tangshan 063000, People's Republic of China
| | - Xuhui Zhang
- Hangzhou Center for Disease Control and Prevention, Hangzhou 310051, Zhejiang, People's Republic of China
| | - Yaohan Zhou
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Di He
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Zongxue Cheng
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Yimin Zhu
- Department of Epidemiology & Biostatistics, School of Public Health, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China.,Department of Respiratory Diseases, Sir Run Run Shaw Hospital Affiliated to School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310060, People's Republic of China.,Department of Pathology, School of Medicine, Zhejiang University, Hangzhou 310058, Zhejiang, People's Republic of China
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, 063000, People's Republic of China
| |
Collapse
|
199
|
Malik S, Naithani M, Mirza A, Darbari A, Saxena R. Possible mechanisms of cardiovascular complications and troponin elevation in coronavirus disease: A narrative review. HEART AND MIND 2021. [DOI: 10.4103/hm.hm_16_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
200
|
Mnafgui K, Khdhiri E, Hajji R, Feriani A, Ivan da Silva F, Laíres da Silva Santos A, Tlili A, Mlayeh S, Bouzidi M, Ammar H, Abid S. Potential effect of new (E)-4-hydroxy -N'-(1-(7-hydroxy-2-oxo-2H-chromen-3-yl) ethylidene) benzohydrazide against acute myocardial infarction: Haemodynamic, biochemical and histological studies. Clin Exp Pharmacol Physiol 2021; 48:107-120. [PMID: 32780517 DOI: 10.1111/1440-1681.13397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 01/14/2023]
Abstract
This study aimed to explore the cardioprotective effect of new synthesized coumarin (E)-4-hydroxy-N'-(1-(7-hydroxy-2-oxo-2H-chromen-3-yl) ethylidene) benzohydrazide denoted (Hyd.Cou) against myocardial infarction disorders. Male Wistar rats were divided into four groups; Control, isoproterenol (ISO), ISO + Acenocoumarol (Ac) and ISO + Hyd.Cou. Results showed that the ISO group exhibited serious alteration in EGC pattern, significant heart hypertrophy (+33%), haemodynamic disturbance and increase in plasma rate of CK-MB, LDH and troponin-T by 44, 53, and 170%, respectively, as compared to Control. Moreover, isoproterenol induced a rise in plasma angiotensin-converting enzyme activity (ACE) by 49%, dyslipidaemia, and increased thiobarbituric acid-reactive substances (TBARS) by 117% associated with decrease in the activity of superoxide dismutase (SOD) and glutathione peroxidase (GPx) by 46% and 58%, respectively in myocardium. Interestingly, the molecular docking calculation demonstrated strong interactions of Hyd.Cou with the receptors of the protein disulphide isomerase (PDI) which could highlight the antithrombotic effect. Moreover, Hyd.Cou improved plasma cardiac dysfunction biomarkers, mitigated the ventricle remodelling process and alleviated heart oxidative stress damage. Overall, Hyd.Cou prevented the heart from the remodelling process through inhibition of ACE activity and oxidative stress improvement.
Collapse
Affiliation(s)
- Kais Mnafgui
- Laboratory of Animal Physiology, Faculty of Sciences of Sfax, University of Sfax, Sfax, Tunisia
| | - Emna Khdhiri
- Laboratoire de Chimie Appliquée 'Hétérocycles Corps Gras & Polymères', Faculté des Sciences, Université de Sfax, Sfax, Tunisie
| | - Raouf Hajji
- Service de Médecine Interne, Hôpital de Sidi Bouzid, Sidi Bouzid, Tunisie
- Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisie
| | - Anouar Feriani
- Laboratory of Animal Physiology, Faculty of Sciences of Sfax, University of Sfax, Sfax, Tunisia
| | | | | | - Abir Tlili
- Faculty of Medicine of Monastir, Monastir, Tunisia
| | - Souhail Mlayeh
- Faculté de Médecine de Sousse, Université de Sousse, Sousse, Tunisie
| | - Moncef Bouzidi
- Service de Médecine Interne, Hôpital de Sidi Bouzid, Sidi Bouzid, Tunisie
| | - Houcine Ammar
- Laboratoire de Chimie Appliquée 'Hétérocycles Corps Gras & Polymères', Faculté des Sciences, Université de Sfax, Sfax, Tunisie
| | - Souhir Abid
- Laboratoire de Chimie Appliquée 'Hétérocycles Corps Gras & Polymères', Faculté des Sciences, Université de Sfax, Sfax, Tunisie
| |
Collapse
|