1
|
Wu C, Bao S, Sun H, Chen X, Yang L, Li R, Peng Y. Noncoding RNAs regulating ferroptosis in cardiovascular diseases: novel roles and therapeutic strategies. Mol Cell Biochem 2024; 479:2827-2841. [PMID: 38064139 PMCID: PMC11473578 DOI: 10.1007/s11010-023-04895-w] [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: 03/30/2023] [Accepted: 11/06/2023] [Indexed: 10/15/2024]
Abstract
The morbidity and mortality rates of cardiovascular diseases (CVDs) are increasing; thus, they impose substantial health and economic burdens worldwide, and effective interventions are needed for immediate resolution of this issue. Recent studies have suggested that noncoding RNAs (ncRNAs) play critical roles in the occurrence and development of CVDs and are potential therapeutic targets and novel biomarkers for these diseases. Newly discovered modes of cell death, including necroptosis, pyroptosis, apoptosis, autophagy-dependent cell death and ferroptosis, also play key roles in CVD progression. However, ferroptosis, which differs from the other aforementioned forms of regulated cell death in terms of cell morphology, biochemistry and inhereditability, is a unique iron-dependent mode of nonapoptotic cell death induced by abnormal iron metabolism and excessive accumulation of iron-dependent lipid peroxides and reactive oxygen species (ROS). Increasing evidence has confirmed that ncRNA-mediated ferroptosis is involved in regulating tissue homeostasis and CVD-related pathophysiological conditions, such as cardiac ischemia/reperfusion (I/R) injury, myocardial infarction (MI), atrial fibrillation (AF), cardiomyopathy and heart failure (HF). In this review, we summarize the underlying mechanism of ferroptosis, discuss the pathophysiological effects of ncRNA-mediated ferroptosis in CVDs and provide ideas for effective therapeutic strategies.
Collapse
Affiliation(s)
- Changyong Wu
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Suli Bao
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Huang Sun
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xiaocui Chen
- Department of Gastroenterology, Affiliated Hospital of Panzhihua University, Panzhihua, China
| | - Lu Yang
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruijie Li
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China.
| | - Yunzhu Peng
- Department of Cardiology, the First Affiliated Hospital of Kunming Medical University, Kunming, China.
| |
Collapse
|
2
|
Wang Q, Xue X, Wang P, Yu Y, Wang J, Jiang Q, Xiao J. Angiotensin 1 peptide-conjugated CdSe/ZnS quantum dots for cardiac-specific hydrogen sulfide targeted therapy in myocardial ischemia-reperfusion injury. Front Pharmacol 2024; 15:1435282. [PMID: 39415837 PMCID: PMC11479923 DOI: 10.3389/fphar.2024.1435282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Myocardial ischemia/reperfusion (I/R) injury remains a major obstacle in cardiovascular therapies. Hydrogen sulfide (H2S) shows promise for mitigating I/R injury, but conventional delivery methods, such as NaHS injections or adenovirus-mediated CSE gene therapy, face low efficiency and systemic side effects. This study explores the use of angiotensin 1 (AT1) peptide-conjugated CdSe/ZnS quantum dots (QDs) for targeted delivery of cystathionine-γ-lyase (CSE) plasmids to the myocardium, aiming to boost local H2S production and minimize I/R injury. Methods CdSe/ZnS QDs were conjugated with AT1 peptides to create a nanocarrier system capable of delivering the CSE plasmid specifically to the myocardium. In vivo fluorescence imaging confirmed heart-specific accumulation. Myocardial infarct size, cardiac function, cell death, and oxidative stress were evaluated. Endoplasmic reticulum stress and mitophagy markers, including CHOP/GRP78/eIF2α, were analyzed, and the CHOP gene's role was further assessed using an adenovirus vector. Results The AT1-conjugated nanocarriers significantly increased CSE expression in the myocardium, as confirmed by fluorescence imaging, without affecting other organs. This localized delivery reduced myocardial infarct size, improved cardiac function, and decreased oxidative stress and cell death. Importantly, a reduction in endoplasmic reticulum stress and mitophagy markers was observed, suggesting that cardioprotection was mediated via the CHOP/GRP78/eIF2α signaling pathway. Reintroduction of CHOP using an adenovirus vector reversed these protective effects, confirming the pathway's involvement. Discussion This study demonstrates that AT1 peptide-conjugated QDs can effectively deliver CSE plasmids to the heart, providing significant protection against I/R injury through enhanced localized H2S production. This approach offers a promising, targeted, and side-effect-free therapy for myocardial I/R injury, with potential for clinical translation.
Collapse
Affiliation(s)
- Qing Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaofei Xue
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Pei Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yue Yu
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jun Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qixia Jiang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
3
|
Zeng J, Cao J, Yang H, Wang X, Liu T, Chen Z, Shi F, Xu Z, Lin X. Overview of mechanism of electroacupuncture pretreatment for prevention and treatment of cardiovascular and cerebrovascular diseases. CNS Neurosci Ther 2024; 30:e14920. [PMID: 39361504 PMCID: PMC11448663 DOI: 10.1111/cns.14920] [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: 04/23/2024] [Revised: 07/19/2024] [Accepted: 08/01/2024] [Indexed: 10/05/2024] Open
Abstract
Cardio-cerebrovascular disease (CCVD) is a serious threat to huma strategy to prevent the occurrence and development of disease by giving electroacupuncture intervention before the disease occurs. EAP has been shown in many preclinical studies to relieve ischemic symptoms and improve damage from ischemia-reperfusion, with no comprehensive review of its mechanisms in cardiovascular disease yet. In this paper, we first systematically discussed the meridian and acupoint selection law of EAP for CCVD and focused on the progress of the mechanism of action of EAP for the prevention and treatment of CCVD. As a result, in preclinical studies, AMI and MCAO models are commonly used to simulate ischemic injury in CCVD, while MIRI and CI/RI models are used to simulate reperfusion injury caused by blood flow recovery after focal tissue ischemia. According to the meridian matching rules of EAP for CCVD, PC6 in the pericardial meridian is the most commonly used acupoint in cardiovascular diseases, while GV20 in the Du meridian is the most commonly used acupoint in cerebrovascular diseases. In terms of intervention parameters, EAP intervention generally lasts for 30 min, with acupuncture depths mostly between 1.5 and 5 mm, stimulation intensities mostly at 1 mA, and commonly used frequencies being low frequencies. In terms of molecular mechanisms, the key pathways of EAP in preventing and treating cardiovascular and cerebrovascular diseases are partially similar. EAP can play a protective role in cardiovascular and cerebrovascular diseases by promoting autophagy, regulating Ca2+ overload, and promoting vascular regeneration through anti-inflammatory reactions, antioxidant stress, and anti-apoptosis. Of course, both pathways involved have their corresponding specificities. When using EAP to prevent and treat cardiovascular diseases, it involves the metabolic pathway of glutamate, while when using EAP to prevent and treat cerebrovascular diseases, it involves the homeostasis of the blood-brain barrier and the release of neurotransmitters and nutritional factors. I hope these data can provide experimental basis and reference for the clinical promotion and application of EAP in CCVD treatment.
Collapse
Affiliation(s)
- Jiaming Zeng
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
| | - Jiaojiao Cao
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
| | - Haitao Yang
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xue Wang
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
| | - Tingting Liu
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
| | - Zhihan Chen
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
| | - Fangyuan Shi
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
| | - Zhifang Xu
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, School of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| | - Xiaowei Lin
- Research Center of Experimental Acupuncture Science, School of Acupuncture‐Moxibustion and TuinaTianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Modern Chinese Medicine Theory of Innovation and Application, School of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
- National Clinical Research Center for Chinese Medicine Acupuncture and MoxibustionTianjinChina
| |
Collapse
|
4
|
Lai G, Shen J, Hu Y, Yang F, Zhang C, Le D, Liu Q, Liang Y. LncRNA RNA ROR Aggravates Hypoxia/Reoxygenation-Induced Cardiomyocyte Ferroptosis by Targeting miR-769-5p/CBX7 Axis. Biochem Genet 2024; 62:3586-3604. [PMID: 38157079 DOI: 10.1007/s10528-023-10587-3] [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: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
Ferroptosis is a new way of cell death which is reported to participate in the pathology of myocardial ischemia-reperfusion (MI/R) injury, but it's mechanism remains unclear. The present investigation is to study the emerging role of long non-coding RNA (lncRNA) regulator of reprogramming (ROR) in cardiomyocyte ferroptosis after hypoxia/reoxygenation (H/R) administration. RT-qPCR and/or Western blot methods were performed to examine the gene/or protein levels, and CCK-8, ELISA, and DCFH-DA staining determined the cellular viability and ferroptosis. Dual-luciferase and RNA immunoprecipitation were applied to verify molecular interaction. LncRNA ROR and miR-769-5p were overexpressed and reduced in blood samples from MI patients and H/R-treated AC16 cells, respectively. Mechanistically, lncROR sponged to miR-769-5p, thus upregulating CBX7 expression. Functional experiments presented that lncRNA ROR silence mitigated H/R-stimulated inflammatory damage, oxidative stress, and ferroptosis in AC16 cells, whereas these roles could be reversed by co-downregulation of miR-769-5p or co-overexpression of CBX7. These data uncovered that lncRNA ROR prevented against H/R-induced cardiomyocyte ferroptosis by modulating miR-769-5p/CBX7 signaling, emphasizing the therapeutic value of lncRNA ROR in MI/R injury.
Collapse
Affiliation(s)
- Guorong Lai
- Department of Pain Management, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Jie Shen
- Department of Rehabilitation, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yanhui Hu
- Department of Anesthesiology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Fan Yang
- Department of Pain Management, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Chao Zhang
- Department of Pain Management, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Dongsheng Le
- Department of Pain Management, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Qin Liu
- Department of Anesthesiology, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, People's Republic of China
| | - Yingping Liang
- Department of Pain Management, The 2nd Affiliated Hospital, Jiangxi Medical College, Nanchang University, No.1 Minde Road, Nanchang, 330006, Jiangxi Province, People's Republic of China.
| |
Collapse
|
5
|
Pissas G, Tziastoudi M, Divani M, Poulianiti C, Konsta MAP, Lykotsetas E, Liakopoulos V, Stefanidis I, Eleftheriadis T. Malate dehydrogenase-2 inhibition shields renal tubular epithelial cells from anoxia-reoxygenation injury by reducing reactive oxygen species. J Biochem Mol Toxicol 2024; 38:e23854. [PMID: 39287333 DOI: 10.1002/jbt.23854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/19/2024]
Abstract
Ischemia-reperfusion (I-R) injury is the most common cause of acute kidney injury. In experiments involving primary human renal proximal tubular epithelial cells (RPTECs) exposed to anoxia-reoxygenation, we explored the hypothesis that mitochondrial malate dehydrogenase-2 (MDH-2) inhibition redirects malate metabolism from the mitochondria to the cytoplasm, towards the malate-pyruvate cycle and reversed malate-aspartate shuttle. Colorimetry, fluorometry, and western blotting showed that MDH2 inhibition accelerates the malate-pyruvate cycle enhancing cytoplasmic NADPH, thereby regenerating the potent antioxidant reduced glutathione. It also reversed the malate-aspartate shuttle and potentially diminished mitochondrial reactive oxygen species (ROS) production by transferring electrons, in the form of NADH, from the mitochondria to the cytoplasm. The excessive ROS production induced by anoxia-reoxygenation led to DNA damage and protein modification, triggering DNA damage and unfolded protein response, ultimately resulting in apoptosis and senescence. Additionally, ROS induced lipid peroxidation, which may contribute to the process of ferroptosis. Inhibiting MDH-2 proved effective in mitigating ROS overproduction during anoxia-reoxygenation, thereby rescuing RPTECs from death or senescence. Thus, targeting MDH-2 holds promise as a pharmaceutical strategy against I-R injury.
Collapse
Affiliation(s)
- Georgios Pissas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Maria Tziastoudi
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Maria Divani
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Christina Poulianiti
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | - Evangelos Lykotsetas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Vasilios Liakopoulos
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | |
Collapse
|
6
|
Li Y, Zhou Y, Pei H, Li D. Disruption of BACH1 Protects AC16 Cardiomyocytes Against Hypoxia/Reoxygenation-Evoked Injury by Diminishing CDKN3 Transcription. Cardiovasc Toxicol 2024; 24:1105-1115. [PMID: 39060883 DOI: 10.1007/s12012-024-09900-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024]
Abstract
Reperfusion after myocardial infarction (MI) can lead to myocardial ischemia/reperfusion (I/R) damage. The transcription factor (TF) broad-complex, tramtrack, and bric-a-brac (BTB) and cap'n'collar (CNC) homology 1 (BACH1) is implicated in the injury. However, the downstream mechanisms of BACH1 in affecting myocardial hypoxia/reoxygenation (H/R) damage are still fully understood. AC16 cells were stimulated with H/R conditions to model cardiomyocytes under H/R. mRNA analysis was performed by quantitative real-time PCR. Protein levels were gauged by immunoblot analysis. The effect of BACH1/cyclin-dependent kinase inhibitor 3 (CDKN3) on H/R-evoked injury was assessed by measuring cell viability via Cell Counting Kit-8 (CCK-8), apoptosis (flow cytometry and caspase 3 activity), ferroptosis via Fe2+, glutathione (GSH), reactive oxygen species (ROS) and malondialdehyde (MDA) markers and inflammation cytokines interleukin-1beta (IL-1β) and tumor necrosis factor alpha (TNF-α). The BACH1/CDKN3 relationship was examined by chromatin immunoprecipitation (ChIP) experiment and luciferase assay. BACH1 was increased in MI serum and H/R-stimulated AC16 cardiomyocytes. Functionally, disruption of BACH1 mitigated H/R-evoked in vitro apoptosis, ferroptosis and inflammation of AC16 cardiomyocytes. Mechanistically, BACH1 activated CDKN3 transcription and enhanced CDKN3 protein expression in AC16 cardiomyocytes. Our rescue experiments validated that BACH1 disruption attenuated H/R-evoked AC16 cardiomyocyte apoptosis, ferroptosis and inflammation by downregulating CDKN3. Additionally, BACH1 disruption could activate the adenosine monophosphate-activated protein kinase (AMPK) signaling by downregulating CDKN3 in H/R-stimulated AC16 cardiomyocytes. Our study demonstrates that BACH1 activates CDKN3 transcription to induce H/R-evoked damage of AC16 cardiomyocytes partially via AMPK signaling.
Collapse
Affiliation(s)
- Yanping Li
- Department of Cardiovascular Medicine, Western Theater Command General Hospital, No. 270, Tianhui Road, Rongdu Avenue, Chengdu, 610083, China
| | - Yi Zhou
- Department of Clinic, Western Theater Command General Hospital, Chengdu, 610083, China
| | - Haifeng Pei
- Department of Cardiovascular Medicine, Western Theater Command General Hospital, No. 270, Tianhui Road, Rongdu Avenue, Chengdu, 610083, China
| | - De Li
- Department of Cardiovascular Medicine, Western Theater Command General Hospital, No. 270, Tianhui Road, Rongdu Avenue, Chengdu, 610083, China.
| |
Collapse
|
7
|
Zhou J, Luo W, Xie Z, Xia C, Zhao J. KLF4-induced upregulation of SOCS1 ameliorates myocardial ischemia/reperfusion injury by attenuating AC16 cardiomyocyte damage and enhancing M2 macrophage polarization. J Biochem Mol Toxicol 2024; 38:e23816. [PMID: 39185902 DOI: 10.1002/jbt.23816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/12/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
Reperfusion strategies, the standard therapy for acute myocardial infarction (AMI), may result in ischemia/reperfusion (I/R) damage. Suppressor of cytokine signaling1 (SOCS1) exerts a cardioprotective function in myocardial I/R damage. Here, we investigated epigenetic modulators that deregulate SOCS1 in cardiomyocytes under hypoxia/reoxygenation (H/R) conditions. Human AC16 cardiomyocytes were exposed to H/R conditions to generate a cell model of myocardial I/R damage. Expression of mRNA and protein was detected by quantitative PCR and western blot analysis, respectively. Cell migratory and invasive abilities were evaluated by transwell assay. Cell apoptosis and M2 macrophage polarization were assessed by flow cytometry. TNF-α, IL-1β, and IL-6 levels were examined by ELISA. The interaction of KLF4 with SOCS1 was verified by chromatin immunoprecipitation and luciferase assays. SOCS1 and transcription factor KLF4 protein levels were underexpressed by 75% and 57%, respectively, in H/R-exposed AC16 cardiomyocytes versus control cells. Under H/R conditions, forced SOCS1 expression (2.7 times) induced cell migration (2.2 times) and invasion (1.9 times) and hindered cell apoptosis (by 45%) of AC16 cardiomyocytes as well as enhanced M2 macrophage polarization (4.6 times). Mechanistically, KLF4 upregulation promoted SOCS1 transcription (2.6 times) and expression (2.6 times) by binding to the SOCS1 promoter. Decrease of SOCS1 (by 51%) reversed the effects of KLF4 upregulation on cardiomyocyte migration, invasion and apoptosis, and M2 macrophage polarization under H/R conditions. Additionally, SOCS1 and KLF4 were underexpressed by 56% and 63%, respectively, in AMI serum. Our study indicates that KLF4-induced upregulation of SOCS1 can attenuate H/R-triggered apoptosis of AC16 cardiomyocytes and enhance M2 macrophage polarization.
Collapse
Affiliation(s)
- Jiming Zhou
- Department of Cardiology, The First Affiliated Hospital,Hengyang Medical School, University of South China, Hengyang, China
| | - Wei Luo
- Department of Cardiology, The First Affiliated Hospital,Hengyang Medical School, University of South China, Hengyang, China
| | - Zhong Xie
- Department of Cardiology, The First Affiliated Hospital,Hengyang Medical School, University of South China, Hengyang, China
| | - Chunchen Xia
- Department of Cardiology, The First Affiliated Hospital,Hengyang Medical School, University of South China, Hengyang, China
| | - Junbi Zhao
- Department of Cardiology, The First Affiliated Hospital,Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
8
|
Sivakumar B, Kurian GA. Investigating the temporal link between PM 2.5 exposure and acceleration of myocardial ischemia reperfusion injury: Emphasizing the hazardous presence of metals in inhaled air. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 355:124113. [PMID: 38734051 DOI: 10.1016/j.envpol.2024.124113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/12/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
Exposure to PM2.5 is widely acknowledged to induce cardiotoxic effects, leading to decreased myocardial tolerance to revascularization procedures and subsequent ischemia reperfusion injury (IR). However, the temporal relationship between PM2.5 exposure and vulnerability to IR, along with the underlying mechanisms, remains unclear and is the focus of this study. Female Wistar rats were exposed to PM2.5 at a concentration of 250 μg/m³ for 3 h daily over varying durations (7, 14, and 21 days), followed by IR induction. Our results demonstrated a significant increase in cardiac injury, as evidenced by increased infarct size and elevated cardiac injury markers, starting from day 14 of PM2.5 exposure, accompanied by declined cardiac function. These adverse effects were associated with apoptosis and impaired mitochondrial function, including reduced bioenergetics, mitochondrial DNA copy number and quality control mechanisms, along with inactivation of the PI3K/AKT/AMPK signalling pathways. Furthermore, analysis of myocardial tissue revealed elevated metal accumulation, particularly within mitochondria. Chelation of PM2.5 -associated metals using EDTA significantly mitigated the toxic effects on cardiac IR pathology, as confirmed in both rat myocardium and H9c2 cells. These findings suggest that metals in PM2.5 play a crucial role in inducing cardiotoxicity, impairing myocardial resilience to stress through mitochondrial accumulation and dysfunction.
Collapse
Affiliation(s)
- Bhavana Sivakumar
- Vascular Biology lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India
| | - Gino A Kurian
- Vascular Biology lab, School of Chemical and Biotechnology, SASTRA Deemed University, Thanjavur, Tamil Nadu, India; School of Chemical and Biotechnology, SASTRA Deemed University, Tirumalaisamudram, Thanjavur-613401, Tamil Nadu, India.
| |
Collapse
|
9
|
Fudim M, Konecny F, Heuring JJ, Durst CA, Fain ES, Patel MR. Left Ventricular Unloading Using Intra-aortic Entrainment Pumping Before Reperfusion Reduces Post-AMI Infarct Size. J Card Fail 2024:S1071-9164(24)00316-6. [PMID: 39147311 DOI: 10.1016/j.cardfail.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 07/10/2024] [Accepted: 07/18/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND Anterior myocardial infarction standard of care prioritizes swift coronary reperfusion. Recent studies show left ventricular (LV) unloading with transvalvular axial flow pumps for 30 minutes before reperfusion (vs immediate reperfusion) decreases 28-day infarct size. Intra-aortic entrainment pumping, using hardware located away from the heart to provide support throughout the cardiac cycle, decreases effective systemic vascular resistance and augments visceral blood flow and pressure, and may reproduce this benefit with a decreased risk. This study characterized the hemodynamic effects of unloading before and during reperfusion using intra-aortic entrainment pumping and investigated whether unloading decreased anterior myocardial infarction scar size. METHODS AND RESULTS Yorkshire swine were subjected to 90 minutes of left anterior descending artery balloon occlusion and randomly assigned to immediate reperfusion (n = 6) vs 30 minutes unloading before reperfusion followed by 120 minutes of further unloading (n = 7). Unloading was achieved using percutaneous entrainment pumping in the descending aorta. The anterior myocardial infarction model matches that used in recent transvalvular pumping studies. Mortality before randomization was 22%. After randomization, mortality was 36% for immediate reperfusion and 0% for unloading. Unloading showed immediate hemodynamic benefit that increased through reperfusion and continued support, leading to distinct differences in cardiac function between groups after 30 minutes of reperfusion. Unloading increased stroke volume and cardiac efficiency at this timepoint relative to preocclusion baseline and reduced 28-day LV scar size by 37%-45%. CONCLUSIONS We present the first preclinical data showing extracardiac LV unloading before coronary reperfusion using intra-aortic entrainment pumping decreases 28-day infarct size. Extracardiac unloading to decrease LV scar size may provide an alternative to transvalvular pumping with potential advantages, including reduced risk.
Collapse
Affiliation(s)
- Marat Fudim
- Duke University Medical Center, Durham, North Carolina
| | - Filip Konecny
- Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | | | | | | | | |
Collapse
|
10
|
Wang H, Rouhi N, Slotabec LA, Seale BC, Wen C, Filho F, Adenawoola MI, Li J. Myeloid Cells in Myocardial Ischemic Injury: The Role of the Macrophage Migration Inhibitory Factor. Life (Basel) 2024; 14:981. [PMID: 39202723 PMCID: PMC11355293 DOI: 10.3390/life14080981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/23/2024] [Accepted: 07/26/2024] [Indexed: 09/03/2024] Open
Abstract
Ischemic heart disease, manifesting as myocardial infarction (MI), remains the leading cause of death in the western world. Both ischemia and reperfusion (I/R) cause myocardial injury and result in cardiac inflammatory responses. This sterile inflammation in the myocardium consists of multiple phases, involving cell death, tissue remodeling, healing, and scar formation, modulated by various cytokines, including the macrophage migration inhibitory factor (MIF). Meanwhile, different immune cells participate in these phases, with myeloid cells acting as first responders. They migrate to the injured myocardium and regulate the initial phase of inflammation. The MIF modulates the acute inflammatory response by affecting the metabolic profile and activity of myeloid cells. This review summarizes the role of the MIF in regulating myeloid cell subsets in MI and I/R injury and discusses emerging evidence of metabolism-directed cellular inflammatory responses. Based on the multifaceted role of the MIF affecting myeloid cells in MI or I/R, the MIF can be a therapeutic target to achieve metabolic balance under pathology and alleviate inflammation in the heart.
Collapse
Affiliation(s)
- Hao Wang
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Nadiyeh Rouhi
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Lily A. Slotabec
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS 39216, USA
| | - Blaise C. Seale
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Changhong Wen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Fernanda Filho
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Michael I. Adenawoola
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, MS 39216, USA; (H.W.); (N.R.); (L.A.S.); (B.C.S.); (C.W.); (F.F.); (M.I.A.)
- G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
11
|
Artimovič P, Špaková I, Macejková E, Pribulová T, Rabajdová M, Mareková M, Zavacká M. The ability of microRNAs to regulate the immune response in ischemia/reperfusion inflammatory pathways. Genes Immun 2024; 25:277-296. [PMID: 38909168 PMCID: PMC11327111 DOI: 10.1038/s41435-024-00283-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/24/2024]
Abstract
MicroRNAs play a crucial role in regulating the immune responses induced by ischemia/reperfusion injury. Through their ability to modulate gene expression, microRNAs adjust immune responses by targeting specific genes and signaling pathways. This review focuses on the impact of microRNAs on the inflammatory pathways triggered during ischemia/reperfusion injury and highlights their ability to modulate inflammation, playing a critical role in the pathophysiology of ischemia/reperfusion injury. Dysregulated expression of microRNAs contributes to the pathogenesis of ischemia/reperfusion injury, therefore targeting specific microRNAs offers an opportunity to restore immune homeostasis and improve patient outcomes. Understanding the complex network of immunoregulatory microRNAs could provide novel therapeutic interventions aimed at attenuating excessive inflammation and preserving tissue integrity.
Collapse
Affiliation(s)
- Peter Artimovič
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Ivana Špaková
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Ema Macejková
- Department of Vascular Surgery, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Timea Pribulová
- Department of Vascular Surgery, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Miroslava Rabajdová
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Mária Mareková
- Department of Medical and Clinical Biochemistry, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia
| | - Martina Zavacká
- Department of Vascular Surgery, Pavol Jozef Šafárik University in Košice, Faculty of Medicine, Košice, Slovakia.
| |
Collapse
|
12
|
Israni DK, Patel ML, Dodiya RK. Exploring the versatility of miRNA-128: a comprehensive review on its role as a biomarker and therapeutic target in clinical pathways. Mol Biol Rep 2024; 51:860. [PMID: 39068606 DOI: 10.1007/s11033-024-09822-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
MicroRNAs (miRNAs/ miRs) are short, noncoding RNAs, usually consisting of 18 to 24 nucleotides, that control gene expression after the process of transcription and have crucial roles in several clinical processes. This article seeks to provide an in-depth review and evaluation of the many activities of miR-128, accentuating its potential as a versatile biomarker and target for therapy; The circulating miR-128 has garnered interest because of its substantial influence on gene regulation and its simplicity in extraction. Several miRNAs, such as miR-128, have been extracted from circulating blood cells, cerebrospinal fluid, and plasma/serum. The miR-128 molecule can specifically target a diverse range of genes, enabling it to have intricate physiological impacts by concurrently regulating many interrelated pathways. It has a vital function in several biological processes, such as modulating the immune system, regulating brain plasticity, organizing the cytoskeleton, and inducing neuronal death. In addition, miR-128 modulates genes associated with cell proliferation, the cell cycle, apoptosis, plasma LDL levels, and gene expression regulation in cardiac development. The dysregulation of miR-128 expression and activity is associated with the development of immunological responses, changes in neural plasticity, programmed cell death, cholesterol metabolism, and heightened vulnerability to autoimmune illnesses, neuroimmune disorders, cancer, and cardiac problems; The paper highlights the importance of studying the consequences of miR-128 dysregulation in these specific locations. By examining the implications of miRNA-128 dysregulation in these areas, the article underscores its significance in diagnosis and treatment, providing a foundation for research and clinical applications.
Collapse
Affiliation(s)
- Dipa K Israni
- Department of Pharmacology, L.J. Institute of Pharmacy, LJ University, SG Highway, Sanand Cross-Road, Ahmedabad, Gujarat, 382210, India.
| | - Manish L Patel
- LJ Institute of Pharmacy, LJ University, Ahmedabad, Gujarat, India
| | - Rohinee K Dodiya
- Department of Pharmacology, L.J. Institute of Pharmacy, LJ University, SG Highway, Sanand Cross-Road, Ahmedabad, Gujarat, 382210, India
| |
Collapse
|
13
|
Yin L, Li L, Gao M, Qi Y, Xu L, Peng J. circMIRIAF aggravates myocardial ischemia-reperfusion injury via targeting miR-544/WDR12 axis. Redox Biol 2024; 73:103175. [PMID: 38795544 PMCID: PMC11140810 DOI: 10.1016/j.redox.2024.103175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/28/2024] Open
Abstract
Exploring and discovering novel circRNAs is one of the ways to develop innovative drugs for the diagnosis and treatment of myocardial ischemia-reperfusion injury (MI/RI). In the work, some dysregulated circRNAs were found by microarray screening analysis in AC16 cells, and hsa_circRNA_104852 named circMIRIAF was screened, which was up-regulated in AC16 cells damaged by hypoxia-reoxygenation injury (H/RI). The comprehensive analysis of ceRNA network revealed the potential relationship of circMIRIAF/miR-544/WDR12. Then, the results of interaction research confirmed that circMIRIAF acted as sponge of miR-544 to positively regulate WDR12 protein expression. Further, the validation results indicate that miR-544 silencing increased the expression of WDR12, and WDR12 activated Notch1 signal to aggravate H/RI of AC16 cells and MI/RI of mice via regulating oxidative stress and inflammation. Furthermore, silencing circMIRIAF caused the decreased circMIRIAF levels and the increased miR-544 levels in cardiomyocytes, while excessive miR-544 inhibited WDR12 expression to alleviate the disorder. On the contrary, excessive circMIRIAF increased WDR12 expression by adsorbing miR-544 to exacerbate H/RI in AC16 cells. In addition, circMIRIAF siRNA reversed the aggravation of H/RI in cells caused by WDR12 overexpression. Overall, circMIRIAF can serve as a drug target or treating MI/RI, and circMIRIAF could sponge miR-544 and enhance WDR12 expression to aggravate MI/RI, which may provide a novel therapeutic strategy for MI/RI treatment.
Collapse
Affiliation(s)
- Lianhong Yin
- Department of Pharmaceutical Analysis, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Lili Li
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Meng Gao
- Department of Pharmaceutical Analysis, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Yan Qi
- Department of Pharmaceutical Analysis, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Lina Xu
- Department of Pharmaceutical Analysis, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China.
| | - Jinyong Peng
- Department of Pharmaceutical Analysis, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China; College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Hubei Shizhen Laboratory, Wuhan, 430065, China.
| |
Collapse
|
14
|
Yang Q, Murata K, Ikeda T, Minatoya K, Masumoto H. miR-124-3p downregulates EGR1 to suppress ischemia-hypoxia reperfusion injury in human iPS cell-derived cardiomyocytes. Sci Rep 2024; 14:14811. [PMID: 38926457 PMCID: PMC11208498 DOI: 10.1038/s41598-024-65373-x] [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: 02/04/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024] Open
Abstract
Ischemic heart diseases are a major global cause of death, and despite timely revascularization, heart failure due to ischemia-hypoxia reperfusion (IH/R) injury remains a concern. The study focused on the role of Early Growth Response 1 (EGR1) in IH/R-induced apoptosis in human cardiomyocytes (CMs). Human induced pluripotent stem cell (hiPSC)-derived CMs were cultured under IH/R conditions, revealing higher EGR1 expression in the IH/R group through quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting (WB). Immunofluorescence analysis (IFA) showed an increased ratio of cleaved Caspase-3-positive apoptotic cells in the IH/R group. Using siRNA for EGR1 successfully downregulated EGR1, suppressing cleaved Caspase-3-positive apoptotic cell ratio. Bioinformatic analysis indicated that EGR1 is a plausible target of miR-124-3p under IH/R conditions. The miR-124-3p mimic, predicted to antagonize EGR1 mRNA, downregulated EGR1 under IH/R conditions in qRT-PCR and WB, as confirmed by IFA. The suppression of EGR1 by the miR-124-3p mimic subsequently reduced CM apoptosis. The study suggests that treatment with miR-124-3p targeting EGR1 could be a potential novel therapeutic approach for cardioprotection in ischemic heart diseases in the future.
Collapse
Affiliation(s)
- Qiaoke Yang
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kozue Murata
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Tadashi Ikeda
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kenji Minatoya
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hidetoshi Masumoto
- Department of Cardiovascular Surgery, Graduate School of Medicine, Kyoto University, 54 Kawara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
- Clinical Translational Research Program, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima Minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| |
Collapse
|
15
|
Lin L, Wang L, Li A, Li Y, Gu X. CircDiaph3 aggravates H/R-induced cardiomyocyte apoptosis and inflammation through miR-338-3p/SRSF1 axis. J Bioenerg Biomembr 2024; 56:235-245. [PMID: 38613636 PMCID: PMC11116235 DOI: 10.1007/s10863-023-09992-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 10/24/2023] [Indexed: 04/15/2024]
Abstract
Acute myocardial infarction (AMI) is one of the most prevalent cardiovascular diseases, accounting for a high incidence rate and high mortality worldwide. Hypoxia/reoxygenation (H/R)-induced myocardial cell injury is the main cause of AMI. Several studies have shown that circular RNA contributes significantly to the pathogenesis of AMI. Here, we established an AMI mouse model to investigate the effect of circDiaph3 in cardiac function and explore the functional role of circDiaph3 in H/R-induced cardiomyocyte injury and its molecular mechanism. Bioinformatics tool and RT-qPCR techniques were applied to detect circDiaph3 expression in human patient samples, heart tissues of AMI mice, and H/R-induced H9C2 cells. CCK-8 was used to examine cell viability, while annexin-V/PI staining was used to assess cell apoptosis. Myocardial reactive oxygen species (ROS) levels were detected by immunofluorescence. Western blot was used to detect the protein expression of anti-apoptotic Bcl-2 while pro-apoptotic Bax and cleaved-Caspase-3. Furthermore, ELISA was used to detect inflammatory cytokines production. While bioinformatics tool and RNA pull-down assay were used to verify the interaction between circDiaph3 and miR-338-3p. We found that circDiaph3 expression was high in AMI patients and mice, as well as in H/R-treated H9C2 cells. CircDiaph3 silencing ameliorated apoptosis and inflammatory response of cardiomyocytes in vivo. Moreover, the knockdown of cirDiaph3 mitigated H/R-induced apoptosis and the release of inflammatory mediators like IL-1β, IL-6, and TNF-α in H9C2 cells. Mechanistically, circDiaph3 induced cell apoptosis and inflammatory responses in H/R-treated H9C2 cells by sponging miR-338-3p. Overexpressing miR-338-3p in H/R-treated cells prominently reversed circDiaph3-induced effects. Notably, miR-338-3p inhibited SRSF1 expression in H/R-treated H9C2 cells. While overexpressing SRSF1 abrogated miR-338-3p-mediated alleviation of apoptosis and inflammation after H/R treatment. To summarize, circDiaph3 aggravates H/R-induced cardiomyocyte apoptosis and inflammation through the miR-338-3p/SRSF1 axis. These findings suggest that the circDiaph3/miR-338-3pp/SRSF1 axis could be a potential therapeutic target for treating H/R-induced myocardial injury.
Collapse
Affiliation(s)
- Lin Lin
- Department of Cardiovascular Medicine, PLA Southern Theater Command General Hospital, 11 Liuhua Road, Guangzhou, 510000, China
| | - Li Wang
- Department of Emergency, PLA Southern Theater Command General Hospital, 11 Liuhua Road, Guangzhou, 510000, China
| | - Aimin Li
- Department of Cardiovascular Medicine, PLA Southern Theater Command General Hospital, 11 Liuhua Road, Guangzhou, 510000, China
| | - Yanzhuo Li
- Department of Cardiovascular Medicine, PLA Southern Theater Command General Hospital, 11 Liuhua Road, Guangzhou, 510000, China
| | - Xiaolong Gu
- Department of Cardiovascular Medicine, PLA Southern Theater Command General Hospital, 11 Liuhua Road, Guangzhou, 510000, China.
| |
Collapse
|
16
|
Wu J, Cui Y, Ding W, Zhang J, Wang L. The protective effect of Macrostemonoside T from Allium macrostemon Bunge against Isoproterenol-Induced myocardial injury via the PI3K/Akt/mTOR signaling pathway. Int Immunopharmacol 2024; 133:112086. [PMID: 38642441 DOI: 10.1016/j.intimp.2024.112086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/31/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024]
Abstract
Myocardial injury (MI) signifies a pathological aspect of cardiovascular diseases (CVDs) such as coronary artery disease, diabetic cardiomyopathy, and myocarditis. Macrostemonoside T (MST) has been isolated from Allium macrostemon Bunge (AMB), a key traditional Chinese medicine (TCM) used for treating chest stuffiness and pains. Although MST has demonstrated considerable antioxidant activity in vitro, its protective effect against MI remains unexplored. To investigate MST's effects in both in vivo and in vitro models of isoproterenol (ISO)-induced MI and elucidate its underlying molecular mechanisms. This study established an ISO-induced MI model in rats and assessed H9c2 cytotoxicity to examine MST's impact on MI. Various assays, including histopathological staining, TUNEL staining, immunohistochemical staining, DCFH-DA staining, JC-1 staining, ELISA technique, and Western blot (WB), were utilized to explore the potential molecular mechanisms of MI protection. In vivo experiments demonstrated that ISO caused myocardial fiber disorders, elevated cardiac enzyme levels, and apoptosis. However, pretreatment with MST significantly mitigated these detrimental changes. In vitro experiments revealed that MST boosted antioxidant enzyme levels and suppressed malondialdehyde (MDA) production in H9c2 cells. Concurrently, MST inhibited ISO-induced reactive oxygen species (ROS) production and mitigated the decline in mitochondrial membrane potential, thereby reducing the apoptosis rate. Moreover, pretreatment with MST elevated the expression levels of p-PI3K, p-Akt, and p-mTOR, indicating activation of the PI3K/Akt/mTOR signaling pathway and consequent protection against MI. MST attenuated ISO-induced MI in rats by impeding apoptosis through activation of the PI3K/Akt/mTOR signaling pathway. This study presents potential avenues for the development of precursor drugs for CVDs.
Collapse
Affiliation(s)
- Jianfa Wu
- Department of Traditional Chinese Medicine, College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Ying Cui
- Department of Traditional Chinese Medicine, College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Weixing Ding
- Department of Traditional Chinese Medicine, College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Jing Zhang
- Department of Traditional Chinese Medicine, College of Traditional Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China.
| | - Lulu Wang
- School of Medicine, Changchun Sci-Tech University, Changchun 130600, China.
| |
Collapse
|
17
|
Behzad H, Asham H, Beheshtirouy S, Mashayekhi S, Jafari N, Entezari-Maleki T. Sodium-glucose cotransporter-2 inhibitors in individuals with ischemia reperfusion injury: A systematic review. Perfusion 2024:2676591241257371. [PMID: 38813587 DOI: 10.1177/02676591241257371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Background: The effects of sodium-glucose cotransporter-2 (SGLT-2) inhibitors on ischemia reperfusion injury (IRI) is a novel concept and only limited number of animals studies have yet been investigated. We aimed to perform a systematic review of literature to explore the clinical studies which investigated the effects of SGLT-2 inhibitors on myocardial IRI setting.Methods: We searched MEDLINE, Embase, and Cochrane Library from inception until December 7th, 2023. ClinicalTrials.gov was also explored for ongoing studies. Two authors independently conducted the literature search, examined the studies, and evaluated the eligibility criteria. Any disagreements or uncertainties were resolved by the corresponding author. The search strategy followed the PICO process (Population, Intervention, Comparison, and Outcome) and Emtree was used to select relevant keywords.Results: Of 220 articles identified from the literature research, five articles were included in the study, of which three studies lately were retracted. The remaining studies included 1229 participants, with 209 receiving SGLT-2 inhibitors and 1090 not receiving them. All of the participants were diabetic patients admitted with acute myocardial infarction (AMI), undergoing percutaneous coronary intervention (PCI). The results demonstrated that the use of SGLT-2 inhibitors is associated with lower troponin levels, and higher rates of ST resolution. The results of the studies also showed smaller infarct sizes, lower inflammatory biomarkers and improved left ventricular function at discharge among SGLT-2 inhibitor users.Conclusion: In line with in vivo and ex vivo findings, the results of this systematic review supported benefits of SGLT-2 inhibitors in IRI through reducing infarct size and inflammatory biomarkers. However, further clinical trials are warranted to provide robust evidence.
Collapse
Affiliation(s)
- Hossein Behzad
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hila Asham
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samineh Beheshtirouy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sina Mashayekhi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Jafari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari-Maleki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
18
|
Rong W, Shi Q, Yang Y, Su W, Li M, Qin M, Bai S, Zhu Q, Wang A. Fructus choerospondiatis: A comprehensive review of its traditional uses, chemical composition, pharmacological activities, and clinical studies. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117696. [PMID: 38171468 DOI: 10.1016/j.jep.2023.117696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/11/2023] [Accepted: 12/30/2023] [Indexed: 01/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fructus Choerospondiatis is the dried and mature fruit of Choerospondias axillaris (Roxb.) Burtt et Hill. It has been used for a long time in Tibetan and Mongolian medicine, first recorded in the ancient Tibetan medicine book "Medicine Diagnosis of the King of the Moon" in the early 8th century. Fructus Choerospondiatis shows multiple pharmacological activities, especially in treating cardiovascular diseases. AIM OF THIS REVIEW This paper reviews the progress in research on the botanical characteristics, traditional uses, chemical constituents, pharmacological activity, clinical studies, and quality control of Fructus Choerospondiatis. This review aims to summarize current research and provide a reference for further development and utilization of Fructus Choerospondiatis resources. METHOD The sources for this review include the Pharmacopeia of the People's Republic of China (2020), theses, and peer-reviewed papers (in both English and Chinese). Theses and papers were downloaded from electronic databases including Web of Science, PubMed, SciFinder, Scholar, Springer, and China National Knowledge Infrastructure.The search terms used were "Choerospondias axillaris", "C. axillaris", "Choerospondias axillaris (Roxb.) Burtt et Hill", "Fructus choerospondiatis", "Guangzao", "Lapsi", and "Lupsi". RESULTS Fructus Choerospondiatis contains polyphenols, organic acids, amino acids, fatty acids, polysaccharides, and other chemical components. These ingredients contribute to its diverse pharmacological activities such as antioxidant activity, protection against myocardial ischemia-reperfusion injury, anti-myocardial fibrosis, heart rhythm regulation, anti-tumor, liver protection, and immunity enhancement. It also affects the central nervous system, with the ability to repair damaged nerve cells. CONCLUSION Fructus Choerospondiatis, with its various chemical compositions and pharmacological activities, is a promising medicinal resource. However, it remains under-researched, particularly in pharmacodynamic material basis and quality control. These areas require further exploration by researchers in the future.
Collapse
Affiliation(s)
- Weiwei Rong
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, 226001, Jiangsu, China
| | - Qilin Shi
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Yuru Yang
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Weiyi Su
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Mingna Li
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Minni Qin
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Shuang Bai
- Livzon Pharmaceutical Group Inc., Zhuhai, 519000, Guangdong, China
| | - Qing Zhu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, 226001, Jiangsu, China.
| | - Andong Wang
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China; Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
19
|
Liu Q, Lai G, Hu Y, Yang F, Zhang C, Le D, Deng F, Xing X, Tang B, Jie H, Liang Y, Lei E. CircRbms1 fosters MST1 mRNA and protein levels to motivate myocardial ischaemia-reperfusion injury via autophagic status. ESC Heart Fail 2024; 11:1205-1217. [PMID: 38288506 DOI: 10.1002/ehf2.14673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/29/2023] [Accepted: 12/18/2023] [Indexed: 03/28/2024] Open
Abstract
AIMS Acute myocardial infarction (MI) is a significant contributor to death in individuals diagnosed with coronary heart disease on a worldwide level. The specific mechanism by which circRbms1 contributes to the damage caused by myocardial ischaemia-reperfusion (I/R) is not well understood. The primary aim of this study was to examine the role of circRbms1 and its associated mechanisms in the setting of I/R injury. METHODS AND RESULTS An in vivo MI mice model and an in vitro MI cell model was established. The expression levels were detected using quantitative real-time PCR (qRT-PCR) and western blot. Cellular proliferation, apoptosis, pyroptosis, and autophagy were detected by immunostaining, immunohistochemistry, western blot, and transmission electron microscopy (TEM). Dual-luciferase reporter assay, RNA pull-down assay, and RIP assay were performed to validate the molecular interactions. CircRbms1 was up-regulated in A/R-induced HCMs and acted as a sponge for miR-142-3p, thereby targeting MST1. CircRbms1 could improve stability of MST1 by recruiting IGF2BP2 (all P < 0.05). CircRbms1 knockout reduced cell pyroptosis, improved autophagy and proliferation level in A/R-induced HCMs (all P < 0.05). CircRbms1 knockout alleviated cardiac dysfunction and cell pyroptosis and enhanced autophagy and proliferation in mice through the miR-142-3p/MST1 axis. CONCLUSIONS CircRbms1 inhibited the miR-142-3p/MST1 axis and played a protective role in myocardial I/R injury. It may provide a new therapeutic target for I/R heart injury.
Collapse
Affiliation(s)
- Qin Liu
- Department of Anesthesiology, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Guorong Lai
- Department of Pain Management, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanhui Hu
- Department of Anesthesiology, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fan Yang
- Department of Pain Management, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Chao Zhang
- Department of Pain Management, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dongsheng Le
- Department of Pain Management, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Fumou Deng
- Department of Anesthesiology, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xianliang Xing
- Department of Anesthesiology, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Binquan Tang
- Department of Anesthesiology, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Huanhuan Jie
- Department of Anesthesiology, Ganzhou People's Hospital, Ganzhou, China
| | - Yingping Liang
- Department of Pain Management, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Enjun Lei
- Department of Anesthesiology, First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Liu WL, Wu LT, Wang JL, Sun J, Cheng XR, Zhou ZH, Guan JX, Wang YL, Meng ZY. Effect of PCI on ophthalmic artery hemodynamics in patients with acute coronary syndrome. Front Med (Lausanne) 2024; 11:1367900. [PMID: 38500953 PMCID: PMC10944952 DOI: 10.3389/fmed.2024.1367900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/19/2024] [Indexed: 03/20/2024] Open
Abstract
Purpose We aimed to explore the effects of percutaneous coronary intervention (PCI) on the ophthalmic artery (OA) hemodynamics in patients with acute coronary syndrome (ACS). Methods A total of 73 participants (Group0: healthy controls, Group1: Patients with ACS underwent PCI < 3 months, Group2: Patients with ACS underwent PCI ≥ 3 months) were enrolled. Computed tomographic angiography images were used to construct three-dimensional models of participants' OAs. Numerical simulations based on computational fluid dynamics were used to acquire hemodynamic parameters. Results The angle between the OA and internal carotid artery in Group2 was significantly larger compared with Group0 and Group1 (P = 0.003 and P = 0.044). Hemodynamic simulation showed a significantly slower OA blood velocity in Group1 than in the control (P < 0.001) and Group2 (P = 0.033). Lower wall shear stress was found in Group1 than that in control (P = 0.040). Patients after PCI had a higher wall pressure than healthy controls (P = 0.012 and P = 0.004). Mass flow ratios were decreased in Group1 and Group2 (P = 0.021 and P = 0.002). The hemodynamic parameters of OA were correlated with several clinical indicators. Conclusions The OA blood flow velocity of patients with ACS after PCI initially slowed down, which increased the risk of plaque formation, and then showed an increasing trend. There was a correlation between OA hemodynamic parameters and clinical indexes related to cardiac stress. Ischemia-reperfusion injury and changes in blood flow status after PCI may affect OA morphology and hemodynamics, leading to ocular lesions. Trial registration ChiCTR2100050428.
Collapse
|
21
|
Li H, Bu L, Sun X, Chu X, Xue Y, Zhang M, Shi J, Liu Y, Guan S, Han X, Wang H. Mechanistic investigation of the ameliorative effect of liquiritin on hypoxia/reoxygenation‑induced cardiomyocyte injury based on network pharmacology and in vitro validation. Exp Ther Med 2024; 27:117. [PMID: 38361515 PMCID: PMC10867724 DOI: 10.3892/etm.2024.12405] [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: 09/10/2023] [Accepted: 12/18/2023] [Indexed: 02/17/2024] Open
Abstract
Liquiritin (LIQ) is a flavonoid known for its cardioprotective properties, extracted from Glycyrrhiza uralensis Fisch. The purpose of the present study was to investigate the protective mechanism of LIQ against hypoxia/reoxygenation (H/R) injury through in vitro experiments, with the goal of enhancing its pharmacological effects. Initially, network pharmacology was employed to explore the targets and mechanisms of LIQ. Subsequently, an in vitro H/R model was established using H9c2 cells. Potential targets for LIQ and myocardial ischemia-reperfusion injury (MIRI) were identified through online databases. The STRING, Cytoscape and DAVID databases were used to extract intersecting targets and mechanisms. In vitro experiments were conducted to validate these findings, assessing cardiac enzymes, oxidative stress indicators, mitochondrial fluorescence, apoptotic fluorescence, inflammation and related protein expression. The network pharmacological analysis revealed that the protective effects of LIQ on MIRI involve oxidative stress, inflammation and apoptosis. The results of in vitro experimental validation demonstrated that LIQ significantly reduced the activities of lactated dehydrogenase and creatine kinase isoenzyme-MB (P<0.05 or 0.01), as well as the level of malondialdehyde (P<0.01). It also inhibited the production of reactive oxygen species (P<0.01), the release of inflammatory factors (P<0.05 or 0.01) and apoptosis (P<0.01). By contrast, the LIQ pre-treatment group exhibited a significant increase in mitochondrial membrane potential level (P<0.05 or 0.01) and the activities of antioxidant enzymes superoxide dismutase, catalase and glutathione peroxidase (P<0.05 or 0.01). Furthermore, LIQ reduced the protein expressions of TNF-α receptor type 1 (TNFR1) and MMP9, along with the level of NF-κB phosphorylation (P<0.05 or 0.01). In conclusion, LIQ mitigated H/R-induced cardiomyocyte injury through mechanisms that may involve antioxidants, anti-apoptotic effects, protection against mitochondrial damage and suppression of inflammatory levels. These effects are achieved via inhibition of the TNFR1/NF-κB/MMP9 pathway.
Collapse
Affiliation(s)
- Haoying Li
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Linlin Bu
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xiaoqi Sun
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Xi Chu
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yucong Xue
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Muqing Zhang
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050000, P.R. China
| | - Jing Shi
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, P.R. China
| | - Yanshuang Liu
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
- College of Integrative Medicine, Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shengjiang Guan
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050000, P.R. China
| | - Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| | - Hongfang Wang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei 050200, P.R. China
| |
Collapse
|
22
|
Lee H, Kim W, Song MY, Kim DH, Jung HS, Kim W, Choi S. One-Stop Plasmonic Nanocube-Excited SERS Immunoassay Platform of Multiple Cardiac Biomarkers for Rapid Screening and Progressive Tracing of Acute Myocardial Infarction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304999. [PMID: 37821412 DOI: 10.1002/smll.202304999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/11/2023] [Indexed: 10/13/2023]
Abstract
Rapid and precise acute myocardial infarction (AMI) diagnosis is essential for preventing patient death. In addition, the complementary roles of creatine kinase muscle brain (CK-MB) and cardiac troponin I (cTnI) cardiac biomarkers in the early and late stages of AMI demand their simultaneous detection, which is difficult to implement using conventional fluorescence and electrochemical technologies. Here, a nanotechnology-based one-stop immuno-surface-enhanced Raman scattering (SERS) detection platform is reported for multiple cardiac indicators for the rapid screening and progressive tracing of AMI events. Optimal SERS is achieved using optical property-based, excitation wavelength-optimized, and high-yield anisotropic plasmonic gold nanocubes. Optimal immunoassay reaction efficiencies are achieved by increasing immobilized antibodies. Multiple simultaneous detection strategies are implemented by incorporating two different Raman reports with narrow wavenumbers corresponding to two indicators and by establishing a computational SERS mapping process to accurately detect their concentrations, irrespective of multiple enzymes in the human serum. The SERS platform precisely estimated AMI onset and progressive timing in human serum and made rapid AMI identification feasible using a portable Raman spectrometer. This integrated platform is hypothesized to significantly contribute to emergency medicine and forensic science by providing timely treatment and observation.
Collapse
Affiliation(s)
- Hyerin Lee
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, 02447, Seoul, South Korea
| | - Wansun Kim
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, 02447, Seoul, South Korea
| | - Min-Young Song
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University, 02447, Seoul, South Korea
| | - Dong-Ho Kim
- Department of Nano-Bio Convergence, Korea Institute of Materials Science (KIMS), 51508, Gyeongnam, South Korea
| | - Ho Sang Jung
- Department of Nano-Bio Convergence, Korea Institute of Materials Science (KIMS), 51508, Gyeongnam, South Korea
- School of Convergence Science and Technology, Medical Science and Engineering, POSTECH, Kyungbuk, 37673, South Korea
| | - Weon Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University, 02447, Seoul, South Korea
| | - Samjin Choi
- Department of Biomedical Engineering, College of Medicine, Kyung Hee University, 02447, Seoul, South Korea
| |
Collapse
|
23
|
Chen W, Ma M, Song Y, Hua Y, Jia H, Liu J, Wang Y. Exercise Attenuates Myocardial Ischemia-Reperfusion Injury by Regulating Endoplasmic Reticulum Stress and Mitophagy Through M 2 Acetylcholine Receptor. Antioxid Redox Signal 2024; 40:209-221. [PMID: 37294203 DOI: 10.1089/ars.2022.0168] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Aims: Adaptive changes in the heart by exercise have been shown to reduce the risk of cardiovascular disease, and M2 Acetylcholine receptor (M2AChR), a receptor abundantly present on cardiac parasympathetic nerves, is closely associated with the development of cardiovascular disease. The present study intends to investigate whether exercise can regulate endoplasmic reticulum stress (ERS) and mitophagy through M2AChR to resist myocardial ischemia-reperfusion (I/R) injury and to elucidate its mechanism of action. Results: Exercise enhanced parasympathetic nerve function and increased myocardial M2AChR protein expression in I/R rats. In addition, it promoted the protein expression of MFN2 and inhibited the expression of Drp1, Chop, PINK1/Parkin, and PERK/eIF2α/ATF4 signaling pathways, effectively reducing mitophagy, ERS, and apoptosis. At the cellular level, 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) reduced hypoxia/reoxygenation (H/R)-induced ERS through the downregulated expression of PERK/eIF2α/ATF4 pathway proteins in H9C2 cardiomyocytes. When intervened with M2AChR inhibitors, the levels of ERS and phosphorylation levels of the PERK/eIF2α/ATF4 pathway were increased in H/R cells. Innovation and Conclusion: Exercise intervention activated the parasympathetic state in rats. It inhibited myocardial mitophagy and ERS levels, and reduced myocardial apoptosis through M2AChR, thereby resisting I/R-induced myocardial injury and improving cardiac function. Antioxid. Redox Signal. 40, 209-221.
Collapse
Affiliation(s)
- Wei Chen
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
- School of Educational Science, Shaanxi University of Technology, Hanzhong, China
| | - Mei Ma
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Yinping Song
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Yijie Hua
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hao Jia
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| | - Jiankang Liu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Youhua Wang
- Institute of Sports and Exercise Biology, School of Physical Education, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
24
|
Bononi G, Citi V, Martelli A, Poli G, Tuccinardi T, Granchi C, Testai L, Calderone V, Minutolo F. Sirtuin 1-activating derivatives belonging to the anilinopyridine class displaying in vivo cardioprotective activities. RSC Med Chem 2024; 15:267-282. [PMID: 38283227 PMCID: PMC10809423 DOI: 10.1039/d3md00489a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/27/2023] [Indexed: 01/30/2024] Open
Abstract
Sirtuin 1 (SIRT1) is an enzyme that relies on NAD+ cofactor and functions as a deacetylase. It has been associated with various biological and pathological processes, including cancer, diabetes, and cardiovascular diseases. Recent studies have shown that compounds that activate SIRT1 exhibit protective effects on the heart. Consequently, targeting SIRT1 has emerged as a viable approach to treat cardiovascular diseases, leading to the identification of several SIRT1 activators derived from natural or synthetic sources. In this study, we developed anilinopyridine-based SIRT1 activators that displayed significantly greater potency in activating SIRT1 compared to the reference compound resveratrol, as demonstrated in enzymatic assays. In particular, compounds 8 and 10, representative 6-aryl-2-anilinopyridine derivatives from this series, were further investigated pharmacologically and found to reduce myocardial damage caused by occlusion and subsequent reperfusion in vivo, confirming their cardioprotective properties. Notably, the cardioprotective effects of 8 and 10 were significantly superior to that of resveratrol. Significantly, compound 10 emerged as the most potent among the tested compounds, demonstrating the ability to substantially decrease the size of the ischemic area at a dosage one hundred times lower (0.1 mg kg-1) than that of resveratrol/compound 1. These promising findings open avenues for expanding and optimizing this chemical class of potent SIRT1 activators as potential agents for cardioprotection.
Collapse
Affiliation(s)
- Giulia Bononi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | - Valentina Citi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
- Center for Instrument Sharing of the University of Pisa (CISUP) Lungarno Pacinotti 43 56126 Pisa Italy
| | - Giulio Poli
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
- Center for Instrument Sharing of the University of Pisa (CISUP) Lungarno Pacinotti 43 56126 Pisa Italy
| | - Carlotta Granchi
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
- Center for Instrument Sharing of the University of Pisa (CISUP) Lungarno Pacinotti 43 56126 Pisa Italy
| | - Lara Testai
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
- Center for Instrument Sharing of the University of Pisa (CISUP) Lungarno Pacinotti 43 56126 Pisa Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
- Center for Instrument Sharing of the University of Pisa (CISUP) Lungarno Pacinotti 43 56126 Pisa Italy
| | - Filippo Minutolo
- Department of Pharmacy, University of Pisa Via Bonanno 6 56126 Pisa Italy
- Center for Instrument Sharing of the University of Pisa (CISUP) Lungarno Pacinotti 43 56126 Pisa Italy
| |
Collapse
|
25
|
Carmo HRP, Bonilha I, Barreto J, Tognolini M, Zanotti I, Sposito AC. High-Density Lipoproteins at the Interface between the NLRP3 Inflammasome and Myocardial Infarction. Int J Mol Sci 2024; 25:1290. [PMID: 38279290 PMCID: PMC10816227 DOI: 10.3390/ijms25021290] [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: 12/30/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Despite significant therapeutic advancements, morbidity and mortality following myocardial infarction (MI) remain unacceptably high. This clinical challenge is primarily attributed to two significant factors: delayed reperfusion and the myocardial injury resulting from coronary reperfusion. Following reperfusion, there is a rapid intracellular pH shift, disruption of ionic balance, heightened oxidative stress, increased activity of proteolytic enzymes, initiation of inflammatory responses, and activation of several cell death pathways, encompassing apoptosis, necroptosis, and pyroptosis. The inflammatory cell death or pyroptosis encompasses the activation of the intracellular multiprotein complex known as the NLRP3 inflammasome. High-density lipoproteins (HDL) are endogenous particles whose components can either promote or mitigate the activation of the NLRP3 inflammasome. In this comprehensive review, we explore the role of inflammasome activation in the context of MI and provide a detailed analysis of how HDL can modulate this process.
Collapse
Affiliation(s)
- Helison R. P. Carmo
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | - Isabella Bonilha
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | - Joaquim Barreto
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| | | | - Ilaria Zanotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy;
| | - Andrei C. Sposito
- Atherosclerosis and Vascular Biology Laboratory (Aterolab), Division of Cardiology, State University of Campinas (UNICAMP), Campinas 13084-971, SP, Brazil; (H.R.P.C.); (I.B.); (J.B.); (A.C.S.)
| |
Collapse
|
26
|
Zhang J, Si R, Gao Y, Shan H, Su Q, Feng Z, Huang P, Kong D, Wang W. dECM restores macrophage immune homeostasis and alleviates iron overload to promote DTPI healing. Regen Biomater 2024; 11:rbad118. [PMID: 38404617 PMCID: PMC10884736 DOI: 10.1093/rb/rbad118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 02/27/2024] Open
Abstract
Due to its highly insidious and rapid progression, deep tissue pressure injury (DTPI) is a clinical challenge. Our previous study found that DTPI may be a skeletal muscle injury dominated by macrophage immune dysfunction due to excessive iron accumulation. Decellularized extracellular matrix (dECM) hydrogel promotes skeletal muscle injury repair. However, its role in polarizing macrophages and regulating iron metabolism in DTPI remains unclear. Here, porcine dECM hydrogel was prepared, and its therapeutic function and mechanism in repairing DTPI were investigated. The stimulus of dECM hydrogel toward RAW264.7 cells resulted in a significantly higher percentage of CD206+ macrophages and notably decreased intracellular divalent iron levels. In mice DTPI model, dECM hydrogel treatment promoted M1 to M2 macrophage conversion, improved iron metabolism and reduced oxidative stress in the early stage of DTPI. In the remodeling phase, the dECM hydrogel remarkably enhanced revascularization and accelerated skeletal muscle repair. Furthermore, the immunomodulation of dECM hydrogels in vivo was mainly involved in the P13k/Akt signaling pathway, as revealed by GO and KEGG pathway analysis, which may ameliorate the iron deposition and promote the healing of DTPI. Our findings indicate that dECM hydrogel is promising in skeletal muscle repair, inflammation resolution and tissue injury healing by effectively restoring macrophage immune homeostasis and normalizing iron metabolism.
Collapse
Affiliation(s)
- Ju Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
- School of Nursing, Qingdao University, Ningde Road, Shinan District, Qingdao, Shandong, 266071, China
| | - Ruijuan Si
- Cancer Hospital of Tianjin Medical University, North Huanhu West Road, Tianjin, China
| | - Yu Gao
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Hui Shan
- The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Shinan District, Qingdao, China
| | - Qi Su
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Zujian Feng
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | | | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
27
|
Papakyriakopoulou P, Valsami G, Kadoglou NPE. Nose-to-Heart Approach: Unveiling an Alternative Route of Acute Treatment. Biomedicines 2024; 12:198. [PMID: 38255302 PMCID: PMC10813812 DOI: 10.3390/biomedicines12010198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
Intranasal (IN) administration has emerged as a novel approach for rapid systemic absorption, with potential applicability in the management of acute cardiovascular events. This review explores the evolution of IN cardiovascular pharmacotherapy, emphasizing its potential in achieving systemic effects and bypassing the first-pass metabolism associated with oral administration. The extensive vascularization of nasal mucosa and a porous endothelial basement membrane facilitate efficient drug absorption into the bloodstream. The IN route ensures a critical swift onset of action, which allows self-administration in at-home settings. For instance, etripamil nasal spray, a first-in-class formulation, exemplifies the therapeutic potential of this approach in the treatment of spontaneous supraventricular tachycardia. The review critically assesses studies on IN formulations for angina, acute myocardial infarction, hypertensive episodes, and cardiac arrhythmias. Preclinical evaluations of beta-blockers, calcium-channel blockers, and antianginal drugs demonstrate the feasibility of IN administration for acute cardiovascular events. A small number of clinical trials have revealed promising results, emphasizing the superiority of IN drug delivery over oral administration in terms of bioavailability and onset of action. Unambiguously, the limited clinical trials and patient enrollment pose challenges in generalizing experimental outcomes. However, the nose-to-heart approach has clinical potential.
Collapse
Affiliation(s)
- Paraskevi Papakyriakopoulou
- Laboratory of Biopharmaceutics and Pharmacokinetics, Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens, Greece; (P.P.); (G.V.)
| | - Georgia Valsami
- Laboratory of Biopharmaceutics and Pharmacokinetics, Section of Pharmaceutical Technology, Department of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Athens, Greece; (P.P.); (G.V.)
| | | |
Collapse
|
28
|
Tan T, Tu L, Yu Y, He M, Zhou X, Yang L. Mechanisms by which silencing long-stranded noncoding RNA KCNQ1OT1 alleviates myocardial ischemia/reperfusion injury (MI/RI)-induced cardiac injury via miR-377-3p/HMOX1. BMC Cardiovasc Disord 2024; 24:19. [PMID: 38172743 PMCID: PMC10765944 DOI: 10.1186/s12872-023-03693-y] [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: 09/26/2023] [Accepted: 12/25/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The key complication of myocardial infarction therapy is myocardial ischemia/reperfusion injury (MI/RI), and there is no effective treatment. The present study elucidates the mechanism of action of lncRNA KCNQ1OT1 in alleviating MI/RI and provides new perspectives and therapeutic targets for cardiac injury-related diseases. METHODS An ischemia/reperfusion (I/R) injury model of human adult cardiac myocytes (HACMs) was constructed, and the expression of KCNQ1OT1 and miR-377-3p was determined by RT‒qPCR. The levels of related proteins were detected by western blot analysis. Cell proliferation was detected by a CCK-8 assay, and cell apoptosis and ROS content were determined by flow cytometry. SOD and MDA expression as well as Fe2+ changes were detected by related analysis kits. The target binding relationships between lncRNA KCNQ1OT1 and miR-377-3p as well as between miR-377-3p and heme oxygenase 1 (HMOX1) were verified by a dual-luciferase reporter gene assay. RESULTS Myocardial ischemia‒reperfusion caused oxidative stress in HACMs, resulting in elevated ROS levels, increased Fe2+ levels, decreased cell viability, and increased LDH release (a marker of myocardial injury), and apoptosis. KCNQ1OT1 and HMOX1 were upregulated in I/R-induced myocardial injury, but the level of miR-377-3p was decreased. A dual-luciferase reporter gene assay indicated that lncRNA KCNQ1OT1 targets miR-377-3p and that miR-377-3p targets HMOX1. Inhibition of HMOX1 alleviated miR-377-3p downregulation-induced myocardial injury. Furthermore, lncRNA KCNQ1OT1 promoted the level of HMOX1 by binding to miR-377-3p and aggravated myocardial injury. CONCLUSION LncRNA KCNQ1OT1 aggravates ischemia‒reperfusion-induced cardiac injury via miR-377-3P/HMOX1.
Collapse
Affiliation(s)
- Tongcai Tan
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Liang Tu
- Medical Experimental Center, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College (The 6th People Hospital of Chongqing), Chongqing, 400060, China
| | - Yanmei Yu
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - MinJie He
- Geriatric Department, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650000, China
| | - Xingchao Zhou
- Department of Medical Equipment, The First Affiliated Hospital of Dali University, Dali, Yunnan, 671000, China
| | - Lei Yang
- Department of Rehabilitation Medicine, The Second People's Hospital of Kunming, Kunming, Yunnan, 650506, China.
| |
Collapse
|
29
|
Zhang Q, Gou F, Shi P, Xu Z, Yan Z, He M, Yin X, He Y, Zhang J. Angiotensin-converting enzyme inhibitors provide a protective effect on hypoxia-induced injury in human coronary artery endothelial cells via Nrf2 signaling and PLVAP. Clin Hemorheol Microcirc 2024; 87:141-170. [PMID: 38339922 DOI: 10.3233/ch-232007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
BACKGROUND Angiotensin-converting enzyme inhibitors (ACEIs) were reported to protect from hypoxia-induced oxidative stress in coronary endothelial cells (CECs) after acute myocardial infarction (AMI). Nrf2 shows a protective effect in hypoxia-induced CECs after AMI. Plasmalemma vesicle-associated protein (PLVAP) plays a pivotal role in angiogenesis after AMI. AIM To explore the protective effect of ACEIs and the involved mechanisms under hypoxia challenge. METHODS Human coronary endothelial cells (HCAECs) were used to establish hypoxia-induced oxidative stress injury in vitro. Flow cytometry was used to evaluate the protective effect of ACEI on hypoxia conditions.ET-1, NO, ROS, and VEGF were detected by ELISA. HO-1, Nrf2, and Keap-1, the pivotal member in the Nrf2 signaling pathway, eNOS and PLVAP were detected in HEAECs treated with ACEI by immunofluorescence, qPCR, and western blotting. RESULTS The hypoxia ACEI or Nrf2 agonist groups showed higher cell viability compared with the hypoxia control group at 24 (61.75±1.16 or 61.23±0.59 vs. 44.24±0.58, both P < 0.05) and 48 h (41.85±1.19 or 59.64±1.13 vs. 22.98±0.25, both P < 0.05). ACEI decreased the levels of ET-1 and ROS under hypoxia challenge at 24 and 48 h (all P < 0.05); ACEI increased the VEGF and NO levels (all P < 0.05). ACEI promoted the expression level of eNOS, HO-1, Nrf2 and PLVAP but inhibited Keap-1 expression at the mRNA and protein levels (all P < 0.05). Blockade of the Nrf2 signaling pathway significantly decreased the expression level of PLVAP. CONCLUSION ACEI protects hypoxia-treated HEAECs by activating the Nrf2 signaling pathway and upregulating the expression of PLVAP.
Collapse
Affiliation(s)
- Qiubing Zhang
- Tianjin Medical University, Tianjin, China
- Department of Cardiology, Guang Yuan Central Hospital, Guang Yuan, China
| | - Fang Gou
- Department of Cardiology, Guang Yuan Central Hospital, Guang Yuan, China
| | - Ping Shi
- Department of Cardiology, Guang Yuan Central Hospital, Guang Yuan, China
| | - Zhe Xu
- Department of Cardiology, Guang Yuan Central Hospital, Guang Yuan, China
| | - Zhitao Yan
- Department of Cardiology, Guang Yuan Central Hospital, Guang Yuan, China
| | - Mingfang He
- Department of Cardiology, Guang Yuan Central Hospital, Guang Yuan, China
| | - Xiaohong Yin
- Department of Cardiology, Guang Yuan Central Hospital, Guang Yuan, China
| | - Yuanjun He
- Department of Cardiology, Guang Yuan Central Hospital, Guang Yuan, China
| | - Jun Zhang
- Department of Cardiology, Cangzhou Central Hospital, Tianjin Medical University, Cangzhou, China
| |
Collapse
|
30
|
Gholami S, Badalzadeh R, Alihemmati A. Alpha-lipoic acid enhances ischemic postconditioning-mediated improvement of myocardial infarction and apoptosis in diabetic rats with ischemia/reperfusion injury. Can J Physiol Pharmacol 2023; 101:682-691. [PMID: 37523770 DOI: 10.1139/cjpp-2023-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
This work evaluated the combined effects of alpha-lipoic acid (ALA) and ischemic postconditioning (Post) on myocardial infarction and cell death in rats with chronic type-II diabetes following ischemia/reperfusion injury. The rats received a high-fat diet and were given one intraperitoneal injection of 35 mg/kg streptozotocin to induce chronic diabetes. They were then pretreated with ALA (100 mg/kg/day, orally) for 5 weeks before undergoing ischemia/reperfusion (I/R) insult. The hearts experienced 35 min regional ischemia through ligating the left anterior descending coronary artery, followed by 60 min reperfusion. The Post protocol involved 6 cycles of a 10/10 s algorithm, applied during the early stage of reperfusion. The use of Post alone did not significantly alter lactate dehydrogenase and infarct size levels, while ALA showed positive effects. Similar findings were observed for apoptotic changes with single treatments. However, the concurrent administration of ALA and Post significantly reduced the protein expressions of Bax, Bax/Bcl2, and cleaved caspase-3 while increasing Bcl2 expression. Additionally, the histopathological findings of the combined therapy were superior to those of single treatments. The concomitant use of ALA and Post effectively inhibited apoptosis, leading to cardiac recovery after I/R injury in diabetic conditions. This strategy could improve outcomes for preserving diabetic hearts following I/R insults.
Collapse
Affiliation(s)
- Sanaz Gholami
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Reza Badalzadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
- Department of PhysiologyFaculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| | - Alireza Alihemmati
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
- Department of Anatomical SciencesFaculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Islamic Republic of Iran
| |
Collapse
|
31
|
Haybar H, Sadati NS, Purrahman D, Mahmoudian-Sani MR, Saki N. lncRNA TUG1 as potential novel biomarker for prognosis of cardiovascular diseases. Epigenomics 2023; 15:1273-1290. [PMID: 38088089 DOI: 10.2217/epi-2023-0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024] Open
Abstract
Globally, cardiovascular diseases (CVDs) are among the leading causes of death. In light of the high prevalence and mortality of CVDs, it is imperative to understand the molecules involved in CVD pathogenesis and the signaling pathways that they initiate. This may facilitate the development of more precise and expedient diagnostic techniques, the identification of more effective prognostic molecules and the identification of potential therapeutic targets. Numerous studies have examined the role of lncRNAs, such as TUG1, in CVD pathogenesis in recent years. According to this review article, TUG1 can be considered a biomarker for predicting the prognosis of CVD.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Narjes Sadat Sadati
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Daryush Purrahman
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Reza Mahmoudian-Sani
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
32
|
Dong W, Peng Q, Liu Z, Xie Z, Guo X, Li Y, Chen C. Estrogen plays an important role by influencing the NLRP3 inflammasome. Biomed Pharmacother 2023; 167:115554. [PMID: 37738797 DOI: 10.1016/j.biopha.2023.115554] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023] Open
Abstract
The nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome is an important part of the natural immune system that plays an important role in many diseases. Estrogen is a sex hormone that plays an important role in controlling reproduction and regulates many physiological and pathological processes. Recent studies have indicated that estrogen is associated with disease progression. Estrogen can ameliorate some diseases (e. g, sepsis, mood disturbances, cerebral ischemia, some hepatopathy, Parkinson's disease, amyotrophic lateral sclerosis, inflammatory bowel disease, spinal cord injury, multiple sclerosis, myocardial ischemia/reperfusion injury, osteoarthritis, and renal fibrosis) by inhibiting the NLRP3 inflammasome. Estrogen can also promote the development of diseases (e.g., ovarian endometriosis, dry eye disease, and systemic lupus erythematosus) by upregulating the NLRP3 inflammasome. In addition, estrogen has a dual effect on the development of cancers and asthma. However, the mechanism of these effects is not summarized. This article reviewed the progress in understanding the effects of estrogen on the NLRP3 inflammasome and its mechanisms in recent years to provide a theoretical basis for an in-depth study.
Collapse
Affiliation(s)
- Wanglin Dong
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Qianwen Peng
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Zhuoxin Liu
- Clinical College of Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhenxing Xie
- School of Basic Medical Science, Henan University, Jinming Avenue, Kaifeng, Henan 475004, China.
| | - Xiajun Guo
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Yuanyuan Li
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Chaoran Chen
- Institute of Nursing and Health, College of Nursing and Health, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
33
|
Eleftheriadis T, Pissas G, Golfinopoulos S, Liakopoulos V, Stefanidis I. Indoleamine 2,3-dioxygenase controls purinergic receptor-mediated ischemia-reperfusion injury in renal tubular epithelial cells. J Basic Clin Physiol Pharmacol 2023; 34:745-754. [PMID: 35918786 DOI: 10.1515/jbcpp-2022-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/03/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVES Ischemia-reperfusion (I-R) injury is the most common cause of acute kidney injury (AKI). Experimental studies have shown that indoleamine 2,3-dioxygenase 1 (IDO) and the purinergic receptor P2X7 contribute to kidney I-R injury. We evaluated whether there is an interplay between IDO and P2X7 in the pathogenesis of I-R injury. METHODS Primary renal proximal tubular epithelial cells (RPTECs) were subjected to anoxia or reoxygenation with or without specific inhibitors. Cell imaging, colorimetric assays, and Western blotting were used. RESULTS Cell imaging revealed that inhibition of IDO, or all the purinergic receptors with an ATPase, or specific inhibition of P2X7 rescued the cells from anoxia or reoxygenation-induced cell death. This was confirmed with LDH release assay, which also detected the ferroptotic nature of cell death due to reoxygenation. On the contrary, activated cleaved caspase 3 increased during anoxia, showing that apoptosis prevails. All the aforementioned treatments prevented caspase increase. Both anoxia and reoxygenation increased extracellular ATP, IDO, and P2X7 expression. IDO remained unaffected by the above-mentioned treatments. On the contrary, treatment with apyrase or inhibition of P2X7decreased extracellular ATP and P2X7 expression, which are also decreased by inhibition of IDO. The first indicates a positive feedback loop regarding P2X7 activation, expression and function, while the latter implies that IDO controls P2X7 expression. CONCLUSIONS In RPRECs subjected to anoxia or reoxygenation, IDO is upregulated, increasing P2X7 and contributing to anoxia or reoxygenation-induced cell death. Clarifying the molecular mechanisms implicated in kidney I-R injury is of particular interest since it may lead to new therapeutic strategies against AKI.
Collapse
Affiliation(s)
| | - Georgios Pissas
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | - Vassilios Liakopoulos
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| |
Collapse
|
34
|
Gouchoe DA, Vijayakumar A, Aly AH, Cui EY, Essandoh M, Gumina RJ, Black SM, Whitson BA. The role of CD38 in ischemia reperfusion injury in cardiopulmonary bypass and thoracic transplantation: a narrative review. J Thorac Dis 2023; 15:5736-5749. [PMID: 37969313 PMCID: PMC10636473 DOI: 10.21037/jtd-23-725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 08/25/2023] [Indexed: 11/17/2023]
Abstract
Background and Objective Ischemia reperfusion injury (IRI) is often the underlying cause of endothelium breakdown and damage in cardiac or transplantation operations, which can lead to disastrous post-operative consequences. Recent studies of cluster of differentiation 38 (CD38) have identified its critical role in IRI. Our objective is to provide a comprehensive overview of CD38-mediated axis, pathways, and potential CD38 translational therapies for reducing inflammation associated with cardiopulmonary bypass (CPB) or thoracic transplantation and IRI. Methods We conducted a review of the literature by performing a search of the PubMed database on 2 April 2023. To find relevant publications on CD38, we utilized the MeSH terms: "CD38" AND "Ischemia" OR "CD38" AND "Transplant" OR "CD38" AND "Heart" from 1990-2023. Additional papers were included if they were felt to be relevant but were not captured in the MeSH terms. We found 160 papers that met this criterion, and following screening, exclusion and consensus a total of 36 papers were included. Key Content and Findings CD38 is most notably a nicotine adenine dinucleotide (NAD)+ glycohydrolase (NADase), and a generator of Ca2+ signaling secondary messengers. Ultimately, the release of these secondary messengers leads to the activation of important mediators of cellular death. In the heart and during thoracic transplantation, this pathway is intimately involved in a wide variety of injuries; namely the endothelium. In the heart, activation generally results in vasoconstriction, poor myocardial perfusion, and ultimately poor cardiac function. CD38 activation also prevents the accumulation of atherosclerotic disease. During transplantation, intracellular activation leads to infiltration of recipient innate immune cells, tissue edema, and ultimately primary graft dysfunction (PGD). Specifically, in heart transplantation, extracellular activation could be protective and improve allograft survival. Conclusions The knowledge gap in understanding the molecular basis of IRI has prevented further development of novel therapies and treatments. The possible interaction of CD38 with CD39 in the endothelium, and the modulation of the CD38 axis may be a pathway to improve cardiovascular outcomes, heart and lung donor organ quality, and overall longevity.
Collapse
Affiliation(s)
- Doug A. Gouchoe
- COPPER Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- 88 Surgical Operations Squadron, Wright-Patterson Medical Center, Wright Patterson AFB, OH, USA
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ammu Vijayakumar
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ahmed H. Aly
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ervin Y. Cui
- COPPER Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Michael Essandoh
- Department of Anesthesiology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Richard J. Gumina
- Division of Cardiovascular Medicine, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Davis Heart and Lung Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Sylvester M. Black
- COPPER Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Division of Transplantation, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Bryan A. Whitson
- COPPER Laboratory, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Division of Cardiac Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Davis Heart and Lung Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
35
|
Mohindra P, Zhong JX, Fang Q, Cuylear DL, Huynh C, Qiu H, Gao D, Kharbikar BN, Huang X, Springer ML, Lee RJ, Desai TA. Local decorin delivery via hyaluronic acid microrods improves cardiac performance, ventricular remodeling after myocardial infarction. NPJ Regen Med 2023; 8:60. [PMID: 37872196 PMCID: PMC10593781 DOI: 10.1038/s41536-023-00336-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 10/09/2023] [Indexed: 10/25/2023] Open
Abstract
Heart failure (HF) remains a global public health burden and often results following myocardial infarction (MI). Following injury, cardiac fibrosis forms in the myocardium which greatly hinders cellular function, survival, and recruitment, thus severely limits tissue regeneration. Here, we leverage biophysical microstructural cues made of hyaluronic acid (HA) loaded with the anti-fibrotic proteoglycan decorin to more robustly attenuate cardiac fibrosis after acute myocardial injury. Microrods showed decorin incorporation throughout the entirety of the hydrogel structures and exhibited first-order release kinetics in vitro. Intramyocardial injections of saline (n = 5), microrods (n = 7), decorin microrods (n = 10), and free decorin (n = 4) were performed in male rat models of ischemia-reperfusion MI to evaluate therapeutic effects on cardiac remodeling and function. Echocardiographic analysis demonstrated that rats treated with decorin microrods (5.21% ± 4.29%) exhibited significantly increased change in ejection fraction (EF) at 8 weeks post-MI compared to rats treated with saline (-4.18% ± 2.78%, p < 0.001) and free decorin (-3.42% ± 1.86%, p < 0.01). Trends in reduced end diastolic volume were also identified in decorin microrod-treated groups compared to those treated with saline, microrods, and free decorin, indicating favorable ventricular remodeling. Quantitative analysis of histology and immunofluorescence staining showed that treatment with decorin microrods reduced cardiac fibrosis (p < 0.05) and cardiomyocyte hypertrophy (p < 0.05) at 8 weeks post-MI compared to saline control. Together, this work aims to contribute important knowledge to guide rationally designed biomaterial development that may be used to successfully treat cardiovascular diseases.
Collapse
Affiliation(s)
- Priya Mohindra
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Justin X Zhong
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Qizhi Fang
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Darnell L Cuylear
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Graduate Program in Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - Cindy Huynh
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Division of Vascular and Endovascular Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Huiliang Qiu
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Dongwei Gao
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Bhushan N Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Xiao Huang
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew L Springer
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
| | - Randall J Lee
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
- Division of Cardiology, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - Tejal A Desai
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA.
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA.
- School of Engineering, Brown University, Providence, RI, USA.
| |
Collapse
|
36
|
Bernini Di Michele A, Onofri V, Pesaresi M, Turchi C. The Role of miRNA Expression Profile in Sudden Cardiac Death Cases. Genes (Basel) 2023; 14:1954. [PMID: 37895303 PMCID: PMC10606010 DOI: 10.3390/genes14101954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Sudden cardiac death (SCD) is one of the leading causes of death in the world and for this reason it has attracted the attention of numerous researchers in the field of legal medicine. It is not easy to determine the cause in a SCD case and the available methods used for diagnosis cannot always give an exhaustive answer. In addition, the molecular analysis of genes does not lead to a clear conclusion, but it could be interesting to focus attention on the expression level of miRNAs, a class of non-coding RNA of about 22 nucleotides. The role of miRNAs is to regulate the gene expression through complementary binding to 3'-untraslated regions of miRNAs, leading to the inhibition of translation or to mRNA degradation. In recent years, several studies were performed with the aim of exploring the use of these molecules as biomarkers for SCD cases, and to also distinguish the causes that lead to cardiac death. In this review, we summarize experiments, evidence, and results of different studies on the implication of miRNAs in SCD cases. We discuss the different biological starting materials with their respective advantages and disadvantages, studying miRNA expression on tissue (fresh-frozen tissue and FFPE tissue), circulating cell-free miRNAs in blood of patients affected by cardiac disease at high risk of SCD, and exosomal miRNAs analyzed from serum of people who died from SCD.
Collapse
Affiliation(s)
- Alessia Bernini Di Michele
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| | - Valerio Onofri
- Legal Medicine Unit, AOU Azienda Ospedaliero Universitaria delle Marche, 60126 Ancona, Italy;
| | - Mauro Pesaresi
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| | - Chiara Turchi
- Section of Legal Medicine, Department of Biomedical Sciences and Public Health, Polytechnic University of Marche, Via Tronto, 60126 Ancona, Italy; (A.B.D.M.); (M.P.)
| |
Collapse
|
37
|
Wang L, Wang C, Sun Z, Du A, Shan F, Sun Z. Knockdown of Mmu-circ-0001380 Attenuates Myocardial Ischemia/Reperfusion Injury via Modulating miR-106b-5p/Phlpp2 Axis. J Cardiovasc Transl Res 2023; 16:1064-1077. [PMID: 37474690 DOI: 10.1007/s12265-023-10383-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 03/22/2023] [Indexed: 07/22/2023]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury induces myocardial damage and dysfunction. Increasing evidence has confirmed that circular RNAs (circRNAs) play crucial roles in regulating MI/R. Mmu-circ-0001380 has identified to be highly expressed in myocardium of MI/R mouse model. However, its biological function and molecular mechanism in MI/R injury are still unclear. Here, we demonstrated that knockdown of cric-0001380 attenuated myocardial injury of MI/R mice. In vitro, silence of circ-0001380 significantly enhanced viability, and inhibited apoptosis and oxidative stress in HL-1 cells under oxygen-glucose deprivation/reoxygenation (OGD/R). Mmu-miR-106b-5p interacted with circ-0001380, and suppressed the expression of pleckstrin homology domain and leucine rich repeat protein phosphatase 2 (Phlpp2). The miR-106b-5p/Phlpp2 axis mediated the effect of circ-0001380 on OGD/R-induced apoptosis through regulating the phosphorylation of p38, and further involved in regulating the viability and oxidative stress of HL-1 cells. In conclusion, circ-0001380 downregulation relieves MI/R injury via regulating the miR-106b-5p/Phlpp2 axis. The present study indicates that mmu-circ-0001380 exacerbates the myocardial ischemia/reperfusion injury through modulating the miR-106b-5p/Phlpp2 axis in vitro and in vivo.
Collapse
Affiliation(s)
- Li Wang
- Department of Cardiology, Dalian Municipal Central Hospital, No. 826, Xinan Road, Dalian, Liaoning, China.
| | - Chuanhe Wang
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 39, Huaxiang Road, Shenyang, Liaoning, China
| | - Zhaoqing Sun
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 39, Huaxiang Road, Shenyang, Liaoning, China
| | - Aolin Du
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 39, Huaxiang Road, Shenyang, Liaoning, China
| | - Fei Shan
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 39, Huaxiang Road, Shenyang, Liaoning, China
| | - Zhijun Sun
- Department of Cardiology, Shengjing Hospital of China Medical University, No. 39, Huaxiang Road, Shenyang, Liaoning, China.
| |
Collapse
|
38
|
Skaria RS, Lopez‐Pier MA, Kathuria BS, Leber CJ, Langlais PR, Aras SG, Khalpey ZI, Hitscherich PG, Chnari E, Long M, Churko JM, Runyan RB, Konhilas JP. Epicardial placement of human placental membrane protects from heart injury in a swine model of myocardial infarction. Physiol Rep 2023; 11:e15838. [PMID: 37849042 PMCID: PMC10582231 DOI: 10.14814/phy2.15838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 10/19/2023] Open
Abstract
Cardiac ischemic reperfusion injury (IRI) is paradoxically instigated by reestablishing blood-flow to ischemic myocardium typically from a myocardial infarction (MI). Although revascularization following MI remains the standard of care, effective strategies remain limited to prevent or attenuate IRI. We hypothesized that epicardial placement of human placental amnion/chorion (HPAC) grafts will protect against IRI. Using a clinically relevant model of IRI, swine were subjected to 45 min percutaneous ischemia followed with (MI + HPAC, n = 3) or without (MI only, n = 3) HPAC. Cardiac function was assessed by echocardiography, and regional punch biopsies were collected 14 days post-operatively. A deep phenotyping approach was implemented by using histological interrogation and incorporating global proteomics and transcriptomics in nonischemic, ischemic, and border zone biopsies. Our results established HPAC limited the extent of cardiac injury by 50% (11.0 ± 2.0% vs. 22.0 ± 3.0%, p = 0.039) and preserved ejection fraction in HPAC-treated swine (46.8 ± 2.7% vs. 35.8 ± 4.5%, p = 0.014). We present comprehensive transcriptome and proteome profiles of infarct (IZ), border (BZ), and remote (RZ) zone punch biopsies from swine myocardium during the proliferative cardiac repair phase 14 days post-MI. Both HPAC-treated and untreated tissues showed regional dynamic responses, whereas only HPAC-treated IZ revealed active immune and extracellular matrix remodeling. Decreased endoplasmic reticulum (ER)-dependent protein secretion and increased antiapoptotic and anti-inflammatory responses were measured in HPAC-treated biopsies. We provide quantitative evidence HPAC reduced cardiac injury from MI in a preclinical swine model, establishing a potential new therapeutic strategy for IRI. Minimizing the impact of MI remains a central clinical challenge. We present a new strategy to attenuate post-MI cardiac injury using HPAC in a swine model of IRI. Placement of HPAC membrane on the heart following MI minimizes ischemic damage, preserves cardiac function, and promotes anti-inflammatory signaling pathways.
Collapse
Affiliation(s)
- Rinku S. Skaria
- Department of PhysiologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Marissa A. Lopez‐Pier
- Department of Biomedical EngineeringUniversity of Arizona College of EngineeringTucsonArizonaUSA
| | - Brij S. Kathuria
- Department of PhysiologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Christian J. Leber
- Department of PhysiologyUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Paul R. Langlais
- Department of MedicineUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Shravan G. Aras
- Center for Biomedical and InformaticsUniversity of Arizona Health SciencesTucsonArizonaUSA
| | | | | | | | | | - Jared M. Churko
- Department of Cellular and Molecular MedicineUniversity of Arizona College of MedicineTucsonArizonaUSA
- Sarver Molecular Cardiovascular Research ProgramUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - Raymond B. Runyan
- Department of Cellular and Molecular MedicineUniversity of Arizona College of MedicineTucsonArizonaUSA
- Sarver Molecular Cardiovascular Research ProgramUniversity of Arizona College of MedicineTucsonArizonaUSA
| | - John P. Konhilas
- Department of PhysiologyUniversity of Arizona College of MedicineTucsonArizonaUSA
- Department of Biomedical EngineeringUniversity of Arizona College of EngineeringTucsonArizonaUSA
- Sarver Molecular Cardiovascular Research ProgramUniversity of Arizona College of MedicineTucsonArizonaUSA
| |
Collapse
|
39
|
Wang J, Tan Y, Dai Y, Hu K, Tan X, Jiang S, Li G, Zhang X, Kang L, Wang X, Xu B. Intranasal Delivery of Endothelial Cell-Derived Extracellular Vesicles with Supramolecular Gel Attenuates Myocardial Ischemia-Reperfusion Injury. Int J Nanomedicine 2023; 18:5495-5510. [PMID: 37791323 PMCID: PMC10544033 DOI: 10.2147/ijn.s420301] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/12/2023] [Indexed: 10/05/2023] Open
Abstract
Purpose Myocardial ischemia-reperfusion injury after myocardial infarction has always been a difficult problem in clinical practice. Endothelial cells and their secreted extracellular vesicles are closely related to inflammation, thrombosis formation, and other processes after injury. Meanwhile, low-molecular-weight gelators have shown great potential for nasal administration. This study aims to explore the therapeutic effects and significance of endothelial cell-derived extracellular vesicles combined with a hydrogel for nasal administration on myocardial ischemia-reperfusion injury. Methods We chose a gel system composed of a derivative of glutamine amide and benzaldehyde as the extracellular vesicle delivery vehicle. This hydrogel was combined with extracellular vesicles extracted from mouse aortic endothelial cells and administered multiple times intranasally in a mouse model of ischemia-reperfusion injury to the heart. The delivery efficiency of the extracellular vesicle-hydrogel combination was evaluated by flow cytometry and immunofluorescence. Echocardiography, TTC Evan's Blue and Masson's staining were used to assess mouse cardiac function, infarct area, and cardiac fibrosis level. Flow cytometry, ELISA, and immunofluorescence staining were used to investigate changes in mouse inflammatory cells, cytokines, and vascular neogenesis. Results The vesicles combined with the hydrogel have good absorption in the nasal cavity. The hydrogel combined with vesicles reduces the levels of pro-inflammatory Ly6C (high) monocytes/macrophages and neutrophils. It can also reduce the formation of microcirculation thrombi in the infarcted area, improve endothelial barrier function, and increase microvascular density in the injured area. As a result, the heart function of mice is improved and the infarct area is reduced. Conclusion We first demonstrated that the combination of extracellular vesicles and hydrogel has a better absorption efficiency in the nasal cavity, which can improve myocardial ischemia-reperfusion injury by inhibiting inflammatory reactions and protecting endothelial function. Nasal administration of vesicles combined with hydrogel is a potential therapeutic direction.
Collapse
Affiliation(s)
- Junzhuo Wang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Ying Tan
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Yang Dai
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Geriatrics, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Ke Hu
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Xi Tan
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Shaoli Jiang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, People’s Republic of China
| | - Guannan Li
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Xinlin Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Lina Kang
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
| | - Xiaojian Wang
- Institute of Advanced Synthesis, School of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, People’s Republic of China
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, People’s Republic of China
- Department of Cardiology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
40
|
Mak MCE, Gurung R, Foo RSY. Applications of Genome Editing Technologies in CAD Research and Therapy with a Focus on Atherosclerosis. Int J Mol Sci 2023; 24:14057. [PMID: 37762360 PMCID: PMC10531628 DOI: 10.3390/ijms241814057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
Cardiovascular diseases, particularly coronary artery disease (CAD), remain the leading cause of death worldwide in recent years, with myocardial infarction (MI) being the most common form of CAD. Atherosclerosis has been highlighted as one of the drivers of CAD, and much research has been carried out to understand and treat this disease. However, there remains much to be better understood and developed in treating this disease. Genome editing technologies have been widely used to establish models of disease as well as to treat various genetic disorders at their root. In this review, we aim to highlight the various ways genome editing technologies can be applied to establish models of atherosclerosis, as well as their therapeutic roles in both atherosclerosis and the clinical implications of CAD.
Collapse
Affiliation(s)
| | - Rijan Gurung
- Cardiovascular Research Institute, Cardiovascular and Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, MD6, #08-01, Singapore 117599, Singapore; (M.C.E.M.); (R.S.Y.F.)
| | | |
Collapse
|
41
|
Miao Y, Wang X, Yin H, Han R. Effects of Cavitation from Extracorporeal Shock Wave Combined with Sulfur Hexafluoride Microbubble on Myocardial Ultrastructure in Rats. Anatol J Cardiol 2023; 27:519-528. [PMID: 37288863 PMCID: PMC10510423 DOI: 10.14744/anatoljcardiol.2023.2946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/19/2023] [Indexed: 06/09/2023] Open
Abstract
BACKGROUND In the present study, the effects of extracorporeal cardiac shock waves combined with different concentrations of sulfur hexafluoride ultrasound microbubbles on myocardial ultrastructure in rats were observed. METHODS Thirty-six rats were randomly divided into 6 groups: control group (N), extracorporeal cardiac shock wave group, and combined group, i.e., extracorporeal cardiac shock wave combined with different concentrations of sulfur hexafluoride microbubble (0.225 mL/kg/min, 0.45 mL/kg/min, 0.9 mL/kg/min, 1.8 mL/kg/min). The combination of extracorporeal cardiac shock wave combined with sulfur hexafluoride microbubbles of different concentrations had no significant effect on hemodynamic indexes and left ventricular function in rats. RESULTS There were significant differences in cardiac troponin I (cTnI) and nitricoxide among different groups. Histopathology showed that inflammatory cells infiltrated in the shock wave+microbubble 0.9 and shock wave+microbubble 1.8 groups. The myocardial ultrastructural injury score of shock wave+microbubble1.8 group was significantly higher than that of the N group, shock wave group, shock wave+microbubble 0.225 group, and shock wave+microbubble 0.45 group. The score of shock wave+microbubble 0.9 group was higher than that of the control group (P=.009). Western blot results showed that the expression of vascular endothelial growth factor and endothelial nitricoxide synthase (eNOS) protein in the rats treated with extracorporeal cardiac shock wave combined with sulfur hexafluoride microbubbles of different concentrations was higher than that in the N group and shock wave group, with shock wave+microbubble 0.45 group having the strongest expression. CONCLUSION Myocardial ultrastructure damage occurs when high concentrations of sulfur hexafluoride microbubbles are present, but a proper concentration of sulfur hexafluoride microbubbles could promote the cavitation effect of extracorporeal cardiac shock waves. Thus combination therapy may become a new paradigm in coronary heart disease, especially contributing to the treatment of refractory angina. Combination therapy may change coronary heart disease treatment, especially for refractory angina.
Collapse
Affiliation(s)
- Yajing Miao
- Department of Echocardiography, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaoxu Wang
- Department of Echocardiography, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongning Yin
- Department of Echocardiography, the Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ruoling Han
- Department of Ultrasound, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
42
|
Bhuia MS, Wilairatana P, Ferdous J, Chowdhury R, Bappi MH, Rahman MA, Mubarak MS, Islam MT. Hirsutine, an Emerging Natural Product with Promising Therapeutic Benefits: A Systematic Review. Molecules 2023; 28:6141. [PMID: 37630393 PMCID: PMC10458569 DOI: 10.3390/molecules28166141] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
Fruits and vegetables are used not only for nutritional purposes but also as therapeutics to treat various diseases and ailments. These food items are prominent sources of phytochemicals that exhibit chemopreventive and therapeutic effects against several diseases. Hirsutine (HSN) is a naturally occurring indole alkaloid found in various Uncaria species and has a multitude of therapeutic benefits. It is found in foodstuffs such as fish, seafood, meat, poultry, dairy, and some grain products among other things. In addition, it is present in fruits and vegetables including corn, cauliflower, mushrooms, potatoes, bamboo shoots, bananas, cantaloupe, and citrus fruits. The primary emphasis of this study is to summarize the pharmacological activities and the underlying mechanisms of HSN against different diseases, as well as the biopharmaceutical features. For this, data were collected (up to date as of 1 July 2023) from various reliable and authentic literature by searching different academic search engines, including PubMed, Springer Link, Scopus, Wiley Online, Web of Science, ScienceDirect, and Google Scholar. Findings indicated that HSN exerts several effects in various preclinical and pharmacological experimental systems. It exhibits anti-inflammatory, antiviral, anti-diabetic, and antioxidant activities with beneficial effects in neurological and cardiovascular diseases. Our findings also indicate that HSN exerts promising anticancer potentials via several molecular mechanisms, including apoptotic cell death, induction of oxidative stress, cytotoxic effect, anti-proliferative effect, genotoxic effect, and inhibition of cancer cell migration and invasion against various cancers such as lung, breast, and antitumor effects in human T-cell leukemia. Taken all together, findings from this study show that HSN can be a promising therapeutic agent to treat various diseases including cancer.
Collapse
Affiliation(s)
- Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (R.C.); (M.H.B.)
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Jannatul Ferdous
- Department of Biotechnology and Genetic Engineering, Bangabandhu Sheikh Mujibur Rahman, Science and Technology University, Gopalganj 8100, Bangladesh;
| | - Raihan Chowdhury
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (R.C.); (M.H.B.)
| | - Mehedi Hasan Bappi
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (R.C.); (M.H.B.)
| | - Md Anisur Rahman
- Department of Pharmacy, Islamic University, Kushtia 7003, Bangladesh;
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (R.C.); (M.H.B.)
| |
Collapse
|
43
|
Liu Y, Wang M, Yu Y, Li C, Zhang C. Advances in the study of exosomes derived from mesenchymal stem cells and cardiac cells for the treatment of myocardial infarction. Cell Commun Signal 2023; 21:202. [PMID: 37580705 PMCID: PMC10424417 DOI: 10.1186/s12964-023-01227-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/12/2023] [Indexed: 08/16/2023] Open
Abstract
Acute myocardial infarction has long been the leading cause of death in coronary heart disease, which is characterized by irreversible cardiomyocyte death and restricted blood supply. Conventional reperfusion therapy can further aggravate myocardial injury. Stem cell therapy, especially with mesenchymal stem cells (MSCs), has emerged as a promising approach to promote cardiac repair and improve cardiac function. MSCs may induce these effects by secreting exosomes containing therapeutically active RNA, proteins and lipids. Notably, normal cardiac function depends on intracardiac paracrine signaling via exosomes, and exosomes secreted by cardiac cells can partially reflect changes in the heart during disease, so analyzing these vesicles may provide valuable insights into the pathology of myocardial infarction as well as guide the development of new treatments. The present review examines how exosomes produced by MSCs and cardiac cells may influence injury after myocardial infarction and serve as therapies against such injury. Video Abstract.
Collapse
Affiliation(s)
- Yuchang Liu
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Minrui Wang
- School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Yang Yu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Chunxiang Zhang
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- The Key Laboratory of Medical Electrophysiology of the Ministry of Education, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Laboratory of Nucleic Acids in Medicine for National High-Level Talents, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
44
|
Castillo-Casas JM, Caño-Carrillo S, Sánchez-Fernández C, Franco D, Lozano-Velasco E. Comparative Analysis of Heart Regeneration: Searching for the Key to Heal the Heart-Part I: Experimental Injury Models to Study Cardiac Regeneration. J Cardiovasc Dev Dis 2023; 10:325. [PMID: 37623338 PMCID: PMC10455172 DOI: 10.3390/jcdd10080325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/28/2023] [Accepted: 07/30/2023] [Indexed: 08/26/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide, among which, ischemic heart disease is the most prevalent. Myocardial infarction results from occlusion of a coronary artery, which leads to an insufficient blood supply to the myocardium. As is well known, the massive loss of cardiomyocytes cannot be solved due the limited regenerative ability of the adult mammalian heart. In contrast, some lower vertebrate species can regenerate the heart after injury; their study has disclosed some of the involved cell types, molecular mechanisms and signaling pathways during the regenerative process. In this two-part review, we discuss the current state of the principal response in heart regeneration, where several involved processes are essential for full cardiac function in recovery.
Collapse
Affiliation(s)
- Juan Manuel Castillo-Casas
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Sheila Caño-Carrillo
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
| | - Cristina Sánchez-Fernández
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Diego Franco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| | - Estefanía Lozano-Velasco
- Cardiovascular Development Group, Department of Experimental Biology, University of Jaén, 23071 Jaén, Spain; (J.M.C.-C.); (S.C.-C.); (C.S.-F.); (D.F.)
- Medina Foundation, 18007 Granada, Spain
| |
Collapse
|
45
|
Hu Y, Cang J, Hiraishi K, Fujita T, Inoue R. The Role of TRPM4 in Cardiac Electrophysiology and Arrhythmogenesis. Int J Mol Sci 2023; 24:11798. [PMID: 37511555 PMCID: PMC10380800 DOI: 10.3390/ijms241411798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/20/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The transient receptor potential melastatin 4 (TRPM4) channel is a non-selective cation channel that activates in response to increased intracellular Ca2+ levels but does not allow Ca2+ to pass through directly. It plays a crucial role in regulating diverse cellular functions associated with intracellular Ca2+ homeostasis/dynamics. TRPM4 is widely expressed in the heart and is involved in various physiological and pathological processes therein. Specifically, it has a significant impact on the electrical activity of cardiomyocytes by depolarizing the membrane, presumably via Na+ loading. The TRPM4 channel likely contributes to the development of cardiac arrhythmias associated with specific genetic backgrounds and cardiac remodeling. This short review aims to overview what is known so far about the TRPM4 channel in cardiac electrophysiology and arrhythmogenesis, highlighting its potential as a novel therapeutic target to effectively prevent and treat cardiac arrhythmias.
Collapse
Affiliation(s)
- Yaopeng Hu
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Jiehui Cang
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Keizo Hiraishi
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Takayuki Fujita
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| | - Ryuji Inoue
- Department of Physiology, Fukuoka University School of Medicine, Fukuoka 814-0180, Japan
| |
Collapse
|
46
|
Wang Y, Guo L, Zhang Z, Fu S, Huang P, Wang A, Liu M, Ma X. A bibliometric analysis of myocardial ischemia/reperfusion injury from 2000 to 2023. Front Cardiovasc Med 2023; 10:1180792. [PMID: 37383699 PMCID: PMC10293770 DOI: 10.3389/fcvm.2023.1180792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Background Myocardial ischemia/reperfusion injury (MIRI) refers to the more severe damage that occurs in the previously ischemic myocardium after a short-term interruption of myocardial blood supply followed by restoration of blood flow within a certain period of time. MIRI has become a major challenge affecting the therapeutic efficacy of cardiovascular surgery. Methods A scientific literature search on MIRI-related papers published from 2000 to 2023 in the Web of Science Core Collection database was conducted. VOSviewer was used for bibliometric analysis to understand the scientific development and research hotspots in this field. Results A total of 5,595 papers from 81 countries/regions, 3,840 research institutions, and 26,202 authors were included. China published the most papers, but the United States had the most significant influence. Harvard University was the leading research institution, and influential authors included Lefer David J., Hausenloy Derek J., Yellon Derek M., and others. All keywords can be divided into four different directions: risk factors, poor prognosis, mechanisms and cardioprotection. Conclusion Research on MIRI is flourishing. It is necessary to conduct an in-depth investigation of the interaction between different mechanisms and multi-target therapy will be the focus and hotspot of MIRI research in the future.
Collapse
Affiliation(s)
- Yifei Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Lijun Guo
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Zhibo Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Shuangqing Fu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Pingping Huang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mi Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| |
Collapse
|
47
|
Li Z, Wang H, Zoungrana LI, James A, Slotabec L, Didik S, Fatmi MK, Krause-Hauch M, Lesnefsky EJ, Li J. Administration of metformin rescues age-related vulnerability to ischemic insults through mitochondrial energy metabolism. Biochem Biophys Res Commun 2023; 659:46-53. [PMID: 37031594 PMCID: PMC10190118 DOI: 10.1016/j.bbrc.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/03/2023] [Indexed: 04/08/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of death on a global scale. Despite significant advances in the reperfusion treatment of acute myocardial infarction, there is still a significant early mortality rate among the elderly, as angioplasty-achieved reperfusion can exacerbate myocardial damage, leading to severe ischemia/reperfusion (I/R) injury and induce fatal arrhythmias. Mitochondria are a key mediator of ischemic insults; a transient blockade of the electron transport chain (ETC) at complex I during reperfusion can reduce myocardial infarct caused by ischemic insults. The reversible, transient modulation of complex I during early reperfusion is limited by the available of clinically tractable agents. We employed the novel use of acute, high dose metformin to modulate complex I activity during early reperfusion to decrease cardiac injury in the high-risk aged heart. Young (3-6 months) and aged (22-24 months) male and female C57BL/6 J mice were subjected to in vivo regional ischemia for 45 min, followed by metformin (2 mM, i. v.) injection 5 min prior to reperfusion for 24 h. The cardiac functions were measured with echocardiography. A Seahorse XF24 Analyzer was used to ascertain mitochondrial function. Cardiomyocyte sarcomere shortening and calcium transients were measured using the IonOptix Calcium and Contractility System. The results demonstrated that administration of acute, high dose metformin at the onset of reperfusion significantly limited cardiac damage and rescued cardiac dysfunction caused by I/R in both young and aged mice. Importantly, metformin treatment improves contractile functions of isolated cardiomyocytes and maintains mitochondrial integrity under I/R stress conditions. Thus, acute metformin administration at the onset of reperfusion has potential as a mitochondrial-based therapeutic to mitigate reperfusion injury and reduce infarct size in the elderly heart attack patient who remains at greater mortality risk despite reperfusion alone.
Collapse
Affiliation(s)
- Zehui Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Hao Wang
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Linda Ines Zoungrana
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Adewale James
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Lily Slotabec
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Steven Didik
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Mohammad Kasim Fatmi
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Meredith Krause-Hauch
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; James A. Haley Veterans' Hospital, Tampa, FL, USA
| | - Edward J Lesnefsky
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA; Cardiology Section, Medical Service, Richmond Department of Veterans Affairs Medical Center, Richmond, VA, USA
| | - Ji Li
- Department of Surgery, Morsani College of Medicine, University of South Florida, Tampa, FL, USA; James A. Haley Veterans' Hospital, Tampa, FL, USA.
| |
Collapse
|
48
|
Ma K, Liu W, Liu Q, Hu P, Bai L, Yu M, Yang Y. Naringenin facilitates M2 macrophage polarization after myocardial ischemia-reperfusion by promoting nuclear translocation of transcription factor EB and inhibiting the NLRP3 inflammasome pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:1405-1419. [PMID: 36988289 DOI: 10.1002/tox.23774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 02/22/2023] [Accepted: 02/25/2023] [Indexed: 05/18/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) remains an unsolved puzzle in medical circles. Naringenin (NAR) is a flavonoid with cardioprotective potential. The purpose of this article was to discuss the protective mechanism of NAR in MIRI by regulating macrophage polarization. The MIRI mouse model was established and perfused with NAR before surgery. In the in vitro experiment, macrophages RAW264.7 were treated with lipopolysaccharide to induce M1 polarization after pretreatment with NAR. Rescue experiments were carried out to validate the functions of transcription factor EB (TFEB), the NLR pyrin domain containing 3 (NLRP3) inflammasome, and autophagy in macrophage polarization. NAR reduced histopathological injury and infarction of myocardial tissues in MIRI mice, inhibited M1 polarization and promoted M2 polarization of macrophages, diminished levels of pro-inflammatory factors, and augmented levels of anti-inflammatory factors. NAR facilitated TFEB nuclear translocation and inhibited the NLRP3 inflammasome pathway. Silencing TFEB or Nigericin partly nullified the effect of NAR on macrophage polarization. NAR increased autophagosome formation, autophagy flux, and autophagy level. Autophagy inhibitor 3-methyladenine partly invalidated the inhibition of NAR on the NLRP3 inflammasome pathway. In animal experiments, NAR protected MIRI mice through the TFEB-autophagy-NLRP3 inflammasome pathway. Collectively, NAR inhibited NLRP3 inflammasome activation and facilitated M2 macrophage polarization by stimulating TFEB nuclear translocation, thus protecting against MIRI.
Collapse
Affiliation(s)
- Kuiying Ma
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Wenqing Liu
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Qi Liu
- Emergency Department, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Pengfei Hu
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Lingyu Bai
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Miao Yu
- Department of Cardiovascular Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| | - Yan Yang
- Department of General Medicine, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao City, China
| |
Collapse
|
49
|
Yap J, Irei J, Lozano-Gerona J, Vanapruks S, Bishop T, Boisvert WA. Macrophages in cardiac remodelling after myocardial infarction. Nat Rev Cardiol 2023; 20:373-385. [PMID: 36627513 DOI: 10.1038/s41569-022-00823-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2022] [Indexed: 01/12/2023]
Abstract
Myocardial infarction (MI), as a result of thrombosis or vascular occlusion, is the most prevalent cause of morbidity and mortality among all cardiovascular diseases. The devastating consequences of MI are compounded by the complexities of cellular functions involved in the initiation and resolution of early-onset inflammation and the longer-term effects related to scar formation. The resultant tissue damage can occur as early as 1 h after MI and activates inflammatory signalling pathways to elicit an immune response. Macrophages are one of the most active cell types during all stages after MI, including the cardioprotective, inflammatory and tissue repair phases. In this Review, we describe the phenotypes of cardiac macrophage involved in MI and their cardioprotective functions. A specific subset of macrophages called resident cardiac macrophages (RCMs) are derived from yolk sac progenitor cells and are maintained as a self-renewing population, although their numbers decrease with age. We explore sophisticated sequencing techniques that demonstrate the cardioprotective properties of this cardiac macrophage phenotype. Furthermore, we discuss the interactions between cardiac macrophages and other important cell types involved in the pathology and resolution of inflammation after MI. We summarize new and promising therapeutic approaches that target macrophage-mediated inflammation and the cardioprotective properties of RCMs after MI. Finally, we discuss future directions for the study of RCMs in MI and cardiovascular health in general.
Collapse
Affiliation(s)
- Jonathan Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Jason Irei
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Javier Lozano-Gerona
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Selena Vanapruks
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - Tianmai Bishop
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, USA.
| |
Collapse
|
50
|
Sun M, Jiang W, Mu N, Zhang Z, Yu L, Ma H. Mitochondrial transplantation as a novel therapeutic strategy for cardiovascular diseases. J Transl Med 2023; 21:347. [PMID: 37231493 DOI: 10.1186/s12967-023-04203-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/13/2023] [Indexed: 05/27/2023] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of noncommunicable disease-related death worldwide, and effective therapeutic strategies against CVD are urgently needed. Mitochondria dysfunction involves in the onset and development of CVD. Nowadays, mitochondrial transplantation, an alternative treatment aimed at increasing mitochondrial number and improving mitochondrial function, has been emerged with great therapeutic potential. Substantial evidence indicates that mitochondrial transplantation improves cardiac function and outcomes in patients with CVD. Therefore, mitochondrial transplantation has profound implications in the prevention and treatment of CVD. Here, we review the mitochondrial abnormalities that occur in CVD and summarize the therapeutic strategies of mitochondrial transplantation for CVD.
Collapse
Affiliation(s)
- Mingchu Sun
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, P.R. China
| | - Wenhua Jiang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, P.R. China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Zihui Zhang
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, P.R. China.
| | - Lu Yu
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|