1
|
Cheng H, Wu X, Li J, Wang L. CircRNA Networks in CAD: Multi-Cellular Mechanisms and Clinical Potential. Int J Gen Med 2025; 18:3129-3150. [PMID: 40529348 PMCID: PMC12170827 DOI: 10.2147/ijgm.s524189] [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: 02/21/2025] [Accepted: 05/26/2025] [Indexed: 06/20/2025] Open
Abstract
Coronary artery disease (CAD), is a global cardiovascular disease that is characterized by myocardial ischemia and hypoxia caused by coronary artery occlusion. Circular RNAs (CircRNAs) is a particular kind of endogenous non-coding RNA, which can affect the occurrence and development of CAD. Concurrently, several circRNAs display stable persistence in CAD patients, attributable to their exceptional exonuclease resistance, thereby harboring the capacity to evolve into a biomarker for CAD diagnosis and prognosis. This article endeavors to clarify the pivotal role of circRNAs in the intricate pathophysiological processes underlying CAD patients or CAD disease models based on their unique biological characteristics and functionalities, and further discuss their prospects in clinical applications of CAD.
Collapse
Affiliation(s)
- Huan Cheng
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, People’s Republic of China
| | - Xinyu Wu
- Department of Cardiology, Zhumadian City Central Hospital, Zhumadian, Henan, 463000, People’s Republic of China
| | - Jingru Li
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, People’s Republic of China
| | - Luqiao Wang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650032, People’s Republic of China
| |
Collapse
|
2
|
Liu CP, Zheng S, Zhang P, Chen GH, Zhang YY, Sun HL, Peng L. Decreased serum SLC7A11 and GPX4 levels may reflect disease severity of acute ischaemic stroke. Ann Clin Biochem 2025; 62:191-201. [PMID: 39632577 DOI: 10.1177/00045632241305927] [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: 12/07/2024]
Abstract
ObjectiveThis study aimed to examine the levels of solute carrier family seven number 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) in the serum of patients with acute ischaemic stroke (AIS) and their relationship with disease severity.MethodsA total of 148 patients with AIS together with 148 healthy controls (HCs) were enrolled. The expression levels of SLC7A11 and GPX4 in serum were detected immediately as early as possible. Radiographic severity was detected by Alberta Stroke Program Early CT Score (ASPECTS). Disease severity was evaluated using modified Rankin Scale (mRS). High-sensitivity C-reactive protein (hs-CRP) and matrix metalloproteinase-9 (MMP-9) expression levels were also measured. A correlation analysis was conducted to determine the relationship between the expression levels of SLC7A11 and GPX4 with the clinical severity of the disease and the levels of hs-CRP and MMP-9. Furthermore, receiver operating characteristic (ROC) curve analysis was utilized to assess the potential of SLC7A11 and GPX4 as diagnostic markers.ResultsCompared to the HC group, the serum expression levels of SLC7A11 and GPX4 were significantly lower in the AIS group. Serum SLC7A11 levels were positively associated with serum GPX4 levels. The AIS group included 50 patients with mild neurological impairment, 52 with moderate neurological impairment, and 46 with severe neurological impairment. AIS patients with mild neurological impairment had drastically higher serum SLC7A11 and GPX4 levels compared with those with moderate neurological impairment. AIS patients with moderate neurological impairment showed significantly higher serum SLC7A11 and GPX4 concentrations compared with those with severe neurological impairment. ROC curve analysis demonstrated that both serum SLC7A11 and GPX4 may both act as potential indicators for evaluating of AIS disease severity. In addition, both serum SLC7A11 and GPX4 levels were positively correlated with ASPECTS. Both serum SLC7A11 and GPX4 levels were negatively associated with hs-CRP as well as MMP-9 levels. Serum SLC7A11 and GPX4 levels were significantly increased following comprehensive therapy.ConclusionsDecreased SLC7A11 and GPX4 levels may reflect disease severity of AIS.
Collapse
Affiliation(s)
| | - Su Zheng
- Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ping Zhang
- Department of Acupuncture, Shiyan Hospital of Traditional Chinese Medicine, Shiyan, China
| | - Guang-Hui Chen
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Yuan-Yuan Zhang
- Department of Neurology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Hui-Lin Sun
- Department of Radiology, Renmin Hospital, Hubei University of Medicine, Shiyan, China
| | - Li Peng
- Shiyan Hospital of Traditional Chinese Medicine, Hubei University of Chinese Medicine, Shiyan, China
| |
Collapse
|
3
|
Han L, Zhai W. Mechanisms and preventive measures of ALDH2 in ischemia‑reperfusion injury: Ferroptosis as a novel target (Review). Mol Med Rep 2025; 31:105. [PMID: 40017132 PMCID: PMC11876945 DOI: 10.3892/mmr.2025.13470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/31/2025] [Indexed: 03/01/2025] Open
Abstract
Ischemia‑reperfusion injury (IRI) refers to tissue or organ damage that occurs following a period of inadequate blood supply (ischemia) followed by restoration of blood flow (reperfusion) within a short time frame. This phenomenon is prevalent in clinical conditions such as cardiovascular and cerebrovascular disease, organ transplantation and stroke. Despite its frequency, effective therapeutic strategies to mitigate IRI remain elusive in clinical practice, underscoring the need for a deeper understanding of its molecular mechanisms. Aldehyde dehydrogenase 2 (ALDH2), a key enzyme in alcohol metabolism, serves a role in alleviating oxidative stress and cell damage during IRI by modulating oxidative stress, decreasing apoptosis and inhibiting inflammatory responses. ALDH2 exerts protective effects by detoxifying reactive aldehydes, thereby preventing lipid peroxidation and maintaining cellular homeostasis. Furthermore, ferroptosis, a regulated form of cell death driven by iron accumulation and subsequent lipid peroxidation, is a key process in IRI. However, the precise role of ALDH2 in modulating ferroptosis during IRI remains incompletely understood. Although there is an interaction between ALDH2 activity and ferroptosis, the underlying mechanisms have yet to be clarified. The present review examines the role of ALDH2 and ferroptosis in IRI and the potential regulatory influence of ALDH2 on ferroptosis mechanisms, as well as potential targeting of ALDH2 and ferroptosis for IRI treatment and prevention. By elucidating the complex interplay between ALDH2 and ferroptosis, the present review aims to provide new insights for the development of innovative therapeutic strategies to mitigate ischemic tissue damage and improve clinical outcomes.
Collapse
Affiliation(s)
- Liang Han
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, P.R. China
| | - Wen Zhai
- Center for Rehabilitation Medicine, Department of Anesthesiology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
4
|
Liu H, Song Y, Wang H, Zhou Y, Xu M, Xian J. Deciphering the Power of Resveratrol in Mitophagy: From Molecular Mechanisms to Therapeutic Applications. Phytother Res 2025; 39:1319-1343. [PMID: 39754508 DOI: 10.1002/ptr.8433] [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: 10/30/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 01/06/2025]
Abstract
Resveratrol (RES), a natural polyphenolic compound, has garnered significant attention for its therapeutic potential in various pathological conditions. This review explores how RES modulates mitophagy-the selective autophagic degradation of mitochondria essential for maintaining cellular homeostasis. RES promotes the initiation and execution of mitophagy by enhancing PINK1/Parkin-mediated mitochondrial clearance, reducing reactive oxygen species production, and mitigating apoptosis, thereby preserving mitochondrial integrity. Additionally, RES regulates mitophagy through the activation of key molecular targets such as AMP-activated protein kinase (AMPK), the mechanistic target of rapamycin (mTOR), deacetylases (SIRT1 and SIRT3), and mitochondrial quality control (MQC) pathways, demonstrating substantial therapeutic effects in multiple disease models. We provide a detailed account of the biosynthetic pathways, pharmacokinetics, and metabolic characteristics of RES, focusing on its role in mitophagy modulation and implications for medical applications. Potential adverse effects associated with its clinical use are also discussed. Despite its promising therapeutic properties, the clinical application of RES is limited by issues of bioavailability and pharmacokinetic profiles. Future research should concentrate on enhancing RES bioavailability and developing derivatives that precisely modulate mitophagy, thereby unlocking new avenues for disease therapy.
Collapse
Affiliation(s)
- Hongmei Liu
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Yixuan Song
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huan Wang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Zhou
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Xu
- Department of Pharmacy, Affiliated Hospital of Southwest Jiaotong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Jiaxun Xian
- Traditional Chinese Medicine Hospital of Meishan, Meishan, China
| |
Collapse
|
5
|
Tian Y, Hu X, Zhang T, Li B, Fu Q, Li J. Advances in Chinese herbal medicine in modulating mitochondria to treat myocardial ischemia-reperfusion injury: a narrative review. Cardiovasc Diagn Ther 2025; 15:207-232. [PMID: 40115104 PMCID: PMC11921369 DOI: 10.21037/cdt-24-346] [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: 07/22/2024] [Accepted: 11/28/2024] [Indexed: 03/23/2025]
Abstract
Background and Objective The urgent need to identify pathways that can mitigate myocardial ischemia-reperfusion injury (MIRI) has become a central focus in cardiovascular treatment. Chinese herbal medicine (CHM), renowned for its multi-component, multi-channel, and multi-target therapeutic properties, holds significant promise in the management of MIRI. Mitochondria, as pivotal players in MIRI, have been shown to be effectively modulated by CHM through various mechanisms. The objective of this narrative review is to underscore the critical role of mitochondria in MIRI and to provide an up-to-date overview of the latest research advancements in utilizing CHM to treat MIRI by targeting mitochondrial morphology and function. Methods The PubMed and the China National Knowledge Infrastructure (CNKI) databases were searched using keywords related to MIRI. Relevant English-language articles published from January 2019 to July 2024 were included in this narrative review. Key Content and Findings Mitochondria are intimately linked to MIRI. The mechanisms involve the regulation of mitochondrial biogenesis and energy metabolism, the functionality of the mitochondrial respiratory chain, resistance to oxidative stress-induced damage, the maintenance of mitochondrial homeostasis, the modulation of calcium ion homeostasis, the preservation of mitochondrial membrane potential, the opening of adenosine triphosphate (ATP)-sensitive potassium channels, and the effective control over the opening of the mitochondrial permeability transition pore, all of which contribute to the balance between autophagy and apoptosis in cardiomyocytes. Various effective monomers of CHM, extracts of CHM, compounds, and proprietary Chinese medicine have demonstrated promising therapeutic potential in basic research, among them, tonic and blood-activating CHMs account for the largest proportion. Conclusions The prospect of CHM targeting mitochondria for the treatment of MIRI is promising, yet it necessitates overcoming challenges such as low bioavailability and inadequate mechanistic research. By integrating traditional Chinese medicine theories with modern scientific technologies, it is imperative to delve deeper into and optimize the pharmacodynamics, pharmacokinetics, and clinical applications of these herbs.
Collapse
Affiliation(s)
- Yushi Tian
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyang Hu
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tingyu Zhang
- Acupuncture and Tuina Science, Liaoning University of Chinese Medicine, Shenyang, China
| | - Bojia Li
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qiang Fu
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ji Li
- School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
6
|
Pandolfi S, Valdenassi L, Franzini M, Simonetti V, Chirumbolo S. Insights into the use of oxygen-ozone therapy in ischemic cardiopathy and cardiovascular disease: a role for mitochondria? Med Gas Res 2024; 14:164-166. [PMID: 40434379 PMCID: PMC11257178 DOI: 10.4103/mgr.medgasres-d-23-00007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/15/2023] [Accepted: 11/22/2023] [Indexed: 05/29/2025] Open
Affiliation(s)
- Sergio Pandolfi
- Italian Scientific Society of Oxygen Ozone Therapy (SIOOT), Bergamo, Italy and High School Master in Oxygen-Ozone Therapy University of Pavia, Pavia, Italy
| | - Luigi Valdenassi
- Italian Scientific Society of Oxygen Ozone Therapy (SIOOT), Bergamo, Italy and High School Master in Oxygen-Ozone Therapy University of Pavia, Pavia, Italy
| | - Marianno Franzini
- Italian Scientific Society of Oxygen Ozone Therapy (SIOOT), Bergamo, Italy and High School Master in Oxygen-Ozone Therapy University of Pavia, Pavia, Italy
| | - Vincenzo Simonetti
- Italian Scientific Society of Oxygen Ozone Therapy (SIOOT), Bergamo, Italy and High School Master in Oxygen-Ozone Therapy University of Pavia, Pavia, Italy
| | - Salvatore Chirumbolo
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| |
Collapse
|
7
|
Tian L, Liu Q, Guo H, Zang H, Li Y. Fighting ischemia-reperfusion injury: Focusing on mitochondria-derived ferroptosis. Mitochondrion 2024; 79:101974. [PMID: 39461581 DOI: 10.1016/j.mito.2024.101974] [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: 07/04/2024] [Revised: 09/12/2024] [Accepted: 10/12/2024] [Indexed: 10/29/2024]
Abstract
Ischemia-reperfusion injury (IRI) is a major cause of mortality and morbidity. Current treatments for IRI have limited efficacy and novel therapeutic strategies are needed. Mitochondrial dysfunction not only initiates IRI but also plays a significant role in ferroptosis pathogenesis. Recent studies have highlighted that targeting mitochondrial pathways is a promising therapeutic approach for ferroptosis-induced IRI. The association between ferroptosis and IRI has been reviewed many times, but our review provides the first comprehensive overview with a focus on recent mitochondrial research. First, we present the role of mitochondria in ferroptosis. Then, we summarize the evidence on mitochondrial manipulation of ferroptosis in IRI and review recent therapeutic strategies aimed at targeting mitochondria-related ferroptosis to mitigate IRI. We hope our review will provide new ideas for the treatment of IRI and accelerate the transition from bench to bedside.
Collapse
Affiliation(s)
- Lei Tian
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Qian Liu
- Department of Anesthesiology, Zigong First People's Hospital, Zigong Academy of Medical Sciences, Zigong, China
| | - Hong Guo
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Honggang Zang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Yulan Li
- Department of Anesthesiology, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
8
|
Zhu H, Zhu T, Dubiao D, Zhang X. Metformin Attenuates Myocardial Ischemia-Reperfusion Injury through the AMPK-HMGCR-ROS Signaling Axis. KARDIOLOGIIA 2024; 64:48-56. [PMID: 39526518 DOI: 10.18087/cardio.2024.10.n2739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/06/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVE To explore the role and mechanism of metformin (MET) in regulating myocardial injury caused by cardiac ischemia-reperfusion. MATERIAL AND METHODS A rat model of myocardial ischemia-reperfusion injury was established by ligation of the anterior descending branch of the left coronary artery. The myocardial area at risk and the infarction size were measured by Evans blue and 2,3,5‑triphenyltetrazole chloride (TTC) staining, respectively. Terminal Deoxynucleotidyl Transferase-Mediated dUTP Nick End Labeling (TUNEL) staining was used to detect apoptosis of cardiomyocytes. The expression of 4‑hydroxynonenal (4‑HNE) was detected by immunohistochemical staining. Real-time quantitative polymerase chain reaction (RT-PCR) and Western blot were used to detect mRNA and expression of the Adenosine 5'-monophosphate-activated protein kinase (AMPK) - 3‑hydroxy-3‑methylglutaryl-CoA reductase (HMGCR) signaling pathway, respectively. RESULTS MET treatment decreased the infarct size and the activity of the myocardial enzyme profile, thus demonstrating protection of ischemic myocardium. The number of TUNEL positive cells significantly decreased. Immunohistochemical results showed that MET decreased the expression of 4‑HNE in myocardial tissue and the content of malondialdehyde (MDA) in myocardial cells. Further experimental results showed that MET decreased HMGCR transcription and protein expression, and increased AMPK phosphorylation. In the model of hypoxia and reoxygenation injury of cardiomyocytes, MET increased the viability of cardiomyocytes, decreased the activity of lactic dehydrogenase (LDH), decreased malondialdehyde content and intracellular reactive oxygen species (ROS) concentrations, and regulate the AMPK-HMGCR signaling pathway through coenzyme C (ComC). CONCLUSION MET inhibits the expression of HMGCR by activating AMPK, reduces oxidative damage and apoptosis of cardiomyocytes, and alleviates myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- He Zhu
- Department of Vascular Surgery, Zhejiang Chinese Medical University
| | - Tao Zhu
- Department of Vascular Surgery, Zhejiang Chinese Medical University
| | - Dubiao Dubiao
- Department of Cardiology, Kecheng District People's Hospital
| | - Xinmei Zhang
- Department of Vascular Surgery, Quzhou People's Hospital, the Quzhou Affiliated Hospital of Wenzhou Medical University
| |
Collapse
|
9
|
Huan R, Zhang J, Yue J, Yang S, Han G, Cheng Y, Tan Y. Orexin-A mediates glioblastoma proliferation inhibition by increasing ferroptosis triggered by unstable iron pools and GPX4 depletion. J Cell Mol Med 2024; 28:e18318. [PMID: 38685674 PMCID: PMC11058333 DOI: 10.1111/jcmm.18318] [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: 11/01/2023] [Revised: 03/21/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024] Open
Abstract
Glioblastoma (GBM) represents a prevalent form of primary malignant tumours in the central nervous system, but the options for effective treatment are extremely limited. Ferroptosis, as the most enriched programmed cell death process in glioma, makes a critical difference in glioma progression. Consequently, inducing ferroptosis has become an appealing strategy for tackling gliomas. Through the utilization of multi-omics sequencing data analysis, flow cytometry, MDA detection and transmission electron microscopy, the impact of orexin-A on ferroptosis in GBM was assessed. In this report, we provide the first evidence that orexin-A exerts inhibitory effects on GBM proliferation via the induction of ferroptosis. This induction is achieved by instigating an unsustainable increase in iron levels and depletion of GPX4. Moreover, the regulation of TFRC, FTH1 and GPX4 expression through the targeting of NFE2L2 appears to be one of the potential mechanisms underlying orexin-A-induced ferroptosis.
Collapse
Affiliation(s)
- Rengzheng Huan
- Department of NeurosurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Jiqin Zhang
- Department of AnesthesiologyGuizhou Provincial People's HospitalGuiyangChina
| | - Jianhe Yue
- Department of NeurosurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Sha Yang
- Department of biomedical sciencesMedical College of Guizhou UniversityGuiyangChina
| | - Guoqiang Han
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Yuan Cheng
- Department of NeurosurgeryThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Ying Tan
- Department of NeurosurgeryGuizhou Provincial People's HospitalGuiyangChina
| |
Collapse
|
10
|
Mogavero MP, Salemi M, Lanza G, Rinaldi A, Marchese G, Ravo M, Salluzzo MG, Antoci A, DelRosso LM, Bruni O, Ferini-Strambi L, Ferri R. Unveiling the pathophysiology of restless legs syndrome through transcriptome analysis. iScience 2024; 27:109568. [PMID: 38617564 PMCID: PMC11015462 DOI: 10.1016/j.isci.2024.109568] [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: 11/05/2023] [Revised: 01/22/2024] [Accepted: 03/22/2024] [Indexed: 04/16/2024] Open
Abstract
The aim of this study was to analyze signaling pathways associated with differentially expressed messenger RNAs in people with restless legs syndrome (RLS). Seventeen RLS patients and 18 controls were enrolled. Coding RNA expression profiling of 12,857 gene transcripts by next-generation sequencing was performed. Enrichment analysis by pathfindR tool was carried-out, with p-adjusted ≤0.001 and fold-change ≥2.5. Nine main different network groups were significantly dysregulated in RLS: infections, inflammation, immunology, neurodegeneration, cancer, neurotransmission and biological, blood and metabolic mechanisms. Genetic predisposition plays a key role in RLS and evidence indicates its inflammatory nature; the high involvement of mainly neurotropic viruses and the TORCH complex might trigger inflammatory/immune reactions in genetically predisposed subjects and activate a series of biological pathways-especially IL-17, receptor potential channels, nuclear factor kappa-light-chain-enhancer of activated B cells, NOD-like receptor, mitogen-activated protein kinase, p53, mitophagy, and ferroptosis-involved in neurotransmitter mechanisms, synaptic plasticity, axon guidance, neurodegeneration, carcinogenesis, and metabolism.
Collapse
Affiliation(s)
- Maria P. Mogavero
- Vita-Salute San Raffaele University, 20132 Milan, Italy
- San Raffaele Scientific Institute, Division of Neuroscience, Sleep Disorders Center, 20127 Milan, Italy
| | | | - Giuseppe Lanza
- Oasi Research Institute-IRCCS, 94018 Troina, Italy
- University of Catania, Department of Surgery and Medical-Surgical Specialties, 95123 Catania, Italy
| | - Antonio Rinaldi
- Genomix4Life Srl, 84081 Baronissi, Italy
- Genome Research Center for Health-CRGS, 84081 Baronissi, Italy
| | - Giovanna Marchese
- Genomix4Life Srl, 84081 Baronissi, Italy
- Genome Research Center for Health-CRGS, 84081 Baronissi, Italy
| | - Maria Ravo
- Genomix4Life Srl, 84081 Baronissi, Italy
- Genome Research Center for Health-CRGS, 84081 Baronissi, Italy
| | | | | | | | - Oliviero Bruni
- Sapienza University of Rome, Developmental and Social Psychology, 00185 Rome, Italy
| | - Luigi Ferini-Strambi
- Vita-Salute San Raffaele University, 20132 Milan, Italy
- San Raffaele Scientific Institute, Division of Neuroscience, Sleep Disorders Center, 20127 Milan, Italy
| | | |
Collapse
|
11
|
Jin C, Zhong Z, Gao L, Wu X, Zhou C, Zhou G, Liu S. Focus on the Role of Inflammation as a Bridge between Ferroptosis and Atrial Fibrillation: A Narrative Review and Novel Perspective. Rev Cardiovasc Med 2024; 25:110. [PMID: 39076556 PMCID: PMC11264012 DOI: 10.31083/j.rcm2504110] [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: 10/05/2023] [Revised: 11/19/2023] [Accepted: 11/28/2023] [Indexed: 07/31/2024] Open
Abstract
In this comprehensive review, we examine the intricate interplay between inflammation, ferroptosis, and atrial fibrillation (AF), highlighting their significant roles in AF pathophysiology and pathogenesis. Augmented inflammatory responses are pivotal to AF, potentially leading to atrial remodeling and reentry phenomena by impacting calcium channels and atrial tissue fibrosis. A strong correlation exists between inflammatory cytokines and AF, underscoring the importance of inflammatory signaling pathways, such as NOD-like receptor thermal protien domain associated protein 3 (NLRP3) inflammasome, Nuclear Factor kappa B (NF- κ B) signaling, and Tumor necrosis factor- α (TNF- α ) signaling in AF development. Ferroptosis, a non-apoptotic regulated mode of cell death, has been widely studied in relation to cardiovascular diseases including heart failure, myocardial infarction, cardiomyopathy, and reperfusion injury. The interaction between ferroptosis and inflammation is complex and mutually influential. While significant progress has been made in understanding the inflammation-AF relationship, the role of inflammation as a conduit linking ferroptosis and AF remains underexplored. The specific pathogenesis and key molecules of atrial fibrosis caused by ferroptosis are still not fully understood. Here we review the role of inflammatory signaling in ferroptosis and AF. We elucidated the association between ferroptosis and AF, aiming to unveil mechanisms for targeted inhibition of atrial cell fibrosis and to propose novel therapeutic strategies for AF. This exploration is vital for advancing our knowledge and developing more effective interventions for AF, a condition deeply intertwined with inflammatory processes and ferroptotic pathways.
Collapse
Affiliation(s)
- Chenyang Jin
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong
University School of Medicine, 201620 Shanghai, China
| | - Zikan Zhong
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong
University School of Medicine, 201620 Shanghai, China
| | - Longzhe Gao
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong
University School of Medicine, 201620 Shanghai, China
| | - Xiaoyu Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong
University School of Medicine, 201620 Shanghai, China
| | - Changzuan Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong
University School of Medicine, 201620 Shanghai, China
| | - Genqing Zhou
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong
University School of Medicine, 201620 Shanghai, China
| | - Shaowen Liu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong
University School of Medicine, 201620 Shanghai, China
| |
Collapse
|
12
|
Yu M, Cheng X. Editorial Commentary: Top Five Stories of the Cellular Landscape and Therapies of Atherosclerosis: Current Knowledge and Future Perspectives. Curr Med Sci 2024; 44:241-243. [PMID: 38277018 DOI: 10.1007/s11596-023-2825-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Affiliation(s)
- Miao Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiang Cheng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
13
|
Wang H, Huang Z, Du C, Dong M. Iron Dysregulation in Cardiovascular Diseases. Rev Cardiovasc Med 2024; 25:16. [PMID: 39077672 PMCID: PMC11263000 DOI: 10.31083/j.rcm2501016] [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: 08/07/2023] [Revised: 10/07/2023] [Accepted: 10/24/2023] [Indexed: 07/31/2024] Open
Abstract
Iron metabolism plays a crucial role in various physiological functions of the human body, as it is essential for the growth and development of almost all organisms. Dysregulated iron metabolism-manifested either as iron deficiency or overload-is a significant risk factor for the development of cardiovascular disease (CVD). Moreover, emerging evidence suggests that ferroptosis, a form of iron-dependent programed cell death, may also contribute to CVD development. Understanding the regulatory mechanisms of iron metabolism and ferroptosis in CVD is important for improving disease management. By integrating different perspectives and expertise in the field of CVD-related iron metabolism, this overview provides insights into iron metabolism and CVD, along with approaches for diagnosing, treating, and preventing CVD associated with iron dysregulation.
Collapse
Affiliation(s)
- Hui Wang
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and
Translation, Chengdu Fifth People's Hospital, 611137 Chengdu, Sichuan, China
| | - Zhongmin Huang
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and
Translation, Chengdu Fifth People's Hospital, 611137 Chengdu, Sichuan, China
| | - Chenyan Du
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and
Translation, Chengdu Fifth People's Hospital, 611137 Chengdu, Sichuan, China
| | - Mingqing Dong
- Geriatric Diseases Institute of Chengdu, Center for Medicine Research and
Translation, Chengdu Fifth People's Hospital, 611137 Chengdu, Sichuan, China
| |
Collapse
|
14
|
Wang J, Xu RM, Cao QM, Ma BC, Zhang H, Hao HP. Mechanism of DYRK1a in myocardial ischemia-reperfusion injury by regulating ferroptosis of cardiomyocytes. Kaohsiung J Med Sci 2023; 39:1190-1199. [PMID: 37702441 DOI: 10.1002/kjm2.12753] [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: 05/05/2023] [Revised: 07/30/2023] [Accepted: 08/10/2023] [Indexed: 09/14/2023] Open
Abstract
This study aimed to explore the role and mechanism of DYRK1a regulating ferroptosis of cardiomyocytes during myocardial ischemia-reperfusion injury (MIRI). H9c2 cells treated with oxygen-glucose deprivation/reoxygenation (OGD/R) were used as MIRI cell models and transfected with sh-DYRK1a or/and erastin. Cell viability, apoptosis, and DYRK1a mRNA/protein expression were measured accordingly. The levels of reactive oxygen species (ROS), iron, malondialdehyde (MDA), and glutathione (GSH) were determined. The expression of ferroptosis-related proteins (GPX4, SLC7A11, ACSL4, and TFR1) was detected using western blotting. The MIRI rat model was established to explore the possible role of DYRK1a suppression in cell injury and ferroptosis. OGD/R cells showed elevated mRNA and protein expression for DYRK1a. OGD/R cells transfected with sh-DYRK1a showed elevated cell viability, GSH content, increased GPX4 and SLC7A11 expression, suppressed iron content, MDA, ROS, ACSL4, and TFR1 expression, and reduced apoptosis rate, whereas co-transfection of sh-DYRK1a with erastin reversed the attenuation of sh-DYRK1a on MIRI. The suppressive effect of sh-DYRK1a on MI/R injury was confirmed in an MIRI rat model. DYRK1a mediates ferroptosis of cardiomyocytes to deteriorate MIRI progression.
Collapse
Affiliation(s)
- Jing Wang
- Department of Emergency, Beijing Tongren Hospital (South District), Capital Medical University, Beijing, China
| | - Rui-Ming Xu
- Department of Emergency, Beijing Tongren Hospital (South District), Capital Medical University, Beijing, China
| | - Qiu-Mei Cao
- Department of Emergency, Beijing Tongren Hospital (South District), Capital Medical University, Beijing, China
| | - Bing-Chen Ma
- Department of Emergency, Beijing Tongren Hospital (South District), Capital Medical University, Beijing, China
| | - Hao Zhang
- Department of Emergency, Beijing Tongren Hospital (South District), Capital Medical University, Beijing, China
| | - Hua-Peng Hao
- Department of Emergency, Beijing Tongren Hospital (South District), Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Pagliaro P, Penna C. Inhibitors of NLRP3 Inflammasome in Ischemic Heart Disease: Focus on Functional and Redox Aspects. Antioxidants (Basel) 2023; 12:1396. [PMID: 37507935 PMCID: PMC10376505 DOI: 10.3390/antiox12071396] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is caused by several mechanisms, including the production of reactive oxygen species (ROS), altered cellular osmolarity, and inflammatory response. Calcium overload, altered oxygen levels, and mitochondrial ROS are also involved in these MIRI processes, resulting in the irreversible opening of the mitochondrial permeability transition pore (mPTP). These mechanisms and processes are associated with NLRP3 inflammasome priming and activation, which can also induce cell death by pyroptosis through the up-regulation of the caspase-1 pathway and IL-18 release. In addition, endothelial dysfunction, both in the presence and absence of MIRI, is also accompanied by altered oxygen levels, decreased nitric oxide production, and ROS overproduction, resulting in the expression of adhesion molecules and leukocyte infiltration in which the NLRP3 inflammasome plays a central role, thus contributing, through endothelial dysfunction, to the alteration of coronary flow, typical of ischemic heart disease. Given the intricate interrelationship between ROS and NLRP3, ROS inhibitors can reduce NLRP3 inflammasome activation, while NLRP3 inhibitors can reduce oxidative stress and inflammation. NLRP3 inhibitors have been intensively studied as anti-inflammatory agents in basic cardiovascular sciences. In this review, we analyze the interrelation between ROS and NLRP3 in ischemic heart disease and the effects of some NLRP3 inhibitors as possible therapeutic agents in this disease condition. All compounds considered in this review need larger studies to confirm their appropriate use in clinical scenarios as anti-ischemic drugs.
Collapse
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, Turin University, Orbassano, 10043 Turin, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, Turin University, Orbassano, 10043 Turin, Italy
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|