1
|
Yu Z, Wang J, Li T, Gao L. Melatonin promotes diabetic wound healing by mediating mitochondrial function in endothelial cells through the AMPK/SIRT1/HIF-1α pathway. Tissue Cell 2025; 95:102884. [PMID: 40233668 DOI: 10.1016/j.tice.2025.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
OBJECTIVE Diabetic wounds are open lesions that can develop on any part of the body of diabetic patients. Importantly, melatonin (Mel) exerts promotional effects on wound healing. Accordingly, this study explored the mechanism of Mel in diabetic wound healing by mediating mitochondrial function in endothelial cells. METHODS Human umbilical vein vascular endothelial cells (HUVECs) were exposed to high glucose (HG) to mimic a diabetic environment in vitro, followed by Mel treatment. Cell viability, invasion and angiogenic capacity were evaluated with CCK-8, Transwell, and tube formation assays, respectively. CD31 protein expression was determined with Western blot. Wound healing ability was evaluated in vitro, and the levels of adenosine triphosphate (ATP), reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and apoptosis-related proteins (Bcl-2/Bax/CytC) were also detected. To verify the role of the AMPK/SIRT1/HIF-1α pathway in diabetic wound healing, HG-induced HUVECs treated with Mel were subjected to treatment with sh-HIF-1α, AMPK inhibitor (compound c), or SIRT1 inhibitor (Nicotinamide). RESULTS HG impaired the proliferation, invasion, angiogenesis, and wound healing ability of HUVEC, increased ROS, Bax, and CytC levels, and decreased MMP and the levels of ATP and Bcl-2. Mel facilitated viability, angiogenesis, and wound healing ability while ameliorating mitochondrial dysfunction in HG-treated HUVECs. Mel activated the AMPK/SIRT1 pathway to upregulate HIF-1α in HG-treated HUVECs. HIF-1α knockdown, CC, or Nicotinamide negated the effect of Mel on HG-treated HUVECs. CONCLUSIONS Mel fosters angiogenesis and represses mitochondrial dysfunction in endothelial cells by activating the AMPK/SIRT1/HIF-1α pathway, thereby promoting diabetic wound healing.
Collapse
Affiliation(s)
- Zeyang Yu
- Department of Orthopedic, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing 100038, China
| | - Jiangning Wang
- Department of Orthopedic, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing 100038, China
| | - Tianbo Li
- Department of Orthopedic, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing 100038, China
| | - Lei Gao
- Department of Orthopedic, Capital Medical University Affiliated Beijing Shijitan Hospital, No.10 Yangfangdian Tieyi Road, Haidian District, Beijing 100038, China.
| |
Collapse
|
2
|
Tang H, Ning K, Wu B, Wang X, He J, Li P, Pan L, Zhang J, He Y, Bian S, Ma X, Zhang J, Liu C, Qin Z, Hu H. Scutellarein ameliorates pulmonary arterial hypertension via sirtuin 1 mediated deacetylation of nicotinamide nucleotide transhydrogenase. Biochem Pharmacol 2025; 237:116932. [PMID: 40189160 DOI: 10.1016/j.bcp.2025.116932] [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: 02/04/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/13/2025]
Abstract
Scutellarein (Sc), a natural flavonoid, holds potential for treating pulmonary arterial hypertension (PAH), yet its mechanisms remain unexplored. This study investigated Sc's therapeutic effects and underlying pathways in PAH. In vivo experiments demonstrated that Sc significantly attenuated right ventricular hypertension, pulmonary arterial remodeling, αSMA expression, and vascular inflammation in PAH models. In vitro, Sc suppressed hypoxia-induced proliferation, migration, inflammation, and pyroptosis in human pulmonary artery smooth muscle cells (HPASMCs). Mechanistically, Sc activated the SIRT1/NAD+ axis to restore mitochondrial homeostasis: it upregulated SIRT1 expression and elevated NAD+ levels by promoting SIRT1-mediated deacetylation of nicotinamide nucleotide transhydrogenase (NNT), thereby enhancing NNT activity. Elevated NAD+ further activated SIRT1, forming a self-reinforcing SIRT1/NNT/NAD+ feedback loop that mitigated hypoxia-induced mitochondrial dysfunction. This study identifies Sc as a novel regulator of the SIRT1-dependent NNT deacetylation pathway, which stabilizes NAD+ homeostasis to counteract HPASMCs dysregulation in PAH. These findings highlight Sc's potential as a therapeutic candidate for PAH, offering insights into targeting mitochondrial-metabolic pathways for vascular remodeling diseases.
Collapse
Affiliation(s)
- Heng Tang
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ke Ning
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Boji Wu
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Xuhong Wang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Jingyu He
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Pingping Li
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Lina Pan
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jiawen Zhang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Yi He
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Shizhu Bian
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Xingyu Ma
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Jihang Zhang
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Chuan Liu
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Zhexue Qin
- Department of Cardiology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China.
| | - Houyuan Hu
- Department of Cardiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
3
|
Zhang J, Tao J, Zhou Z, Pei W, Xiao Y, Guo Y, Gao J, Jiang C, Dai L, Zhang G, Tan C. Current research on mitochondria‑associated membranes in cardiovascular diseases (Review). Mol Med Rep 2025; 31:141. [PMID: 40183396 PMCID: PMC11976516 DOI: 10.3892/mmr.2025.13506] [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: 10/14/2024] [Accepted: 03/11/2025] [Indexed: 04/05/2025] Open
Abstract
The present study aimed to explore the role of mitochondria‑associated membranes (MAMs) as a key interface between mitochondria and the endoplasmic reticulum (ER) and to evaluate their importance in maintaining the physiological functions of these two organelles. MAMs not only act as a structural bridge between mitochondria and the ER but also widely participate in the regulation of mitochondrial biosynthesis and function, Ca2+ signal transduction, lipid metabolism, oxidative stress response and autophagy. In addition, the specific protein composition of MAMs is increasingly being recognized as having a profound impact on their function, and these proteins play a central role in regulating intercellular communication. Recently, the scientific community has accumulated a large amount of evidence supporting MAMs as potential targets for cardiovascular disease treatment. The present review focuses on the fine structure and multifunctional properties of MAMs and their mechanisms in the occurrence and development of cardiovascular diseases. The goal is to explore the mechanism of MAMs, therapeutic intervention points directly related to cardiovascular diseases, and feasibility of incorporating MAMs into the diagnostic strategy and treatment plan of cardiovascular diseases to provide novel insights and theoretical support for clinical practice in this field. MAMs have great potential as therapeutic targets for various cardiovascular diseases. This finding not only deepens the understanding of the interaction between organelles but also opens up a promising research path for the development of new therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiaheng Zhang
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Jing Tao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Zijuan Zhou
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Wanjuan Pei
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yili Xiao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yanghongxu Guo
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jian Gao
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Chenyv Jiang
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Ling Dai
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Guomin Zhang
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Chao Tan
- First Clinical College of Traditional Chinese Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410208, P.R. China
- The Domestic First-Class Discipline Construction Project of Chinese Medicine of Hunan University of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Inherit Workroom of Medical Master Professor Xiong Ji-bo's Experiences, First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| |
Collapse
|
4
|
Zaky HS, El-Said NT, Aboutaleb AS, Allam A, Mansour M, Ahmed HI, Abdel-Sattar SA. Mito-TEMPO Mitigates Fibromyalgia Induced by Reserpine in Rats: Orchestration Between SIRT1, Mitochondrial Dynamics, Endoplasmic Reticulum and miRNA-320. Neurochem Res 2025; 50:172. [PMID: 40434586 PMCID: PMC12119751 DOI: 10.1007/s11064-025-04424-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/28/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025]
Abstract
Fibromyalgia (FM) is a chronic disorder that lacks both well-defined underlying causes and effective treatments. Mito-TEMPO (MIT) is a mitochondrial-specific antioxidant that has demonstrated benefits in many cancerous, renal, cardiovascular, and neurodegenerative disorders. However, the therapeutic effect of MIT on FM remains ambiguous. The objective of the current work is to illuminate the use of MIT for FM and its prospective mechanisms. Here, we used the FM rat model induced by three days of subcutaneous reserpine injection (1 mg/kg) and examined the role of MIT on SIRT1 activation and other implicated molecular pathways. Behavioral tests showed that MIT (0.7 mg/kg) can effectively alleviate the locomotor, nociceptive, and depressive-like behaviors in reserpinized rats, an effect that simultaneously reconciles the balance of monoamines in the rat brain. Western blot analysis showed that MIT up-regulates SIRT1 and improves the expression of mitochondrial dynamics proteins (DRP1 and OPA1) and the endoplasmic reticulum protein (CHOP). Furthermore, MIT treatment significantly enhanced the SOD and CAT activities and decreased the brain contents of NF-κB, TNF-α, and BAX, but significantly enriching the Bcl-2 content. Lastly, MIT treatment significantly reduced the genetic expression of miRNA-320 following RES treatment. All the measured parameters showed a significant correlation with SIRT1 expression. Our results suggest that MIT provides antioxidant, anti-apoptotic, and anti-inflammatory impacts on the FM rat model, with proposed mechanisms involved activating the SIRT1 pathway to regulate mitochondrial dynamics, endoplasmic reticulum stress, as well as miRNA-320. Thus, MIT has the potential to be an effectual drug candidate for FM treatment.
Collapse
Affiliation(s)
- Heba S Zaky
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Nermin T El-Said
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Amany S Aboutaleb
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Albatoul Allam
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt.
| | - Mona Mansour
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Hebatalla I Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| | - Somaia A Abdel-Sattar
- Pharmacology and Toxicology Department, Faculty of Pharmacy for Girls, Al-Azhar University, Nasr City, Cairo, P.N.11754, Egypt
| |
Collapse
|
5
|
Zhao J, Shu Z, Li X, Zhang W, Sun M, Song W, Cheng H, Shi S. Dehydrodiisoeugenol alleviates palmitate-induced mitochondrial dysfunction in human vascular smooth muscle cells through the activation of SIRT1-mediated Drp1 deacetylation. Lipids Health Dis 2025; 24:187. [PMID: 40413480 DOI: 10.1186/s12944-025-02611-9] [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: 11/18/2024] [Accepted: 05/14/2025] [Indexed: 05/27/2025] Open
Abstract
OBJECTIVE Dehydrodiisoeugenol (Deh) has demonstrated positive effects in the prevention and treatment of cardiovascular disease (CVD) caused by lipid overload, but its specific mechanism of action remains poorly understood. The aim of this study was to investigate the possible mechanisms by which Deh modulates the mitochondrial dysfunction induced by palmitate (PA) in vascular smooth muscle cells (VSMCs). METHODS A PA-induced high-fat model of VSMCs was established, and the effect of PA on the VSMCs on function was detected by evaluating the oxidative stress and apoptosis of cells, as well as mitochondrial function. The expression of dynamin-related protein 1 (Drp1) was detected by immunofluorescence and immunoprecipitation. The key targets of Deh for the treatment of mitochondria-related diseases were screened by bioinformatics analysis and molecular docking techniques. Finally, the role of Silent information regulator 1 (SIRT1) in the treatment of PA-induced mitochondrial dysfunction in VSMCs by Deh was explored by administrating Deh as well as SIRT1 activator (CAY10602, CAY) and SIRT1 inhibitor (JGB1741, JGB). RESULTS The results showed that PA concentration-dependently increased oxidative stress and apoptosis in VSMCs, while modulating the acetylation of Drp1, promoting its expression and mitochondrial ectopia, thereby inducing mitochondrial dysfunction. Bioinformatics analysis and molecular docking indicated that SIRT1 may be a key target of Deh for the treatment of mitochondria-related diseases. Follow-up experiments revealed that Deh significantly inhibited PA-induced mitochondrial dysfunction in VSMCs by suppressing acetylation and expression of Drp1 and reducing mitochondrial ectasia, an effect that was achieved by regulating SIRT1. CONCLUSION Deh was able to inhibit Drp1 expression and mitochondrial ectopia by reducing Drp1 acetylation through activation of SIRT1, thereby inhibiting PA-induced mitochondrial dysfunction effects in VSMCs, ameliorating pathological processes, such as cellular oxidative stress and apoptosis, and maintaining stable cellular functions.
Collapse
Affiliation(s)
- Jianjun Zhao
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
| | - Zhiyun Shu
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130000, China
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Xiangjun Li
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Wenqing Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Mengze Sun
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Wenxiao Song
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Hongyuan Cheng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, 130000, China
| | - Shaomin Shi
- Department of Respiratory Medicine, China-Japan Union Hospital of Jilin University, Changchun, 130000, China.
| |
Collapse
|
6
|
Zhang Y, Cao C, Sun C, Yan J, Wang Y, Gu C. The SIRT1/GPS2/AIP1 axis regulates pulmonary vascular permeability in ventilator-induced lung injury. Mol Cell Biochem 2025:10.1007/s11010-025-05313-z. [PMID: 40397290 DOI: 10.1007/s11010-025-05313-z] [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: 02/17/2025] [Accepted: 05/15/2025] [Indexed: 05/22/2025]
Abstract
Mechanical ventilation (MV) is essential for patients who require life support, but undue mechanical stress leads to airway and alveolar injury, also known as ventilator-induced lung injury (VILI). MV induces changes in pulmonary endothelial barrier integrity by affecting cell junction proteins. The mechanisms of disruption of endothelial barrier integrity during VILI are still unclear. This study aimed to investigate the roles and mechanisms by which ASK1-interacting protein-1 (AIP1), G-protein pathway suppressor 2 (GPS2), and sirtuin 1 (SIRT1) affect VILI. Human lung microvascular endothelial cells (HLMVECs) were transfected with AIP1 small interfering RNA (siRNA), GPS2 siRNA, GPS2 cDNA, and SIRT1 siRNA and subjected to 20% cyclic stretch (CS). C57BL/6N mice were pretreated with the SIRT1 siRNA before MV. We found that CS of 20% activated oxidative stress, increased the reactive oxygen species (ROS) production, and disrupted the pulmonary endothelial cell barrier integrity. AIP1 depletion increased the ROS production and aggravated the disruption of endothelial barrier integrity. Loss of GPS2 decreased the level of AIP1, leading to low expression levels of cell junction proteins. These effects were alleviated by GPS2 overexpression. SIRT1 depletion induced a decrease in GPS2 and AIP1, and increased the ROS production, resulting in decreased expression levels of cell junction proteins. Furthermore, VILI was exacerbated by increased cytokine production (IL-6 and IL-1β), pulmonary oedema, and an elevated wet/dry weight ratio in SIRT1-depleted mice under MV. These results suggest that cyclic mechanical stretching activated oxidative stress and disrupted the expression of cell junction proteins. The SIRT1/GPS2/AIP1 axis influences the production of ROS to regulate the pulmonary endothelial cell barrier integrity during VILI.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Cuicui Cao
- Department of Anesthesiology, Linyi People's Hospital, Linyi, Shandong, China
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Chang Sun
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jie Yan
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yuelan Wang
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Changping Gu
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
7
|
Xu E, Wang J, Ding N, Wang H, Liu C, Wang X, Liu C. Nanoarchitectonics with Zinc-Doped Carbon Dots for Mitochondria-Targeted Repair and Regeneration Signaling Amplification in CLI Therapy. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40377343 DOI: 10.1021/acsami.5c03525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Critical limb ischemia (CLI) faces high rates of amputation and mortality. Despite advancements in surgical and endovascular interventions, their invasiveness and restricted applicability leave many CLI patients classified as "no-option" cases. Therapeutic angiogenesis strategies offer prospects for revascularization, but their efficacy remains suboptimal. Herein, we developed a nanomedicine, mitochondria-targeted zinc-doped ascorbic acid-derived carbon dots (TPP-Zn@ACDs), which simultaneously restores mitochondrial function and amplifies regenerative signaling to synergistically boost angiogenesis in ischemic limbs. TPP-Zn@ACDs integrate potent antioxidative properties of carbon dots and the pro-regenerative effects of Zn2+, with triphenylphosphine (TPP) and polyethylene glycol (PEG) functionalization endowing precise mitochondrial targeting and enhanced biocompatibility, thereby localizing therapeutic effects to the core of oxidative stress mitigation and regenerative signaling transduction. In vitro, TPP-Zn@ACDs improved mitochondrial function by reducing reactive oxygen species, restoring mitochondrial membrane potential and enhancing ATP production, through activation of the SIRT1/PGC-1α signaling pathway. Further, the proliferation, migration, and tube formation activities of endothelial cells were increased, while hypoxia-induced apoptosis and necrosis was inhibited effectively. Notably, leveraging mitochondrial restoration in endothelial cells, TPP-Zn@ACDs subsequently reprogrammed macrophages from an M1 pro-inflammatory phenotype to an M2 anti-inflammatory phenotype. In vivo, TPP-Zn@ACDs demonstrated remarkable therapeutic efficacy in a mouse CLI model, achieving robust blood flow recovery, increased microvascular density, and improved immune microenvironment. Together, this study proposes TPP-Zn@ACDs as a versatile engineering nanomedicine for mitochondria-targeted therapy, providing a scalable approach to breakthrough angiogenic efficacy in ischemic diseases.
Collapse
Affiliation(s)
- Erwei Xu
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Biochemical Engineering, The Affiliated Qingdao Central Hospital of Qingdao University, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Jianyuan Wang
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Biochemical Engineering, The Affiliated Qingdao Central Hospital of Qingdao University, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Ning Ding
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Biochemical Engineering, The Affiliated Qingdao Central Hospital of Qingdao University, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Haoran Wang
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Biochemical Engineering, The Affiliated Qingdao Central Hospital of Qingdao University, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Chunlei Liu
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Biochemical Engineering, The Affiliated Qingdao Central Hospital of Qingdao University, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Xiaoyu Wang
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Biochemical Engineering, The Affiliated Qingdao Central Hospital of Qingdao University, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| | - Chunzhao Liu
- State Key Laboratory of Bio-fibers and Eco-textiles, Institute of Biochemical Engineering, The Affiliated Qingdao Central Hospital of Qingdao University, College of Materials Science and Engineering, Qingdao University, Qingdao 266071, China
| |
Collapse
|
8
|
Xu R, He H, Deng H, Dong Y, Wu X, Xia Z, Zhou Y, Yang L, Huang Z, Xu W, Xu P, Xu H. Study of conductive nerve conduits for anti-inflammatory and antioxidant effects. RSC Adv 2025; 15:14136-14151. [PMID: 40313319 PMCID: PMC12044412 DOI: 10.1039/d5ra00997a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Replacing autologous nerve grafts with nerve conduits is the prevailing direction for the treatment of peripheral nerves, though the repair of hollow nerve conduits remains unsatisfactory. In this study, cysteinylated zein (l-Zein) was prepared through a disulfide exchange reaction between the disulfide bonds of cysteine (Cys) and those of zein (Zein). Subsequently, electrospinning was utilized to fabricate hollow nerve conduits loaded with berberine (BBR) by means of hydrogen bonding and physical encapsulation. Hydrogels were prepared by ionic cross-linking of Zein with pectin (Pec), and were subsequently loaded with melatonin (MT) and graphene oxide (GO) through physical adsorption and encapsulation. A hydrogel was then injected into a hollow catheter to form a hydrogel composite nerve conduit (l-ZBZPGM). The hydrogels exhibited a continuous porous network structure with pore size distribution between 100 and 200 μm. Most of the hydrogels exhibited porosity exceeding 70% and the compressive modulus was 0.42 ± 0.025 MPa. A hydrogel exhibited a residual mass ratio of 35.15% ± 1.87% at the end of the 30 d degradation period, achieved peak release on day 18 with a release rate of 83.31% ± 3.64%, and had an electrical conductivity of 1.23 ± 0.482 × 10-3 S cm-1, meeting the requirements for nerve repair. The lack of cytotoxicity and the anti-inflammatory and antioxidant properties of l-ZBZPGM were demonstrated using RSC96 cells and Raw264.7 cells. Additionally, through electrical stimulation experiments, it was proven that the addition of GO can promote the proliferation of nerve cells. The biological materials used in this study are of simple composition, and their degradation products may have a minimal impact on the microenvironment. The findings suggested that l-ZBZPGM was more conductive to peripheral nerve regeneration.
Collapse
Affiliation(s)
- Runtian Xu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Hanping He
- Department of Radiation Oncology, Hubei Cancer Hospital Wuhan 430079 China
| | - Huan Deng
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Yuehan Dong
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Xiangjie Wu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Zinuo Xia
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Yang Zhou
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Lin Yang
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Zhijun Huang
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Wenjin Xu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Peihu Xu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| | - Haixing Xu
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering, and Life Sciences, Wuhan University of Technology Wuhan 430070 China
| |
Collapse
|
9
|
Wang X, Li Z, Xing Y, Wang Y, Wang S, Wang L, Zhang H. Ameliorating effect of the aldose reductase inhibitor 1-Acetyl-5-phenyl-1 H-pyrrol-3-ylacetate on galactose-induced cataract. Sci Rep 2025; 15:12759. [PMID: 40229517 PMCID: PMC11997150 DOI: 10.1038/s41598-025-98079-9] [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: 01/10/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025] Open
Abstract
Diabetes mellitus, as a common chronic disease, easily leads to significant changes in the structure of the eye, among which diabetic cataract is particularly common. Although surgery is the main treatment for this complication, it may be accompanied by postoperative complications. Therefore, it is particularly important to develop specific drugs for diabetic cataract, aiming to fundamentally reduce its incidence and reduce the need for surgery. At present, the greatest challenge is to develop therapeutic agents with multiple synergistic effects based on the complex pathogenesis of cataract. 1-Acetyl-5-phenyl-1 H-pyrrol-3-ylacetate (APPA) is designed based on the pathological mechanism as a potential drug to alleviate the occurrence of diabetic cataract. Our observations suggest that APPA is more effective than bendazaclysine in alleviating high galactose-induced oxidative stress (The malondialdehyde content in the APPA group and bendazaclysine group was significantly reduced to 0.45-fold and 0.58-fold compared to the high galactose-induced group, respectively.) and apoptosis (The apoptosis rate in the APPA group and bendazaclysine group was significantly reduced to 0.28-fold and 0.35-fold compared to the high galactose-induced group, respectively.) in lens epithelial cells by increasing antioxidant enzyme activity, and restoring mitochondrial homeostasis. Mechanistic studies have shown that APPA restoration of mitochondrial homeostasis is mediated through the SIRT1-PGC-1α pathway. In the galactose-induced cataract rat model, APPA is effective in alleviating the occurrence of galactose-induced cataract. In conclusion, APPA with multiple synergistic functions may be a potential drug to alleviate the occurrence of diabetic cataract, and it has a wider range of indications than benzydalysine.
Collapse
Affiliation(s)
- Xi Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Zhuoya Li
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Ying Xing
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Yaru Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China
| | - Shiyao Wang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Liping Wang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130012, Jilin, China
| | - Hui Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, 130041, Jilin, China.
| |
Collapse
|
10
|
Cui X, Spanos M, Zhao C, Wan W, Cui C, Wang L, Xiao J. Mitochondrial Dysfunction in HFpEF: Potential Interventions Through Exercise. J Cardiovasc Transl Res 2025; 18:442-456. [PMID: 39863753 DOI: 10.1007/s12265-025-10591-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
HFpEF is a prevalent and complex type of heart failure. The concurrent presence of conditions such as obesity, hypertension, hyperglycemia, and hyperlipidemia significantly increase the risk of developing HFpEF. Mitochondria, often referred to as the powerhouses of the cell, are crucial in maintaining cellular functions, including ATP production, intracellular Ca2+ regulation, reactive oxygen species generation and clearance, and the regulation of apoptosis. Exercise plays a vital role in preserving mitochondrial homeostasis, thereby protecting the cardiovascular system from acute stress, and is a fundamental component in maintaining cardiovascular health. In this study, we review the mitochondrial dysfunction underlying the development and progression of HFpEF. Given the pivotal role of exercise in modulating cardiovascular diseases, we particularly focus on exercise as a potential therapeutic strategy for improving mitochondrial function. Graphical abstract Note: This picture was created with BioRender.com.
Collapse
Affiliation(s)
- Xinxin Cui
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Albert Einstein College of Medicine, Department of Internal Medicine, NCB, Bronx, NY, USA
| | - Cuimei Zhao
- Department of Cardiology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Wensi Wan
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Caiyue Cui
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China
| | - Lijun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China.
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, 226011, China.
- Institute of Cardiovascular Sciences, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai Engineering Research Center of Organ Repair, Shanghai University, Shanghai, China.
| |
Collapse
|
11
|
Peng X, Zhao L, Wang J, Zhang Y, Liu Z, Wang K, Zhang L. Melatonin Alleviates Oxidative Stress-Induced Mitochondrial Dysfunction Through Ameliorating NAD + Homeostasis of hDPSCs for Cell-Based Therapy. J Pineal Res 2025; 77:e70058. [PMID: 40391773 DOI: 10.1111/jpi.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 03/29/2025] [Accepted: 04/30/2025] [Indexed: 05/22/2025]
Abstract
Human dental pulp stem cells (hDPSCs) exhibit amazing therapeutic abilities in a variety of diseases due to their remarkable self-renewal capacity and multi-differentiation potential. However, their therapeutic potential could be weakened by various factors such as oxidative stress in cell survival microenvironment In Vivo. Here, we explored the protective effect and mechanism of melatonin (Mel) on hDPSCs transplanted in a type 1 diabetes mellitus (T1DM) rat model. Nicotinamide adenine dinucleotide (NAD+) metabolism and mitochondrial function were remarkably impaired in T1DM rats caused by oxidative stress, while the combination of Mel and post-hDPSCs transplantation could rebalance NAD+ homeostasis through regulating NAMPT-NAD+-SIRT1 axis. Furthermore, Mel significantly reduced intracellular and mitochondrial reactive oxygen species, and alleviated cell senescence and apoptosis of hDPSCs exposed to hydrogen peroxide through ameliorating NAD+ depletion and mitochondrial dysfunction. The protective role of Mel could be extremely essential to stem cells in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Xiu Peng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Li Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Jiale Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yinmo Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Zihan Liu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Kun Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
12
|
Li P, Fan Z, Huang Y, Luo L, Wu X. Mitochondrial dynamics at the intersection of macrophage polarization and metabolism. Front Immunol 2025; 16:1520814. [PMID: 40196123 PMCID: PMC11973336 DOI: 10.3389/fimmu.2025.1520814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Macrophages are vital sentinels in innate immunity, and their functions cannot be performed without internal metabolic reprogramming. Mitochondrial dynamics, especially mitochondrial fusion and fission, contributes to the maintenance of mitochondrial homeostasis. The link between mitochondrial dynamics and macrophages in the past has focused on the immune function of macrophages. We innovatively summarize and propose a link between mitochondrial dynamics and macrophage metabolism. Among them, fusion-related FAM73b, MTCH2, SLP-2 (Stomatin-like protein 2), and mtSIRT, and fission-related Fis1 and MTP18 may be the link between mitochondrial dynamics and macrophage metabolism association. Furthermore, post-translational modifications (PTMs) of mtSIRT play prominent roles in mitochondrial dynamics-macrophage metabolism connection, such as deacetylates and hypersuccinylation. MicroRNAs such as miR-150, miR-15b, and miR-125b are also possible entry points. The metabolic reprogramming of macrophages through the regulation of mitochondrial dynamics helps improve their adaptability and resistance to adverse environments and provides therapeutic possibilities for various diseases.
Collapse
Affiliation(s)
- Pan Li
- Department of Environment and Safety Engineering, Taiyuan Institute of Technology, Taiyuan, China
| | - Zhengbo Fan
- People’s Government of Huangshui Town, Shizhu Tujia Autonomous County, Chongqing, China
| | - Yanlan Huang
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Liang Luo
- College of Veterinary Medicine, Southwest University, Chongqing, China
| | - Xiaoyan Wu
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| |
Collapse
|
13
|
Li Q, Xiao N, Zhang H, Liang G, Lin Y, Qian Z, Yang X, Yang J, Fu Y, Zhang C, Liu A. Systemic aging and aging-related diseases. FASEB J 2025; 39:e70430. [PMID: 40022602 DOI: 10.1096/fj.202402479rrr] [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: 11/05/2024] [Revised: 02/07/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Aging is a biological process along with systemic and multiple organ dysfunction. It is more and more recognized that aging is a systemic disease instead of a single-organ functional disorder. Systemic aging plays a profound role in multiple diseases including neurodegenerative diseases, cardiovascular diseases, and malignant diseases. Aged organs communicate with other organs and accelerate aging. Skeletal muscle, heart, bone marrow, skin, and liver communicate with each other through organ-organ crosstalk. The crosstalk can be mediated by metabolites including lipids, glucose, short-chain fatty acids (SCFA), inflammatory cytokines, and exosomes. Metabolic disorders including hyperglycemia, hyperinsulinemia, and hypercholesterolemia caused by chronic diseases accelerate hallmarks of aging. Systemic aging leads to the destruction of systemic hemostasis, causes the release of inflammatory cytokines, senescence-associated secretory phenotype (SASP), and the imbalance of microbiota composition. Released inflammatory factors further aggregate senescence, which promotes the aging of multiple solid organs. Targeting senescence or delaying aging is emerging as a critical health strategy for solving age-related diseases, especially in the old population. In the current review, we will delineate the mechanisms of organ crosstalk in systemic aging and age-related diseases to provide therapeutic targets for delaying aging.
Collapse
Affiliation(s)
- Qiao Li
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Nanyin Xiao
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Heng Zhang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Guangyu Liang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yan Lin
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Zonghao Qian
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Xiao Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jiankun Yang
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Yanguang Fu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Cuntai Zhang
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anding Liu
- Experimental Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| |
Collapse
|
14
|
Ghavamikia N, Mehrnoosh F, Zare F, Ali-Khiavi P, Sinehsepehr A, Boushehri YG, Vahedinezhad M, Abdollahi E, Hjazi A, Aminnezhad S, Saffarfar H, Hamzehzadeh S, Nourizadeh M, KarkonShayan S. Mitochondrial Quality Control and Melatonin: A Strategy Against Myocardial Injury. J Biochem Mol Toxicol 2025; 39:e70194. [PMID: 40009050 DOI: 10.1002/jbt.70194] [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: 12/31/2024] [Revised: 02/03/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025]
Abstract
Melatonin exhibits various biological functions, including regulation of circadian and endocrine rhythms, anti-inflammatory, and antioxidant effects. Aging and damaged mitochondria are major sources of oxidative stress (OS), and mitochondrial quality control (MQC) is crucial for maintaining normal mitochondrial function. Myocardial ischemia-reperfusion injury is a major complication that can arise during reperfusion therapy for coronary heart disease. However, effective intervention strategies are currently lacking. Mitochondrial dysfunction and OS are considered central mechanisms of myocardial reperfusion injury, with mitochondrial-targeted interventions being a potential treatment direction. Recent studies have shown that melatonin improves mitochondrial structure and function through multiple pathways. This review discusses the mechanisms by which melatonin ameliorates myocardial ischemia-reperfusion injury, focusing on MQC, and explores its potential applications in the prevention and treatment of myocardial ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Nima Ghavamikia
- Cardiovascular Research Institute, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Faranak Mehrnoosh
- Department of Food Science and Technology, Faculty of Agriculture and Natural Resources, Urmia University, Urmia, Iran
| | - Farshad Zare
- Student Research Committee, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Payam Ali-Khiavi
- Student Research Committee, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Sinehsepehr
- School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Milad Vahedinezhad
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Abdollahi
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Siamak Aminnezhad
- Cardiovascular Research Center, Tehran, Iran
- Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Saffarfar
- Cardiovascular Research Center, Tehran, Iran
- Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Hamzehzadeh
- Student Research Committee, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehrdad Nourizadeh
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh KarkonShayan
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
15
|
Yan X, Quan S, Guo R, Li Z, Bai M, Wang B, Su P, Xu E, Li Y. Calycosin‑7‑O‑β‑D‑glucoside downregulates mitophagy by mitigating mitochondrial fission to protect HT22 cells from oxygen‑glucose deprivation/reperfusion‑induced injury. Mol Med Rep 2025; 31:71. [PMID: 39820475 PMCID: PMC11751592 DOI: 10.3892/mmr.2025.13436] [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: 10/04/2024] [Accepted: 11/26/2024] [Indexed: 01/19/2025] Open
Abstract
Calycosin‑7‑O‑β‑D‑glucoside (CG), a major active ingredient of Astragali Radix, exerts neuroprotective effects against cerebral ischemia; however, whether the effects of CG are associated with mitochondrial protection remains unclear. The present study explored the role of CG in improving mitochondrial function in a HT22 cell model of oxygen‑glucose deprivation/reperfusion (OGD/R). The Cell Counting Kit‑8 assay, flow cytometry, immunofluorescence and western blotting were performed to investigate the effects of CG on mitochondrial function. The results demonstrated that mitochondrial function was restored after treatment with CG, as indicated by reduced mitochondrial reactive oxygen species levels, increased mitochondrial membrane potential and improved mitochondrial morphology. Overactivated mitophagy was revealed to be inhibited by the regulation of proteins involved in fission [phosphorylated‑dynamin‑related protein 1 (Drp1) and Drp1] and mitophagy (LC3, p62 and translocase of outer mitochondrial membrane 20), and mitochondrial biogenesis was demonstrated to be enhanced by increased levels of sirtuin 1 (SIRT1) and peroxisome proliferator‑activated receptor γ coactivator‑1α (PGC‑1α). In addition, neuronal apoptosis was ameliorated by CG, as determined by a decreased rate of apoptosis, and levels of caspase‑3 and Bcl‑2/Bax. In conclusion, the present study demonstrated that CG may alleviate OGD/R‑induced injury by upregulating SIRT1 and PGC‑1α protein expression, and reducing excessive mitochondrial fission and overactivation of mitophagy.
Collapse
Affiliation(s)
- Xiangli Yan
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Siqi Quan
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- College of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Roujia Guo
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- College of Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Zibo Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Ming Bai
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Baoying Wang
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Pan Su
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Erping Xu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| | - Yucheng Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, P.R. China
| |
Collapse
|
16
|
Wei X, Guo H, Huang G, Luo H, Gong L, Meng P, Liu J, Zhang W, Mei Z. SIRT1 Alleviates Mitochondrial Fission and Necroptosis in Cerebral Ischemia/Reperfusion Injury via SIRT1-RIP1 Signaling Pathway. MedComm (Beijing) 2025; 6:e70118. [PMID: 40008377 PMCID: PMC11850763 DOI: 10.1002/mco2.70118] [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: 05/04/2024] [Revised: 12/20/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Programmed cell death, including necroptosis, plays a critical role in the pathogenesis of cerebral ischemia/reperfusion injury (CIRI). Silent information regulator 1 (SIRT1) has been identified as a potential therapeutic target for CIRI, yet its precise role in regulating necroptosis remains controversial. Furthermore, the potential interaction between SIRT1 and receptor-interacting protein kinase 1 (RIP1) in this context is not fully understood. Sanpian Decoction (SPD), a classical traditional herbal formula, was previously shown to enhance SIRT1 expression in our studies. Our findings demonstrated that, both in vivo and in vitro, CIRI was associated with a decrease in SIRT1 levels and phosphorylated dynamin-related protein 1 (p-DRP1) at Ser637, alongside an increase in RIP1 and other necroptosis-related proteins. Co-immunoprecipitation and immunofluorescence analyses revealed a weakened interaction between SIRT1 and RIP1. Furthermore, abnormal mitochondrial fission and dysfunction were mediated through the phosphoglycerate mutase 5-DRP1 pathway. Notably, SPD treatment improved neurological outcomes and reversed these pathological changes by enhancing the SIRT1-RIP1 interaction. In conclusion, this study suggests that SIRT1 is a promising therapeutic target for CIRI, capable of inhibiting necroptosis and mitigating mitochondrial fission via the SIRT1-RIP1 pathway. SPD exhibits therapeutic potential by activating SIRT1, thereby attenuating necroptosis and mitochondrial fission during CIRI.
Collapse
Affiliation(s)
- Xuan Wei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Hanjing Guo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Guangshan Huang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Haoyue Luo
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Lipeng Gong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Pan Meng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
| | - Jiyong Liu
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine DiagnosticsHunan University of Chinese MedicineChangshaHunanChina
| | - Wenli Zhang
- School of PharmacyHunan University of Chinese MedicineChangshaHunanChina
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral DiseasesCollege of Integrated Traditional Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunanChina
- Third‐Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese MedicineCollege of Medicine and Health SciencesChina Three Gorges UniversityYichangHubeiChina
| |
Collapse
|
17
|
Luo L, Wu Q, Xiao Q, Chen Y, Deng Z, Cen C, Lin J. Lipotoxicity-induced upregulation of FIS1 exacerbates mitochondrial fragmentation and promotes NLRP3-dependent pyroptosis in diabetic cardiomyopathy. Free Radic Biol Med 2025; 228:183-196. [PMID: 39734056 DOI: 10.1016/j.freeradbiomed.2024.12.049] [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: 09/17/2024] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Lipotoxicity is a significant factor in the pathogenesis of diabetic cardiomyopathy (DbCM), a condition characterized by mitochondrial fragmentation and pyroptosis. Mitochondrial fission protein 1 (FIS1) plays a role in mitochondrial fission by anchoring dynamin-related protein 1 (DRP1). However, the specific contribution of FIS1 to DbCM remains unclear. This study aims to clarify how lipotoxicity-induced upregulation of FIS1 affects mitochondrial fragmentation and the mechanisms linking this fragmentation to NLRP3-dependent pyroptosis in DbCM. METHODS To model lipotoxicity in DbCM, we used db/db mice and primary neonatal rat cardiomyocytes (NRCMs) treated with palmitic acid (PA) and conducted a series of in vivo and in vitro experiments. Gain- and loss-of-function studies on NRCMs were performed using pharmacological inhibitors and small interfering RNA (siRNA) transfection, and we assessed the expression and function of FIS1, mitochondrial dynamics, mitochondrial reactive oxygen species (mitoROS) production, NLRP3-dependent pyroptosis, and their interrelationships. RESULTS Our results show that in the myocardium of db/db mice, NLRP3-dependent pyroptosis is associated with upregulation of FIS1, mitochondrial fragmentation, and increased oxidative stress. In NRCMs subjected to PA, the application of VX-765 and MCC950 to inhibit caspase-1 and NLRP3, respectively, significantly reduced pyroptosis. Additionally, pretreatment with Mito-TEMPO (MT) demonstrated that mitoROS are critical initiators for NLRP3 inflammasome activation and subsequent pyroptosis. Furthermore, PA-induced upregulation of FIS1 exacerbates mitochondrial fragmentation. Downregulation of FIS1 or inhibition of FIS1/DRP1 interaction reversed mitochondrial fragmentation, reduced mitoROS levels, and mitigated pyroptosis. CONCLUSIONS Lipotoxicity-induced FIS1 upregulation exacerbates mitochondrial fragmentation through its interaction with DRP1, leading to increased mitoROS production and the initiation of NLRP3-dependent pyroptosis in DbCM. Therefore, targeting FIS1 emerges as a potential therapeutic approach for managing DbCM.
Collapse
Affiliation(s)
- Libo Luo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Qingrui Wu
- State Key Laboratory of Respiratory Disease, Department of Pulmonary Diseases, Guangzhou Chest Hospital, Guangzhou Medical University, Guangzhou, 510095, China.
| | - Qingyu Xiao
- Department of Anesthesiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, 515041, China.
| | - Yuqiong Chen
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Zhanxiang Deng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Chunren Cen
- Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Jijin Lin
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| |
Collapse
|
18
|
Yaghoobi A, Rezaee M, Hedayati N, Keshavarzmotamed A, Khalilzad MA, Russel R, Asemi Z, Rajabi Moghadam H, Mafi A. Insight into the cardioprotective effects of melatonin: shining a spotlight on intercellular Sirt signaling communication. Mol Cell Biochem 2025; 480:799-823. [PMID: 38980593 DOI: 10.1007/s11010-024-05002-3] [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: 11/30/2023] [Accepted: 03/25/2024] [Indexed: 07/10/2024]
Abstract
Cardiovascular diseases (CVDs) are the leading causes of death and illness worldwide. While there have been advancements in the treatment of CVDs using medication and medical procedures, these conventional methods have limited effectiveness in halting the progression of heart diseases to complete heart failure. However, in recent years, the hormone melatonin has shown promise as a protective agent for the heart. Melatonin, which is secreted by the pineal gland and regulates our sleep-wake cycle, plays a role in various biological processes including oxidative stress, mitochondrial function, and cell death. The Sirtuin (Sirt) family of proteins has gained attention for their involvement in many cellular functions related to heart health. It has been well established that melatonin activates the Sirt signaling pathways, leading to several beneficial effects on the heart. These include preserving mitochondrial function, reducing oxidative stress, decreasing inflammation, preventing cell death, and regulating autophagy in cardiac cells. Therefore, melatonin could play crucial roles in ameliorating various cardiovascular pathologies, such as sepsis, drug toxicity-induced myocardial injury, myocardial ischemia-reperfusion injury, hypertension, heart failure, and diabetic cardiomyopathy. These effects may be partly attributed to the modulation of different Sirt family members by melatonin. This review summarizes the existing body of literature highlighting the cardioprotective effects of melatonin, specifically the ones including modulation of Sirt signaling pathways. Also, we discuss the potential use of melatonin-Sirt interactions as a forthcoming therapeutic target for managing and preventing CVDs.
Collapse
Affiliation(s)
- Alireza Yaghoobi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Malihe Rezaee
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Hedayati
- School of Medicine, Iran University of Medical Science, Tehran, Iran
| | | | | | - Reitel Russel
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA.
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hasan Rajabi Moghadam
- Department of Cardiology, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
- Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
19
|
Le HT, Yu J, Ahn HS, Kim MJ, Chae IG, Cho HN, Kim J, Park HK, Kwon HN, Chae HJ, Kang BH, Seo JK, Kim K, Back SH. eIF2α phosphorylation-ATF4 axis-mediated transcriptional reprogramming mitigates mitochondrial impairment during ER stress. Mol Cells 2025; 48:100176. [PMID: 39756584 PMCID: PMC11786836 DOI: 10.1016/j.mocell.2024.100176] [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: 10/03/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025] Open
Abstract
Eukaryotic translation initiation factor 2α (eIF2α) phosphorylation, which regulates all 3 unfolded protein response pathways, helps maintain cellular homeostasis and overcome endoplasmic reticulum (ER) stress through transcriptional and translational reprogramming. However, transcriptional regulation of mitochondrial homeostasis by eIF2α phosphorylation during ER stress is not fully understood. Here, we report that the eIF2α phosphorylation-activating transcription factor 4 (ATF4) axis is required for the expression of multiple transcription factors, including nuclear factor erythroid 2-related factor 2 and its target genes responsible for mitochondrial homeostasis during ER stress. eIF2α phosphorylation-deficient (A/A) cells displayed dysregulated mitochondrial dynamics and mitochondrial DNA replication, decreased expression of oxidative phosphorylation complex proteins, and impaired mitochondrial functions during ER stress. ATF4 overexpression suppressed impairment of mitochondrial homeostasis in A/A cells during ER stress by promoting the expression of downstream transcription factors and their target genes. Our findings underscore the importance of the eIF2α phosphorylation-ATF4 axis for maintaining mitochondrial homeostasis through transcriptional reprogramming during ER stress.
Collapse
Affiliation(s)
- Hien Thi Le
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Jiyoung Yu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Hee Sung Ahn
- AMC Sciences, Asan Medical Center, Seoul 05505, Korea
| | - Mi-Jeong Kim
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - In Gyeong Chae
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Hyun-Nam Cho
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Juhee Kim
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Hye-Kyung Park
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Hyuk Nam Kwon
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea
| | - Han-Jung Chae
- School of Pharmacy, Jeonbuk National University, Jeonju 54896, Korea
| | - Byoung Heon Kang
- Department of Biological Sciences, School of Life Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Jeong Kon Seo
- Central Research Facilities (UCRF), Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea.
| | - Kyunggon Kim
- Department of Digital Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea.
| | - Sung Hoon Back
- Basic-Clinical Convergence Research Center, School of Biological Sciences, University of Ulsan, Ulsan 44610, Korea.
| |
Collapse
|
20
|
Zhang C, Chang X, Zhao D, He Y, Dong G, Gao L. Mitochondria and myocardial ischemia/reperfusion injury: Effects of Chinese herbal medicine and the underlying mechanisms. J Pharm Anal 2025; 15:101051. [PMID: 39931135 PMCID: PMC11808734 DOI: 10.1016/j.jpha.2024.101051] [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: 02/08/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 02/03/2025] Open
Abstract
Ischemic heart disease (IHD) is associated with high morbidity and mortality rates. Reperfusion therapy is the best treatment option for this condition. However, reperfusion can aggravate myocardial damage through a phenomenon known as myocardial ischemia/reperfusion (I/R) injury, which has recently gained the attention of researchers. Several studies have shown that Chinese herbal medicines and their natural monomeric components exert therapeutic effects against I/R injury. This review outlines the current knowledge on the pathological mechanisms through which mitochondria participate in I/R injury, focusing on the issues related to energy metabolism, mitochondrial quality control disorders, oxidative stress, and calcium. The mechanisms by which mitochondria mediate cell death have also been discussed. To develop a resource for the prevention and management of clinical myocardial I/R damage, we compiled the most recent research on the effects of Chinese herbal remedies and their monomer components.
Collapse
Affiliation(s)
- Chuxin Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xing Chang
- Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, 100053, China
| | - Dandan Zhao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu He
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Guangtong Dong
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lin Gao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
21
|
Rui Y, Guo Y, He L, Wang ME, Wu H. SIRT1/PGC-1α-mediated mitophagy participates the improvement roles of BMAL1 in podocytes injury in diabetic nephropathy: evidences from in vitro experiments. Eur J Med Res 2025; 30:29. [PMID: 39810231 PMCID: PMC11734468 DOI: 10.1186/s40001-025-02280-5] [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: 09/19/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Dysfunction in podocyte mitophagy has been identified as a contributing factor to the onset and progression of diabetic nephropathy (DN), and BMAL1 plays an important role in the regulation of mitophagy. Thus, this study intended to examine the impact of BMAL1 on podocyte mitophagy in DN and elucidate its underlying mechanisms. MATERIALS AND METHODS High D-glucose (HG)-treated MPC5 cells was used as a podocyte injury model for investigating the potential roles of BMAL1 in DN. Mitophagy was examined by detecting autophagosomes using transmission electron microscopy, and detecting the colocalization of LC3 and Tom20 using immunofluorescence staining. The interaction between BMAL1 and SIRT1 was conducted by immunoprecipitation (Co-IP) assay. RESULTS In HG-induced podocyte injury model, we found that BMAL1 and SIRT1 mRNA level was significantly decreased, and positively correlated with mitophagy dysfunction. BMAL1 overexpression could ameliorate HG-induced podocyte injury, evidenced by improved cell viability, decreased cell apoptosis and inflammatory cytokines expression (TNF-α, IL-1β, and IL-6). BMAL1 overexpression could promote podocyte mitophagy coupled with increased expression of mitophagy markers PINK1 and Parkin. In terms of mechanism, Co-IP suggested that BMAL1 could interact with SIRT1. SIRT1 inhibitor Ex-527 addition obviously inhibit the effect of BMAL1 overexpression on the mitophagy, demonstrating that BMAL1 may act on mitophagy by SIRT1//PGC-1α axis. CONCLUSIONS Our in vitro experiments demonstrate that BMAL1/SIRT1/PGC-1α pathway may protect podocytes against HG-induced DN through promoting mitophagy.
Collapse
Affiliation(s)
- Yanxia Rui
- Department of Nephrology, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), No.1882, Zhonghuan North Road, Jiaxing, 314000, Zhejiang, China
| | - Yinfeng Guo
- Department of Nephrology, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), No.1882, Zhonghuan North Road, Jiaxing, 314000, Zhejiang, China
| | - Linying He
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Min-Er Wang
- Jiaxing University Master Degree Cultivation Base, Zhejiang Chinese Medical University, Hangzhou, 310000, Zhejiang, China
| | - Henglan Wu
- Department of Nephrology, Affiliated Hospital of Jiaxing University (The First Hospital of Jiaxing), No.1882, Zhonghuan North Road, Jiaxing, 314000, Zhejiang, China.
- Kidney Disease Center College of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China.
| |
Collapse
|
22
|
Li J, Hao G, Yan Y, Li M, Li G, Lu Z, Sun Z, Chen Y, Liu H, Zhao Y, Wu M, Bao X, Wang Y, Li Y. Hydrogen restores central tryptophan and metabolite levels and maintains mitochondrial homeostasis to protect rats from chronic mild unpredictable stress damage. Neurochem Int 2025; 182:105914. [PMID: 39653185 DOI: 10.1016/j.neuint.2024.105914] [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: 10/12/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND AND PURPOSE The field of hydrogen medicine has garnered extensive attention since Professor Ohsawa established that low concentrations of hydrogen (2%-4%) exert antioxidant effects. The present study aimed to evaluate the therapeutic effect of molecular hydrogen in a CUMS rat model. METHODS A total of 40 SD rats were randomly divided into a control group, a model group, a hydrogen group, and a positive drug group. Four weeks post-modeling, hydrogen inhalation and other treatments were administered. Behavioral, biochemical, and immunohistochemical evaluations were performed after treatment. RESULTS Hydrogen inhalation alleviated depressive behavior and hippocampal neuronal damage in CUMS rats, as well as restored the levels of neurotransmitters, inflammatory factors, and oxidative stress. Moreover, it maintained mitochondrial homeostasis and up-regulated the expression of PGC-1α, PINK1, and Parkin. CONCLUSIONS The results collectively indicated that hydrogen significantly attenuated CUMS-induced depressive-like behavior and monoamine neurotransmitter deficiency, as well as protected the brain from oxidative stress and inflammatory damage and effectively preserved mitochondrial homeostasis.
Collapse
Affiliation(s)
- Jiaxin Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Gaimei Hao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yupeng Yan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ming Li
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Gaifen Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhengmin Lu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zhibo Sun
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanjing Chen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Haixia Liu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yukun Zhao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Meng Wu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiangxin Bao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yubo Li
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
23
|
Fu Y, Yang L, Liu L, Kong L, Sun H, Sun Y, Yin F, Yan G, Wang X. Rhein: An Updated Review Concerning Its Biological Activity, Pharmacokinetics, Structure Optimization, and Future Pharmaceutical Applications. Pharmaceuticals (Basel) 2024; 17:1665. [PMID: 39770507 PMCID: PMC11679290 DOI: 10.3390/ph17121665] [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: 11/20/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Rhein is a natural active ingredient in traditional Chinese medicine that has attracted much attention due to its wide range of pharmacological activities. However, its clinical application is limited by low water solubility, poor oral absorption, and potential toxicity to the liver and kidneys. Recently, advanced extraction and synthesis techniques have made it possible to develop derivatives of rhein, which have better pharmacological properties and lower toxicity. This article comprehensively summarizes the biological activity and action mechanism of rhein. Notably, we found that TGF-β1 is the target of rhein improving tissue fibrosis, while NF-κB is the main target of its anti-inflammatory effect. Additionally, we reviewed the current research status of the pharmacokinetics, toxicology, structural optimization, and potential drug applications of rhein and found that the coupling and combination therapy of rhein and other active ingredients exhibit a synergistic effect, significantly enhancing therapeutic efficacy. Finally, we emphasize the necessity of further studying rhein's pharmacological mechanisms, toxicology, and development of analogs, aiming to lay the foundation for its widespread clinical application as a natural product and elucidate its prospects in modern medicine.
Collapse
Affiliation(s)
- Yuqi Fu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China; (Y.F.); (L.L.); (L.K.); (F.Y.); (G.Y.)
| | - Le Yang
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou 510006, China; (L.Y.); (Y.S.)
| | - Lei Liu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China; (Y.F.); (L.L.); (L.K.); (F.Y.); (G.Y.)
| | - Ling Kong
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China; (Y.F.); (L.L.); (L.K.); (F.Y.); (G.Y.)
| | - Hui Sun
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China; (Y.F.); (L.L.); (L.K.); (F.Y.); (G.Y.)
| | - Ye Sun
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou 510006, China; (L.Y.); (Y.S.)
| | - Fengting Yin
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China; (Y.F.); (L.L.); (L.K.); (F.Y.); (G.Y.)
| | - Guangli Yan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China; (Y.F.); (L.L.); (L.K.); (F.Y.); (G.Y.)
| | - Xijun Wang
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, National Chinmedomics Research Center, National TCM Key Laboratory of Serum Pharmacochemistry, Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Heping Road 24, Harbin 150040, China; (Y.F.); (L.L.); (L.K.); (F.Y.); (G.Y.)
- State Key Laboratory of Dampness Syndrome, The Second Affiliated Hospital Guangzhou University of Chinese Medicine, Dade Road 111, Guangzhou 510006, China; (L.Y.); (Y.S.)
| |
Collapse
|
24
|
Rahmani S, Roohbakhsh A, Pourbarkhordar V, Karimi G. The Cardiovascular Protective Function of Natural Compounds Through AMPK/SIRT1/PGC-1α Signaling Pathway. Food Sci Nutr 2024; 12:9998-10009. [PMID: 39723061 PMCID: PMC11666815 DOI: 10.1002/fsn3.4553] [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: 08/27/2024] [Revised: 10/02/2024] [Accepted: 10/06/2024] [Indexed: 12/28/2024] Open
Abstract
Cardiovascular disease (CVD) poses a major risk to human health and exert a heavy burden on individuals, society, and healthcare systems. Therefore, it is critical to identify CVD's underlying mechanism(s) and target them using effective agents. Natural compounds have shown promise as antioxidants with cardioprotective functions against CVD injuries due to their antioxidative solid capacity and high safety profile. Several CVDs, such as heart failure, ischemia/reperfusion, atherosclerosis, and cardiomyopathies, are closely linked to mitochondrial dysfunction. It is well established that activating the AMPK/SIRT1/PGC-1α pathway during CVD promotes mitochondrial function. Therefore, targeting the AMPK/SIRT1/PGC-1α pathway provides a foundation for novel therapeutic strategies to combat CVD. A key goal of our search was to find natural compounds that target this biological pathway and have beneficial effects on CVD.
Collapse
Affiliation(s)
- Sohrab Rahmani
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
- Pharmaceutical Research Center, Institute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
| | - Vahid Pourbarkhordar
- Student Research CommitteeMashhad University of Medical SciencesMashhadIran
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of PharmacyMashhad University of Medical SciencesMashhadIran
- Pharmaceutical Research Center, Institute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
25
|
Hui Z, Lai-Fa W, Xue-Qin W, Ling D, Bin-Sheng H, Li JM. Mechanisms and therapeutic potential of chinonin in nervous system diseases. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024; 26:1405-1420. [PMID: 38975978 DOI: 10.1080/10286020.2024.2371040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 07/09/2024]
Abstract
The flavonoid compound chinonin is one of the main active components of Rhizoma anemarrhena with multiple activities, including anti-inflammatory and antioxidant properties, protection of mitochondrial function and regulation of immunity. In this paper, we reviewed recent research progress on the protective effect of chinonin on brain injury in neurological diseases. "Chinonin" OR "Mangiferin" AND "Nervous system diseases" OR "Neuroprotection" was used as the terms for search in PumMed. After discarding duplicated and irrelevant articles, a total of 23 articles relevant to chinonin published between 2012 and 2023 were identified in our study.
Collapse
Affiliation(s)
- Zhang Hui
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha 410219, China
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha 410219, China
| | - Wang Lai-Fa
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha 410219, China
| | - Wang Xue-Qin
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha 410219, China
| | - Deng Ling
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha 410219, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha 410219, China
| | - He Bin-Sheng
- Hunan Provincial Key Laboratory of the TCM Agricultural Biogenomics, Changsha Medical University, Changsha 410219, China
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha 410219, China
| | - Jian-Ming Li
- Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Neurodegenerative Diseases, Changsha Medical University, Changsha 410219, China
- The Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha 410219, China
| |
Collapse
|
26
|
Zheng J, Zhao L, Zhang Y, He W, Guo X, Wang J. Melatonin alleviates high glucose-induced cardiomyocyte injury through suppressing mitochondrial FUNDC1-DRP1 axis. J Pharm Pharmacol 2024; 76:1431-1448. [PMID: 39306802 DOI: 10.1093/jpp/rgae114] [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: 03/06/2024] [Accepted: 08/21/2024] [Indexed: 11/05/2024]
Abstract
OBJECTIVES To use H9c2 cardiomyocytes to establish a diabetic cardiomyopathic model by exposing these cells to high glucose (HG), followed by treating them with melatonin (MEL) or plasmid vectors overexpressing FUN14 Domain Containing 1 (FUNDC1). METHODS We employed quantitative real-time PCR, mitochondrial staining, and biochemical assays to measure the activity of various antioxidant and mitochondrial complex functions under various treatment conditions. KEY FINDINGS Our results showed that HG induced the expression of FUNDC1 and increased mitochondrial oxidative stress and fragmentation, while MEL treatment reversed most of these pathological effects. Moreover, HG exposure activated dynamin-related protein 1 expression and its translocation to mitochondria. Modulation of AMP-activated protein kinase level was found to be another pathological hallmark. In silico molecular docking, analysis revealed that MEL could directly bind the catalytic groove of FUNDC1 through Van der Waal's force and hydrogen bonding. Finally, MEL ameliorated diabetic cardiomyopathy-induced mitochondrial injury through FUNDC1 in vivo. CONCLUSIONS Hyperglycemia induced mitochondrial fragmentation and altered electron transport chain complex functions, which could be ameliorated by MEL treatment, suggesting its potential as a cardiovascular therapeutic.
Collapse
Affiliation(s)
- Junyi Zheng
- Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, China
- Department of Cardiology, Chest Hospital, Tianjin University, Tianjin 300222, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin 300222, China
- Tianjin Institute of Cardiovascular Disease, Tianjin 300222, China
| | - Lili Zhao
- Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, China
- Department of Cardiology, Chest Hospital, Tianjin University, Tianjin 300222, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin 300222, China
- Tianjin Institute of Cardiovascular Disease, Tianjin 300222, China
| | - Yingying Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, China
- Department of Cardiology, Chest Hospital, Tianjin University, Tianjin 300222, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin 300222, China
- Tianjin Institute of Cardiovascular Disease, Tianjin 300222, China
| | - Wenbin He
- Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, China
- Department of Cardiology, Chest Hospital, Tianjin University, Tianjin 300222, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin 300222, China
- Tianjin Institute of Cardiovascular Disease, Tianjin 300222, China
| | - Xukun Guo
- Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, China
- Department of Cardiology, Chest Hospital, Tianjin University, Tianjin 300222, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin 300222, China
- Tianjin Institute of Cardiovascular Disease, Tianjin 300222, China
| | - Jixiang Wang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, China
- Department of Cardiology, Chest Hospital, Tianjin University, Tianjin 300222, China
- Clinical School of Thoracic, Tianjin Medical University, Tianjin 300222, China
- Tianjin Institute of Cardiovascular Disease, Tianjin 300222, China
| |
Collapse
|
27
|
Liu S, Yang TN, Wang YX, Ma XY, Shi YS, Zhao Y, Li JL. Parkin-TLR4-NLRP3 Axis Directs Melatonin to Alleviate Atrazine-Induced Immune Impairment in Splenic Macrophages. J Pineal Res 2024; 76:e70014. [PMID: 39648693 DOI: 10.1111/jpi.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 09/10/2024] [Accepted: 11/20/2024] [Indexed: 12/10/2024]
Abstract
Atrazine (ATR) is a widespread environmental herbicide that seriously affects agricultural work and human safety. Melatonin (MLT) as an endogenous neuroendocrine hormone is widely found in animals and plants, which have antioxidant and anti-inflammatory effects. Pink1/Parkin-mediated mitophagy keeps normal physiological processes by degrading damaged mitochondria in cells. Therefore, we investigated the potential role and mechanism of MLT in ATR-induced toxic injury of the spleen. The results showed that MLT alleviated ATR-induced unclear boundary between the white pulp and the red pulp of the spleen. It is also shown that ATR resulted in swollen mitochondria, partial extinction of mitochondrial membranes and cristae, and increased mitophagy under the action of MLT. ATR-induced reactive oxygen species (ROS) activates the Pink1/Parkin pathway, which guides mitophagy development and then causes the activation of TLR4/NF-κB inflammatory pathway. Meanwhile, these damages further exacerbated the production of NLRP3 inflammasomes, leading to spleen necrosis. Interestingly, these changes were improved after MLT treatment. Collectively, we found that MLT alleviates ATR-induced immune impairment in splenic macrophages via regulating Parkin-TLR4-NLRP3 axis which elucidates the effect of melatonin on the spleen and provides a novel perspective on melatonin in splenic inflammatory injury treatment.
Collapse
Affiliation(s)
- Shuo Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Tian-Ning Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yu-Xiang Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiang-Yu Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yu-Sheng Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yi Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
28
|
Huo JY, Hou C, Li XL, Yang L, Jiang WY. Renal denervation ameliorates atrial remodeling in type 2 diabetic rats by regulating mitochondrial dynamics. J Physiol Biochem 2024; 80:935-948. [PMID: 39436584 DOI: 10.1007/s13105-024-01054-7] [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: 05/14/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024]
Abstract
There is no effective treatment for diabetes-related atrial remodeling currently. This study aimed to investigate the effects of renal denervation (RDN) on diabetes-related atrial remodeling and explore the related mechanisms. A type 2 diabetes mellitus model was established by high-fat diet feeding and low-dose streptozotocin injection in Sprague‒Dawley rats. After successful modeling, the diabetic rats were randomly assigned to two groups according to whether they were subjected to RDN or sham RDN surgery. At the end of the experiment, cardiac function and structure were evaluated by echocardiography and histology, respectively. Mitochondrial morphology, function and mitochondrial dynamics were assessed by multiple methods. Mdivi1 was used to verify the mechanism by which RDN improves atrial remodeling. In the 10th week, diabetic rats exhibited obvious atrial remodeling, including atrial enlargement and diastolic dysfunction. Pathological staining showed that diabetic rats had cardiomyocyte hypertrophy and interstitial fibrosis in atrial tissues. In terms of mitochondrial morphology and function, diabetic rats exhibited fragmented mitochondria, reduced adenosine triphosphate production and decreased mitochondrial membrane potential levels. Abnormal mitochondrial dynamics in diabetic rats were characterized by the inhibition of mitochondrial fusion, excessive mitochondrial fission, and the suppression of mitophagy. However, RDN effectively ameliorated diabetes-induced pathological atrial remodeling. In addition, RDN significantly improved mitochondrial morphological and functional abnormalities and corrected the disorders of mitochondrial dynamics. Furthermore, the protective effects of RDN against atrial remodeling were related to the regulation of mitochondrial dynamics. RDN prevented diabetes-induced atrial remodeling. These protective effects might be related to improvements in mitochondrial dynamics.
Collapse
Affiliation(s)
- Jun-Yu Huo
- Department of Cardiology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Can Hou
- Department of Cardiology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiao-Long Li
- Department of Cardiology, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Ling Yang
- Department of Cardiology, The First People's Hospital of Changzhou, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wan-Ying Jiang
- Department of Cardiology, Changzhou Hospital of Traditional Chinese Medicine, Changzhou, China.
| |
Collapse
|
29
|
Zhu J, Jin P, Zhou T, Zhang D, Wang Z, Tang Z, Liu Z, Ren G. SIRT1 modulates microglia phenotypes via inhibiting drp1 phosphorylation reduces neuroinflammation in heatstroke. Brain Res Bull 2024; 218:111101. [PMID: 39396713 DOI: 10.1016/j.brainresbull.2024.111101] [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: 08/24/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Brain injury often results in high mortality rates and significant sequelae following severe heatstroke (HS). Neuroinflammation aggravates HS-induced brain injury, yet the involvement of microglia in heat-induced neuroinflammation deserves further investigation. METHODS Our study investigated activation status, phenotype markers, production of pro-inflammatory cytokine and reactive oxygen species (ROS) of microglia both in vitro and in vivo under HS. Utilizing high-throughput sequencing, we identified SIRT1 as a potential modulator of microglia phenotype, and observed that SIRT1 alleviated severe heatstroke-induced brain injury following intraperitoneal administration of the SIRT1 agonist SRT-1720 and the inhibitor selisistat. Additionally, the effects of SRT-1720 and selisistat on mitochondrial dynamics and microglial phenotype transition were examined in BV2 cells in vitro. RESULTS Heatstroke promotes microglia activation, as evidenced by the increased production of pro-inflammatory cytokine and reactive oxygen species. High-throughput sequencing revealed elevated expression of SIRT1 in BV2 cells under HS. Upon inhibition of SIRT1 expression, there was a corresponding increase in pro-inflammatory cytokine, iNOS, and ROS expression in BV2 cells. In vivo experiments with the SIRT1 agonist SRT-1720 showed a mitigation of neuron injury under HS, as assessed by Nissl and HE staining. Activation of SIRT1 was associated with a reduction in mitochondrial injury and a decrease in the phosphorylation of mitochondrial fission protein Drp1ser616. Furthermore, the heat-induced activation of microglia was reversed by the Drp1 inhibitor, Mdivi. CONCLUSIONS Our findings provided evidence that SIRT1 played a crucial role in inhibiting heat stress-induced microglial activation. By suppressing the phosphorylation of mitochondrial fission protein Drp1, SIRT1 contributed to the reduction of neuroinflammation and severity of heatstroke-induced brain injury.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Pediatric, Daping Hospital, Army Medical University, China; Department of Pediatric, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China.
| | - Panshi Jin
- Department of Plastic Surgery, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - Tingting Zhou
- Department of Pediatric, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - Dingshun Zhang
- Department of Medicine Intensive Care Unit, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - Zixin Wang
- Department of Metabolic Surgery, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510010, China
| | - Zhen Tang
- Department of Pediatric, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China
| | - Zhifeng Liu
- Department of Medicine Intensive Care Unit, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China; Southern Medical University, Guangzhou 510010, China.
| | - Guangli Ren
- Department of Pediatric, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, China; Southern Medical University, Guangzhou 510010, China.
| |
Collapse
|
30
|
Zhao L, Gu C, Zhang Y, Yan J, Qiu L, Qin X, Wang Y. Regulation mechanism of GPS2 on PGC-1α/Drp1-mediated mitochondrial dynamics in inflammation of acute lung injury. Int Immunopharmacol 2024; 140:112838. [PMID: 39116501 DOI: 10.1016/j.intimp.2024.112838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/04/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024]
Abstract
Acute lung injury (ALI) has been a hot topic in the field of critical care research in recent years. Mitochondrial dynamics consists of mitochondrial fusion and mitochondrial fission. Dynamin-related protein 1 (Drp1), a key molecule that regulates mitochondrial fission, is important in the oxidative stress and inflammatory response to ALI. Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is a core protein that mediates mitochondrial biogenesis. G-protein pathway suppressor 2 (GPS2) acts as a transcriptional cofactor with regulatory effects on nuclear-encoded mitochondrial genes. This study aimed to investigate the mechanism of PGC-1α/Drp1-mediated mitochondrial dynamics involved in ALI and to demonstrate the protective mechanism of GPS2 in regulating mitochondrial structure and function and inflammation in ALI. The ALI model was constructed using LPS-induced wild-type mice and human pulmonary microvascular endothelial cells (HPMVECs). It was found that lung injury, oxidative stress and inflammation were exacerbated in the mice ALI model and that mitochondrial structure and function were disrupted in HPMVECs. In vitro studies revealed that LPS led to the upregulated expression of Drp1 and the downregulated expression of PGC-1α and GPS2. Mitochondrial division was reduced and respiratory function was restored in Drp1 knockdown cells, which inhibited oxidative stress and inflammatory response. In addition, the overexpression of PGC-1α and GPS2 significantly inhibited the expression of Drp1, mitochondrial function was restored, and inhibited reactive oxygen species (ROS) production and inflammatory factor release. Moreover, the overexpression of GPS2 promoted the upregulated expression of PGC-1α. This mechanism was also validated in vivo, in which the low expression of GPS2 in mice resulted in the upregulated expression of Drp1 and the downregulated expression of PGC-1α, and further exacerbated LPS-induced ALI. In the present study, we also found that LPS-induced the downregulated expression of GPS2 may be associated with its increased degradation by the proteasome. Therefore, these findings revealed that GPS2 inhibited oxidative stress and inflammation by modulating PGC-1α/Drp1-mediated mitochondrial dynamics to alleviate LPS-induced ALI, which may provide a new approach to the therapeutic orientation for LPS-induced ALI.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Changping Gu
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yi Zhang
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Jie Yan
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Lei Qiu
- School of Anesthesiology, Weifang Medical University, Weifang 261053, China
| | - Xiaofeng Qin
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Yuelan Wang
- Department of Anesthesiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| |
Collapse
|
31
|
Zhang S, Feng X, Yang G, Tan H, Cheng X, Tang Q, Yang H, Zhao Y, Ding X, Li S, Dou X, Li J, Kang H, Li X, Ji Y, Hou Q, An Q, Fang H, Fan H. Dexmedetomidine ameliorates acute kidney injury by regulating mitochondrial dynamics via the α2-AR/SIRT1/PGC-1α pathway activation in rats. Mol Med 2024; 30:184. [PMID: 39455916 PMCID: PMC11505563 DOI: 10.1186/s10020-024-00964-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: 08/22/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Sepsis-associated acute kidney injury (AKI) is a serious complication of systemic infection with high morbidity and mortality in patients. However, no effective drugs are available for AKI treatment. Dexmedetomidine (DEX) is an alpha 2 adrenal receptor agonist with antioxidant and anti-apoptotic effects. This study aimed to investigate the therapeutic effects of DEX on sepsis-associated AKI and to elucidate the role of mitochondrial dynamics during this process. METHODS A lipopolysaccharide (LPS)-induced AKI rat model and an NRK-52E cell model were used in the study. This study investigated the effects of DEX on sepsis-associated AKI and the molecular mechanisms using histologic assessment, biochemical analyses, ultrastructural observation, western blotting, immunofluorescence, immunohistochemistry, qRT-PCR, flow cytometry, and si-mRNA transfection. RESULTS In rats, the results showed that administration of DEX protected kidney structure and function from LPS-induced septic AKI. In addition, we found that DEX upregulated the α2-AR/SIRT1/PGC-1α pathway, protected mitochondrial structure and function, and decreased oxidative stress and apoptosis compared to the LPS group. In NRK-52E cells, DEX regulated the mitochondrial dynamic balance by preventing intracellular Ca2+ overloading and activating CaMKII. CONCLUSIONS DEX ameliorated septic AKI by reducing oxidative stress and apoptosis in addition to modulating mitochondrial dynamics via upregulation of the α2-AR/SIRT1/PGC-1α pathway. This is a confirmatory study about DEX pre-treatment to ameliorate septic AKI. Our research reveals a novel mechanistic molecular pathway by which DEX provides nephroprotection.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xiujing Feng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, China
| | - Guiyan Yang
- College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Haoyang Tan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xin Cheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qichao Tang
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin, China
| | - Haotian Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xuanpan Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Siyao Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xinyi Dou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Junfeng Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Huijie Kang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Xingxing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yaxin Ji
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qingdian Hou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Qiuyue An
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Hao Fang
- College of Optoelectronic Engineering, Chongqing University, Chongqing, China
| | - Honggang Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
32
|
Su X, Li Q, Yang M, Zhang W, Liu X, Ba Y, Deng Q, Zhang Y, Han L, Huang H. Resveratrol protects against a high-fat diet-induced neuroinflammation by suppressing mitochondrial fission via targeting SIRT1/PGC-1α. Exp Neurol 2024; 380:114899. [PMID: 39059737 DOI: 10.1016/j.expneurol.2024.114899] [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: 01/10/2024] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Various health issues have emerged due to consuming high-fat diets (HFD), particularly the detrimental impact they have on mitochondrial dynamics and subsequet cognition functions. Specially, mitochondrial fission can serve as an upstream signal in the regulation of cortical inflammation and neural pyroptosis. Our study was designed to verify the existence of neuroinflammation in the pathogenesis of HFD-induced cognitive dysfunction and demonstrated that resveratrol (RSV) attenuated neural deficits via regulation of cortical mitochondrial fission. A total of 50 male Sprague Dawley rats were randomly divided into five groups: control (Cont, 26 weeks on normal rodent diet); high-fat diet (HFD); dietary adjustments (HFD + ND); resveratrol intervention (HFD + R); joint intervention (HFD + ND + R) for 26 weeks. The spatial learning and memory function, spine density, NLRP3 inflammasome associated protein, mRNA and protein expression involved in mitochondrial dynamics and SIRT1/PGC-1α signaling pathway in brain were measured. Furthermore, reactive oxygen species (ROS) accumulation and resultant mitochondrial membrane potential (MMP) alteration in PC12 cells exposed to palmitic acid (PA) or Drp1 inhibitor (Mdivi-1) were detected to reflect mitochondrial function. The findings suggested that prolonged treatment of RSV improved cognitive deficits and neuronal damage induced by HFD, potentially attributed to activation of the SIRT1/PGC-1α axis. We further indicated that the activation of the NLRP3 inflammasome in PA (200 μM) treated PC12 cells could be inhibited by Mdivi-1. More importantly, Mdivi-1 (10 μM) reduced intracellular ROS levels and enhanced MMP by reversing Drp1-mediated aberrant mitochondrial fission. To summarize, those results clearly indicated that a HFD inhibited the SIRT1/PGC-1α pathway, which contributed to an imbalance in mitochondrial dynamics and the onset of NLRP3-mediated pyroptosis. This effect was mitigated by the RSV possibly through triggering the SIRT1/PGC-1α axis, prevented aberrant mitochondrial fission and thus inhibited the activation of the NLRP3 inflammatory pathway.
Collapse
Affiliation(s)
- Xiao Su
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Qiong Li
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Mingzhi Yang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Wenhui Zhang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Xiaoxue Liu
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Yue Ba
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Qihong Deng
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China
| | - Yu Zhang
- State Key Laboratory of Microbial Technology, Qingdao, Shandong 266000, China; Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong 266000, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250100, China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong 266000, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250100, China
| | - Hui Huang
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China; Environment and Health Innovation Team, College of Public Health, Zhengzhou University, Zhengzhou, Henan province 450001, China.
| |
Collapse
|
33
|
Marino Y, Inferrera F, D'Amico R, Impellizzeri D, Cordaro M, Siracusa R, Gugliandolo E, Fusco R, Cuzzocrea S, Di Paola R. Role of mitochondrial dysfunction and biogenesis in fibromyalgia syndrome: Molecular mechanism in central nervous system. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167301. [PMID: 38878832 DOI: 10.1016/j.bbadis.2024.167301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/22/2024] [Accepted: 06/07/2024] [Indexed: 08/18/2024]
Abstract
A critical role for mitochondrial dysfunction has been shown in the pathogenesis of fibromyalgia. It is a chronic pain syndrome characterized by neuroinflammation and impaired oxidative balance in the central nervous system. Boswellia serrata (BS), a natural polyphenol, is a well-known able to influence the mitochondrial metabolism. The objective of this study was to evaluate the mitochondrial dysfunction and biogenesis in fibromyalgia and their modulation by BS. To induce the model reserpine (1 mg/Kg) was subcutaneously administered for three consecutive days and BS (100 mg/Kg) was given orally for twenty-one days. BS reduced pain like behaviors in reserpine-injected rats and the astrocytes activation in the dorsal horn of the spinal cord and prefrontal cortex that are recognized as key regions associated with the neuropathic pain. Vulnerability to neuroinflammation and impaired neuronal plasticity have been described as consequences of mitochondrial dysfunction. BS administration increased PGC-1α expression in the nucleus of spinal cord and brain tissues, promoting the expression of regulatory genes for mitochondrial biogenesis (NRF-1, Tfam and UCP2) and cellular antioxidant defence mechanisms (catalase, SOD2 and Prdx 3). According with these data BS reduced lipid peroxidation and the GSSG/GSH ratio and increased SOD activity in the same tissues. Our results also showed that BS administration mitigates cytochrome-c leakage by promoting mitochondrial function and supported the movement of PGC-1α protein into the nucleus restoring the quality control of mitochondria. Additionally, BS reduced Drp1 and Fis1, preventing both mitochondrial fission and cell death, and increased the expression of Mfn2 protein, facilitating mitochondrial fusion. Overall, our results showed important mitochondrial dysfunction in central nervous system in fibromyalgia syndrome and the role of BS in restoring mitochondrial dynamics.
Collapse
Affiliation(s)
- Ylenia Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Francesca Inferrera
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Ramona D'Amico
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Marika Cordaro
- Department of Biomedical, Dental and Morphological and Functional Imaging, University of Messina, 98125 Messina, Italy.
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Enrico Gugliandolo
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy.
| | - Roberta Fusco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy.
| | - Rosanna Di Paola
- Department of Veterinary Science, University of Messina, 98168 Messina, Italy.
| |
Collapse
|
34
|
Liu J, Liu X, Guo L, Liu X, Gao Q, Wang E, Dong Z. PPARγ agonist alleviates calcium oxalate nephrolithiasis by regulating mitochondrial dynamics in renal tubular epithelial cell. PLoS One 2024; 19:e0310947. [PMID: 39325731 PMCID: PMC11426502 DOI: 10.1371/journal.pone.0310947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Kidney stone formation is a common disease that causes a significant threat to human health. The crystallization mechanism of calcium oxalate, the most common type of kidney stone, has been extensively researched, yet the damaging effects and mechanisms of calcium oxalate crystals on renal tubular epithelial cells remain incompletely elucidated. Regulated mitochondrial dynamics is essential for eukaryotic cells, but its role in the occurrence and progression of calcium oxalate (CaOx) nephrolithiasis is not yet understood. METHODS An animal model of calcium oxalate-related nephrolithiasis was established in adult male Sprague‒Dawley (SD) rats by continuously administering drinking water containing 1% ethylene glycol for 28 days. The impact of calcium oxalate crystals on mitochondrial dynamics and apoptosis in renal tubular epithelial cells was investigated using HK2 cells in vitro. Blood samples and bilateral kidney tissues were collected for histopathological evaluation and processed for tissue injury, inflammation, fibrosis, oxidative stress detection, and mitochondrial dynamics parameter analysis. RESULTS Calcium oxalate crystals caused higher levels of mitochondrial fission and apoptosis in renal tubular epithelial cells both in vivo and in vitro. Administration of a PPARγ agonist significantly alleviated mitochondrial fission and apoptosis in renal tubular epithelial cells, and improved renal function, accompanied by reduced levels of oxidative stress, increased antioxidant enzyme expression, alleviation of inflammation, and reduced fibrosis in vivo. CONCLUSION Our results indicated that increased mitochondrial fission in renal tubular epithelial cells is a critical component of kidney injury caused by calcium oxalate stones, leading to the accumulation of reactive oxygen species within the tissue and the subsequent initiation of apoptosis. Regulating mitochondrial dynamics represents a promising approach for calcium oxalate nephrolithiasis.
Collapse
Affiliation(s)
- Junfa Liu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xingyang Liu
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Lizhe Guo
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Xiongfei Liu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qian Gao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - E Wang
- Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, China
| | - Zhitao Dong
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
35
|
Zhao M, Li J, Li Z, Yang D, Wang D, Sun Z, Wen P, Gou F, Dai Y, Ji Y, Li W, Zhao D, Yang L. SIRT1 Regulates Mitochondrial Damage in N2a Cells Treated with the Prion Protein Fragment 106-126 via PGC-1α-TFAM-Mediated Mitochondrial Biogenesis. Int J Mol Sci 2024; 25:9707. [PMID: 39273653 PMCID: PMC11395710 DOI: 10.3390/ijms25179707] [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: 07/21/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Mitochondrial damage is an early and key marker of neuronal damage in prion diseases. As a process involved in mitochondrial quality control, mitochondrial biogenesis regulates mitochondrial homeostasis in neurons and promotes neuron health by increasing the number of effective mitochondria in the cytoplasm. Sirtuin 1 (SIRT1) is a NAD+-dependent deacetylase that regulates neuronal mitochondrial biogenesis and quality control in neurodegenerative diseases via deacetylation of a variety of substrates. In a cellular model of prion diseases, we found that both SIRT1 protein levels and deacetylase activity decreased, and SIRT1 overexpression and activation significantly ameliorated mitochondrial morphological damage and dysfunction caused by the neurotoxic peptide PrP106-126. Moreover, we found that mitochondrial biogenesis was impaired, and SIRT1 overexpression and activation alleviated PrP106-126-induced impairment of mitochondrial biogenesis in N2a cells. Further studies in PrP106-126-treated N2a cells revealed that SIRT1 regulates mitochondrial biogenesis through the PGC-1α-TFAM pathway. Finally, we showed that resveratrol resolved PrP106-126-induced mitochondrial dysfunction and cell apoptosis by promoting mitochondrial biogenesis through activation of the SIRT1-dependent PGC-1α/TFAM signaling pathway in N2a cells. Taken together, our findings further describe SIRT1 regulation of mitochondrial biogenesis and improve our understanding of mitochondria-related pathogenesis in prion diseases. Our findings support further investigation of SIRT1 as a potential target for therapeutic intervention of prion diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Lifeng Yang
- National Key Laboratory of Veterinary Public Health and Safety, Key Laboratory of Animal Epidemiology of the Ministry of Agriculture and Rural Affairs, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; (M.Z.)
| |
Collapse
|
36
|
Song X, Fan C, Wei C, Yu W, Tang J, Ma F, Chen Y, Wu B. Mitochondria fission accentuates oxidative stress in hyperglycemia-induced H9c2 cardiomyoblasts in vitro by regulating fatty acid oxidation. Cell Biol Int 2024; 48:1378-1391. [PMID: 38922770 DOI: 10.1002/cbin.12204] [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: 12/17/2023] [Revised: 04/14/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
Oxidative stress plays a pivotal role in the development of diabetic cardiomyopathy (DCM). Previous studies have revealed that inhibition of mitochondrial fission suppressed oxidative stress and alleviated mitochondrial dysfunction and cardiac dysfunction in diabetic mice. However, no research has confirmed whether mitochondria fission accentuates hyperglycemia-induced cardiomyoblast oxidative stress through regulating fatty acid oxidation (FAO). We used H9c2 cardiomyoblasts exposed to high glucose (HG) 33 mM to simulate DCM in vitro. Excessive mitochondrial fission, poor cell viability, and lipid accumulation were observed in hyperglycemia-induced H9c2 cardiomyoblasts. Also, the cells were led to oxidative stress injury, lower adenosine triphosphate (ATP) levels, and apoptosis. Dynamin-related protein 1 (Drp1) short interfering RNA (siRNA) decreased targeted marker expression, inhibited mitochondrial fragmentation and lipid accumulation, suppressed oxidative stress, reduced cardiomyoblast apoptosis, and improved cell viability and ATP levels in HG-exposed H9c2 cardiomyoblasts, but not in carnitine palmitoyltransferase 1 (CPT1) inhibitor etomoxir treatment cells. We also found subcellular localization of CPT1 on the mitochondrial membrane, FAO, and levels of nicotinamide adenine dinucleotide phosphate (NADPH) were suppressed after exposure to HG treatment, whereas Drp1 siRNA normalized mitochondrial CPT1, FAO, and NADPH. However, the blockade of FAO with etomoxir abolished the above effects of Drp1 siRNA in hyperglycemia-induced H9c2 cardiomyoblasts. The preservation of mitochondrial function through the Drp1/CPT1/FAO pathway is the potential mechanism of inhibited mitochondria fission in attenuating oxidative stress injury of hyperglycemia-induced H9c2 cardiomyoblasts.
Collapse
Affiliation(s)
- Xiaogang Song
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu, China
- Department of Cardiology, Xi'an Central Hospital, Xi'an, Shaanxi, China
- Department of Cardiology, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Chongxi Fan
- Department of Gastroenterology, Air Force Medical Center, Beijing, China
| | - Chao Wei
- Department of Neurology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wuhan Yu
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Jichao Tang
- Key Laboratory of Stem Cells and Gene Drugs of Gansu Province, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu, China
| | - Feng Ma
- Department of Cardiology, Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Yongqing Chen
- Department of Cardiology, Gansu Provincial Central Hospital, Lanzhou, Gansu, China
| | - Bing Wu
- Department of Geriatrics, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, Gansu, China
| |
Collapse
|
37
|
Rahmani S, Roohbakhsh A, Pourbarkhordar V, Hayes AW, Karimi G. Melatonin regulates mitochondrial dynamics and mitophagy: Cardiovascular protection. J Cell Mol Med 2024; 28:e70074. [PMID: 39333694 PMCID: PMC11436317 DOI: 10.1111/jcmm.70074] [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: 02/26/2024] [Revised: 07/27/2024] [Accepted: 08/28/2024] [Indexed: 09/29/2024] Open
Abstract
Despite extensive progress in the knowledge and understanding of cardiovascular diseases and significant advances in pharmacological treatments and procedural interventions, cardiovascular diseases (CVD) remain the leading cause of death globally. Mitochondrial dynamics refers to the repetitive cycle of fission and fusion of the mitochondrial network. Fission and fusion balance regulate mitochondrial shape and influence physiology, quality and homeostasis. Mitophagy is a process that eliminates aberrant mitochondria. Melatonin (Mel) is a pineal-synthesized hormone with a range of pharmacological properties. Numerous nonclinical trials have demonstrated that Mel provides cardioprotection against ischemia/reperfusion, cardiomyopathies, atherosclerosis and cardiotoxicity. Recently, interest has grown in how mitochondrial dynamics contribute to melatonin cardioprotective effects. This review assesses the literature on the protective effects of Mel against CVD via the regulation of mitochondrial dynamics and mitophagy in both in-vivo and in-vitro studies. The signalling pathways underlying its cardioprotective effects were reviewed. Mel modulated mitochondrial dynamics and mitophagy proteins by upregulation of mitofusin, inhibition of DRP1 and regulation of mitophagy-related proteins. The evidence supports a significant role of Mel in mitochondrial dynamics and mitophagy quality control in CVD.
Collapse
Affiliation(s)
- Sohrab Rahmani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Roohbakhsh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Vahid Pourbarkhordar
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
38
|
Fan JJ, Ding WD, Liang YF, Wei YX, Huang Y, Ma L, Wang R. Diosgenin derivative ML5 attenuates MPTP-induced neuronal impairment via regulating AMPK/PGC-1α-mediated mitochondrial biogenesis and fusion/fission. Am J Transl Res 2024; 16:3582-3598. [PMID: 39262707 PMCID: PMC11384354 DOI: 10.62347/jbre5043] [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: 04/07/2024] [Accepted: 05/14/2024] [Indexed: 09/13/2024]
Abstract
OBJECTIVE The aim of the present study was to assess the therapeutic impact of diosgenin derivative ML5 on Parkinson's disease (PD) and explore the mechanism underlying mitochondrial biogenesis and fusion/fission. METHODS We established 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse models and N-methyl-4-phenylpyridinium iodide (MPP+)-induced cell models of PD. The pole test and forced swimming test were used to detect the motor coordination and depressive symptoms in mice. The influence of ML5 on dopaminergic neuronal injury was investigated. Meanwhile, adenosine triphosphate (ATP) content, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS) production were measured to evaluate mitochondrial function. Confocal and transmission electron microscopy were used to detect mitochondrial morphology of cell. The expression of mitochondrial biogenesis-related proteins was measured by Western blotting. RESULTS The administration of ML5 showed the neuroprotection against MPTP-induced damage in vivo and in vitro. The levels of ATP, MMP, and ROS were restored after ML5 administration. In addition, we observed that ML5 preserved the mitochondrial network morphology and inhibited mitochondrial fission. Furthermore, the amelioration of mitochondrial dysfunction was mediated by enhancing 5'-monophosphate-activated protein kinase (AMPK) and peroxisome proliferators-activated receptor γ coactivator-l alpha (PGC-1α) expression, which activated its downstream modulators leading to the enhancing of mitochondrial biogenesis and the balance of mitochondrial fusion/fission. CONCLUSION ML5 can protect the PD models against MPTP/MPP+-induced mitochondrial dysfunction and neuronal injury via promoting AMPK/PGC-1α signaling activation and be used as a therapeutic drug for PD treatment.
Collapse
Affiliation(s)
- Jing-Jing Fan
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237, China
| | - Wei-Dong Ding
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237, China
| | - Ying-Fan Liang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237, China
| | - Yao-Xin Wei
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237, China
| | - Yi Huang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237, China
| | - Lei Ma
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237, China
| | - Rui Wang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology Shanghai 200237, China
| |
Collapse
|
39
|
Jiang L, Bai K, Wang T. Bacillus subtilis fmbj ameliorates lipopolysaccharide-induced intestinal dysfunction in broilers by enhancing the SIRT1/PGC1α pathway. Poult Sci 2024; 103:103964. [PMID: 38936217 PMCID: PMC11259727 DOI: 10.1016/j.psj.2024.103964] [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: 03/02/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/29/2024] Open
Abstract
This study aimed to explore the impact of dietary Bacillus subtilis fmbj (BS) supplementation on acute intestinal dysfunction induced by lipopolysaccharide (LPS) in broilers. One hundred and eighty 1-day-old male Arbor Acres broilers were randomly divided into three treatment groups, each comprising ten replicates of 6 birds. On d 20, LPS-challenged (LPS group and LPS-BS group) and LPS-unchallenged (CON group) broilers received intraperitoneal injections of 1 mg/kg body weight LPS solution and an equivalent volume of sterile saline, respectively. Compared to the CON group, LPS disrupted (P < 0.05) the morphology of the small intestine (jejunum or ileum), exacerbated (P < 0.05) serum, small intestinal, and small intestinal mitochondrial antioxidant capacity, induced (P < 0.05) small intestinal oxidative damage, and altered (P < 0.05) the expression of genes and proteins related to antioxidants, cell adhesion, and mitochondrial function in the jejunum. The LPS-BS group exhibited a tendency towards improvement in small intestinal morphology, serum, small intestinal, and small intestinal mitochondrial antioxidant capacity, small intestinal oxidative damage, and the expression of genes and proteins related to antioxidants, cell adhesion, and mitochondrial function in the jejunum when compared to the LPS group. In conclusion, BS supplementation may confer protection against LPS-induced acute intestinal dysfunction in broilers by enhancing the activation of SIRT1/PGC1α, suggesting its potential as a valuable additive for the poultry industry.
Collapse
Affiliation(s)
- Luyi Jiang
- College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, 310023, China; Institute of Dairy Science, Ministry of Education Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310023, China
| | - Kaiwen Bai
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, 310023, China; College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
40
|
Zhang J, Zhao Y, Gong N. Endoplasmic reticulum stress signaling modulates ischemia/reperfusion injury in the aged heart by regulating mitochondrial maintenance. Mol Med 2024; 30:107. [PMID: 39044180 PMCID: PMC11265325 DOI: 10.1186/s10020-024-00869-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: 11/02/2023] [Accepted: 06/27/2024] [Indexed: 07/25/2024] Open
Abstract
Aging is associated with an increased risk of myocardial ischemia/reperfusion injury (IRI). With an increasing prevalence of cardiovascular diseases such as coronary arteriosclerosis in older people, there has been increasing interest in understanding the mechanisms of myocardial IRI to develop therapeutics that can attenuate its damaging effects. Previous studies identified that abnormal mitochondria, involved in cellar senescence and oxidative stress, are the master subcellular organelle that induces IRI. In addition, endoplasmic reticulum (ER) stress is also associated with IRI. Cellular adaptation to ER stress is achieved by the activation of ER molecular chaperones and folding enzymes, which provide an important link between ER stress and oxidative stress gene programs. In this review, we outline how these ER stress-related molecules affect myocardial IRI via the crosstalk of ER stress and mitochondrial homeostasis and discuss how these may offer promising novel therapeutic targets and strategies against age-related cardiovascular diseases.
Collapse
Affiliation(s)
- Ji Zhang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, Wuhan, Hubei, 430030, P.R. China
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Institute of Urology & Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei, Anhui, 230022, P.R. China
| | - Yuanyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, Wuhan, Hubei, 430030, P.R. China
| | - Nianqiao Gong
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation of Ministry of Education, National Health Commission and Chinese Academy of Medical Sciences, Wuhan, Hubei, 430030, P.R. China.
| |
Collapse
|
41
|
Pan X, Hao E, Zhang F, Wei W, Du Z, Yan G, Wang X, Deng J, Hou X. Diabetes cardiomyopathy: targeted regulation of mitochondrial dysfunction and therapeutic potential of plant secondary metabolites. Front Pharmacol 2024; 15:1401961. [PMID: 39045049 PMCID: PMC11263127 DOI: 10.3389/fphar.2024.1401961] [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: 03/16/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a specific heart condition in diabetic patients, which is a major cause of heart failure and significantly affects quality of life. DCM is manifested as abnormal cardiac structure and function in the absence of ischaemic or hypertensive heart disease in individuals with diabetes. Although the development of DCM involves multiple pathological mechanisms, mitochondrial dysfunction is considered to play a crucial role. The regulatory mechanisms of mitochondrial dysfunction mainly include mitochondrial dynamics, oxidative stress, calcium handling, uncoupling, biogenesis, mitophagy, and insulin signaling. Targeting mitochondrial function in the treatment of DCM has attracted increasing attention. Studies have shown that plant secondary metabolites contribute to improving mitochondrial function and alleviating the development of DCM. This review outlines the role of mitochondrial dysfunction in the pathogenesis of DCM and discusses the regulatory mechanism for mitochondrial dysfunction. In addition, it also summarizes treatment strategies based on plant secondary metabolites. These strategies targeting the treatment of mitochondrial dysfunction may help prevent and treat DCM.
Collapse
Affiliation(s)
- Xianglong Pan
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Fan Zhang
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wei Wei
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zhengcai Du
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Guangli Yan
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xijun Wang
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xiaotao Hou
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
42
|
Huyan T, Fan L, Zheng ZY, Zhao JH, Han ZR, Wu P, Ma Q, Du YQ, Shi YD, Gu CY, Li XJ, Wang WH, Zhang L, Tie L. ROCK1 inhibition improves wound healing in diabetes via RIPK4/AMPK pathway. Acta Pharmacol Sin 2024; 45:1477-1491. [PMID: 38538716 PMCID: PMC11192920 DOI: 10.1038/s41401-024-01246-3] [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: 10/04/2023] [Accepted: 02/19/2024] [Indexed: 06/23/2024]
Abstract
Refractory wounds are a severe complication of diabetes mellitus that often leads to amputation because of the lack of effective treatments and therapeutic targets. The pathogenesis of refractory wounds is complex, involving many types of cells. Rho-associated protein kinase-1 (ROCK1) phosphorylates a series of substrates that trigger downstream signaling pathways, affecting multiple cellular processes, including cell migration, communication, and proliferation. The present study investigated the role of ROCK1 in diabetic wound healing and molecular mechanisms. Our results showed that ROCK1 expression significantly increased in wound granulation tissues in diabetic patients, streptozotocin (STZ)-induced diabetic mice, and db/db diabetic mice. Wound healing and blood perfusion were dose-dependently improved by the ROCK1 inhibitor fasudil in diabetic mice. In endothelial cells, fasudil and ROCK1 siRNA significantly elevated the phosphorylation of adenosine monophosphate-activated protein kinase at Thr172 (pThr172-AMPKα), the activity of endothelial nitric oxide synthase (eNOS), and suppressed the levels of mitochondrial reactive oxygen species (mtROS) and nitrotyrosine formation. Experiments using integrated bioinformatics analysis and coimmunoprecipitation established that ROCK1 inhibited pThr172-AMPKα by binding to receptor-interacting serine/threonine kinase 4 (RIPK4). These results suggest that fasudil accelerated wound repair and improved angiogenesis at least partially through the ROCK1/RIPK4/AMPK pathway. Fasudil may be a potential treatment for refractory wounds in diabetic patients.
Collapse
Affiliation(s)
- Tianru Huyan
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
- Department of Wound Healing Center and Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Lu Fan
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhong-Yuan Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Jing-Hui Zhao
- Department of Wound Healing Center and Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, 100191, China
| | - Zhen-Ru Han
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Pin Wu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Qun Ma
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Ya-Qin Du
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Yun-di Shi
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Chun-Yan Gu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xue-Jun Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China
| | - Wen-Hui Wang
- Department of Dermatology, Peking University Third Hospital, Beijing, 100191, China
| | - Long Zhang
- Department of Wound Healing Center and Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, 100191, China.
| | - Lu Tie
- Department of Pharmacology, School of Basic Medical Sciences, Peking University and Beijing Key Laboratory of Tumor Systems Biology, Peking University, Beijing, 100191, China.
| |
Collapse
|
43
|
Kong C, Guo Z, Liu F, Tang N, Wang M, Yang D, Li C, Yang Z, Ma Y, Wang P, Tang Q. Triad3A-Mediated K48-Linked ubiquitination and degradation of TLR9 impairs mitochondrial bioenergetics and exacerbates diabetic cardiomyopathy. J Adv Res 2024; 61:65-81. [PMID: 37625569 PMCID: PMC11258663 DOI: 10.1016/j.jare.2023.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION Targeted protein degradation represents a promising therapeutic approach, while diabetic cardiomyopathy (DCM) arises as a consequence of aberrant insulin secretion and impaired glucose and lipid metabolism in the heart.. OBJECTIVES Considering that the Toll-like receptor 9 (TLR9) signaling pathway plays a pivotal role in regulating energy metabolism, safeguarding cardiomyocytes, and influencing glucose uptake, the primary objective of this study was to investigate the impact of TLR9 on diabetic cardiomyopathy (DCM) and elucidate its underlying mechanism. METHODS Mouse model of DCM was established using intraperitoneal injection of STZ, and mice were transfected with adeno-associated virus serotype 9-TLR9 (AAV9-TLR9) to assess the role of TLR9 in DCM. To explore the mechanism of TLR9 in regulating DCM disease progression, we conducted interactome analysis and employed multiple molecular approaches. RESULTS Our study revealed a significant correlation between TLR9 expression and mouse DCM. TLR9 overexpression markedly mitigated cardiac dysfunction, myocardial fibrosis, oxidative stress, and apoptosis in DCM, while inflammation levels remained relatively unaffected. Mechanistically, TLR9 overexpression positively modulated mitochondrial bioenergetics and activated the AMPK-PGC1a signaling pathway. Furthermore, we identified Triad3A as an interacting protein that facilitated TLR9's proteasomal degradation through K48-linked ubiquitination. Inhibiting Triad3A expression improved cardiac function and pathological changes in DCM by enhancing TLR9 activity. CONCLUSIONS The findings of this study highlight the critical role of TLR9 in maintaining cardiac function and mitigating pathological alterations in diabetic cardiomyopathy. Triad3A-mediated regulation of TLR9 expression and function has significant implications for understanding the pathogenesis of DCM. Targeting TLR9 and its interactions with Triad3A may hold promise for the development of novel therapeutic strategies for diabetic cardiomyopathy. Further research is warranted to fully explore the therapeutic potential of TLR9 modulation in the context of cardiovascular diseases.
Collapse
Affiliation(s)
- Chunyan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Fangyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Mingyu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Chenfei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Yulan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Pan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
44
|
Yan M, Su L, Wu K, Mei Y, Liu Z, Chen Y, Zeng W, Xiao Y, Zhang J, Cai G, Bai Y. USP7 promotes cardiometabolic disorders and mitochondrial homeostasis dysfunction in diabetic mice via stabilizing PGC1β. Pharmacol Res 2024; 205:107235. [PMID: 38815879 DOI: 10.1016/j.phrs.2024.107235] [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: 01/30/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/01/2024]
Abstract
Diabetic cardiomyopathy (DCM) is a major complication of diabetes and is characterized by left ventricular dysfunction. Currently, there is a lack of effective treatments for DCM. Ubiquitin-specific protease 7 (USP7) plays a key role in various diseases. However, whether USP7 is involved in DCM has not been established. In this study, we demonstrated that USP7 was upregulated in diabetic mouse hearts and NMCMs co-treated with HG+PA or H9c2 cells treated with PA. Abnormalities in diabetic heart morphology and function were reversed by USP7 silencing through conditional gene knockout or chemical inhibition. Proteomic analysis coupled with biochemical validation confirmed that PCG1β was one of the direct protein substrates of USP7 and aggravated myocardial damage through coactivation of the PPARα signaling pathway. USP7 silencing restored the expression of fatty acid metabolism-related proteins and restored mitochondrial homeostasis by inhibiting mitochondrial fission and promoting fusion events. Similar effects were also observed in vitro. Our data demonstrated that USP7 promoted cardiometabolic metabolism disorders and mitochondrial homeostasis dysfunction via stabilizing PCG1β and suggested that silencing USP7 may be a therapeutic strategy for DCM.
Collapse
Affiliation(s)
- Meiling Yan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China.
| | - Liyan Su
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China
| | - Kaile Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yu Mei
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhou Liu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yifan Chen
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wenru Zeng
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yang Xiao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jingfei Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guida Cai
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yunlong Bai
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, China; Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou, China; Guangdong Key Laboratory of Metabolic Disease Prevention and Treatment of Traditional Chinese Medicine, China; Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China; Department of Pharmacology (State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Medicine Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China; Translational Medicine Research and Cooperation Center of Northern China, Chronic Disease Research Institute, Heilongjiang Academy of Medical Sciences, Harbin, China.
| |
Collapse
|
45
|
Zhou J, Hao J, Zhong Z, Yang J, Lv T, Zhao B, Lin H, Chi J, Guo H. Fecal Microbiota Transplantation in Mice Exerts a Protective Effect Against Doxorubicin-Induced Cardiac Toxicity by Regulating Nrf2-Mediated Cardiac Mitochondrial Fission and Fusion. Antioxid Redox Signal 2024; 41:1-23. [PMID: 37756370 DOI: 10.1089/ars.2023.0355] [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] [Indexed: 09/29/2023]
Abstract
Aims: The relationship between the gut microbiota and cardiovascular system has been increasingly clarified. Fecal microbiota transplantation (FMT), used to improve gut microbiota, has been applied clinically for disease treatment and has great potential in combating doxorubicin (DOX)-induced cardiotoxicity. However, the application of FMT in the cardiovascular field and its molecular mechanisms are poorly understood. Results: During DOX-induced stress, FMT alters the gut microbiota and serum metabolites, leading to a reduction in cardiac injury. Correlation analysis indicated a close association between serum metabolite indole-3-propionic acid (IPA) and cardiac function. FMT and IPA achieve this by facilitating the translocation of Nfe2l2 (Nrf2) from the cytoplasm to the nucleus, thereby activating the expression of antioxidant molecules, reducing reactive oxygen species production, and inhibiting excessive mitochondrial fission. Consequently, mitochondrial function is preserved, leading to the mitigation of cardiac injury under DOX-induced stress. Innovation: FMT has the ability to modify the composition of the gut microbiota, providing not only protection to the intestinal mucosa but also influencing the generation of serum metabolites and regulating the Nrf2 gene to modulate the balance of cardiac mitochondrial fission and fusion. This study comprehensively demonstrates the efficacy of FMT in countering DOX-induced myocardial damage and elucidates the pathways linking the microbiota and the heart. Conclusion: FMT alters the gut microbiota and serum metabolites of recipient mice, promoting nuclear translocation of Nrf2 and subsequent activation of downstream antioxidant molecule expression, while inhibiting excessive mitochondrial fission to preserve cardiac integrity. Correlation analysis highlights IPA as a key contributor among differentially regulated metabolites.
Collapse
Affiliation(s)
- Jiedong Zhou
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Jinjin Hao
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Zuoquan Zhong
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, China
| | - Juntao Yang
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Tingting Lv
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Bingjie Zhao
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Hui Lin
- Department of Cardiovascular, The Affiliated Lihuili Hospital of Ningbo University, Healthy Science Center, Ningbo University, Ningbo, China
| | - Jufang Chi
- Department of Cardiology, Zhuji People's Hospital, Shaoxing, China
| | - Hangyuan Guo
- School of Medicine, Shaoxing University, Shaoxing, China
| |
Collapse
|
46
|
Zhang BF, Wu ZH, Chen K, Jin HJ, Wu J, Huang ZY, Lu XW, Zheng XT. Dynamin-related protein 1 mediates the therapeutic effect of isoliquiritigenin in diabetic intimal hyperplasia via regulation of mitochondrial fission. Hypertens Res 2024; 47:1908-1924. [PMID: 38750218 DOI: 10.1038/s41440-024-01681-z] [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: 08/08/2023] [Revised: 03/05/2024] [Accepted: 03/22/2024] [Indexed: 07/06/2024]
Abstract
Phenotypic shift of vascular smooth muscle cells (VSMCs) plays a key role in intimal hyperplasia, especially in patients with diabetes mellitus (DM). This study aimed to investigate the role of dynamin-related protein 1 (DRP1) in mitochondrial fission-mediated VSMC phenotypic shift and to clarify whether DRP1 is the therapeutic target of isoliquiritigenin (ISL). Wire injury of carotid artery or platelet-derived growth factor treatment was performed in DM mice or high-glucose cultured human aortic smooth muscle cells (HASMCs), respectively. The effects of DRP1 silencing on DM-induced intimal hyperplasia were investigated both in vivo and in vitro. Phenotypic shift of HASMCs was evaluated by detection of reactive oxygen species (ROS) generation, cell viability, and related protein expressions. The effects of ISL on DM-induced intimal hyperplasia were evaluated both in vivo and in vitro. DRP1 silencing and ISL treatment attenuated DM-induced intimal hyperplasia with reduced ROS generation, cell viability, and VSMC dedifferentiation. The GTPase domain of DRP1 protein played a critical role in mitochondrial fission in DM-induced VSMC phenotypic shift. Cellular experiments showed that ISL inhibited mitochondrial fission and reduced the GTPase activity of DRP1, which was achieved by the directly binding to K216 of the DRP1 GTPase domain. ISL attenuated mouse intimal hyperplasia by reducing GTPase activity of DRP1 and inhibiting mitochondrial fission in vivo. In conclusion, increased GTPase activity of DRP1 aggregated DM-induced intimal hyperplasia by increasing mitochondrial fission-mediated VSMC phenotypic shift. ISL attenuated mouse intimal hyperplasia by reducing DRP1 GTPase activity and inhibiting mitochondrial fission of VSMCs.
Collapse
MESH Headings
- Animals
- Mitochondrial Dynamics/drug effects
- Dynamins/metabolism
- Hyperplasia
- Chalcones/pharmacology
- Chalcones/therapeutic use
- Mice
- Humans
- Male
- Diabetes Mellitus, Experimental/drug therapy
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Reactive Oxygen Species/metabolism
- Myocytes, Smooth Muscle/drug effects
- Cells, Cultured
- Mice, Inbred C57BL
- Tunica Intima/drug effects
- Tunica Intima/pathology
- Tunica Intima/metabolism
Collapse
Affiliation(s)
- Bao-Fu Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Zi-Heng Wu
- Department of Vascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Kui Chen
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Hao-Jie Jin
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Jun Wu
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Zi-Yi Huang
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China
| | - Xin-Wu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiang-Tao Zheng
- Department of Vascular Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325015, China.
| |
Collapse
|
47
|
Huang SQ, Cao KX, Wang CL, Chen PL, Chen YX, Zhang YT, Yu SH, Bai ZX, Guo S, Liao MX, Li QW, Zhang GQ, He J, Xu YM. Decreasing mitochondrial fission ameliorates HIF-1α-dependent pathological retinal angiogenesis. Acta Pharmacol Sin 2024; 45:1438-1450. [PMID: 38565961 PMCID: PMC11192750 DOI: 10.1038/s41401-024-01262-3] [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: 08/27/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
Angiogenesis plays a critical role in many pathological processes, including irreversible blindness in eye diseases such as retinopathy of prematurity. Endothelial mitochondria are dynamic organelles that undergo constant fusion and fission and are critical signalling hubs that modulate angiogenesis by coordinating reactive oxygen species (ROS) production and calcium signalling and metabolism. In this study, we investigated the role of mitochondrial dynamics in pathological retinal angiogenesis. We showed that treatment with vascular endothelial growth factor (VEGF; 20 ng/ml) induced mitochondrial fission in HUVECs by promoting the phosphorylation of dynamin-related protein 1 (DRP1). DRP1 knockdown or pretreatment with the DRP1 inhibitor Mdivi-1 (5 μM) blocked VEGF-induced cell migration, proliferation, and tube formation in HUVECs. We demonstrated that VEGF treatment increased mitochondrial ROS production in HUVECs, which was necessary for HIF-1α-dependent glycolysis, as well as proliferation, migration, and tube formation, and the inhibition of mitochondrial fission prevented VEGF-induced mitochondrial ROS production. In an oxygen-induced retinopathy (OIR) mouse model, we found that active DRP1 was highly expressed in endothelial cells in neovascular tufts. The administration of Mdivi-1 (10 mg·kg-1·d-1, i.p.) for three days from postnatal day (P) 13 until P15 significantly alleviated pathological angiogenesis in the retina. Our results suggest that targeting mitochondrial fission may be a therapeutic strategy for proliferative retinopathies and other diseases that are dependent on pathological angiogenesis.
Collapse
Affiliation(s)
- Shu-Qi Huang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kai-Xiang Cao
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Cai-Ling Wang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Pei-Ling Chen
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi-Xin Chen
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Ting Zhang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shi-Hui Yu
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zai-Xia Bai
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shuai Guo
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Mu-Xi Liao
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, China
| | - Qiao-Wen Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511520, China
| | - Guo-Qi Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511520, China.
| | - Jun He
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, China.
| | - Yi-Ming Xu
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
48
|
Nagata Y, Quynh NT, Aono H, Harada K, Miyamoto H, Fujimoto N. Melatonin Inhibits Chemical Carcinogen-mediated Malignant Transformation of Urothelial Cells: In Vitro Evidence. Cancer Genomics Proteomics 2024; 21:388-394. [PMID: 38944424 PMCID: PMC11215424 DOI: 10.21873/cgp.20456] [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: 01/21/2024] [Revised: 05/15/2024] [Accepted: 05/24/2024] [Indexed: 07/01/2024] Open
Abstract
BACKGROUND/AIM The efficacy of melatonin and its biological significance in human bladder cancer remain poorly understood. This study aimed to investigate the functional role of melatonin in urothelial carcinogenesis. MATERIALS AND METHODS In human normal urothelial SVHUC cells with exposure to the chemical carcinogen 3-methylcholanthrene, we assessed the effects of melatonin on the neoplastic/malignant transformation. RESULTS In the in vitro system with carcinogen challenge, melatonin significantly prevented the neoplastic transformation of SV-HUC-1 cells. In addition, melatonin treatment resulted in increased expression of SIRT1, Rb1, and E-cadherin, and decreased expression of N-cadherin and FGFR3 in SV-HUC-1 cells. Furthermore, publicly available datasets from GSE3167 revealed that the expression of melatonin receptor 1 and melatonin receptor 2 was significantly down-regulated in bladder urothelial carcinoma tissues, compared with adjacent normal urothelial tissues. CONCLUSION These findings indicate that melatonin serves as a suppressor for urothelial tumorigenesis. To the best of our knowledge, this is the first preclinical study demonstrating the impact of melatonin on the development of urothelial cancer.
Collapse
Affiliation(s)
- Yujiro Nagata
- Department of Urology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan;
| | - Nguyen Thu Quynh
- Department of Urology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Hisami Aono
- Department of Urology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Kenichi Harada
- Department of Urology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| | - Hiroshi Miyamoto
- Departments of Pathology & Laboratory Medicine and Urology, University of Rochester Medical Center, Rochester, NY, U.S.A
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health School of Medicine, Kitakyushu, Japan
| |
Collapse
|
49
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
50
|
Zhang X, Zhang W, Wang Y, Zhang Y, Zhang D, Qin G, Zhou J, Chen L. SIRT1-regulated ROS generation activates NMDAR2B phosphorylation to promote central sensitization and allodynia in a male chronic migraine rat model. Front Mol Neurosci 2024; 17:1387481. [PMID: 38840778 PMCID: PMC11150646 DOI: 10.3389/fnmol.2024.1387481] [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: 02/17/2024] [Accepted: 04/25/2024] [Indexed: 06/07/2024] Open
Abstract
Background Central sensitization is one of the pivotal pathological mechanisms in chronic migraine (CM). Silent information regulator 1 (SIRT1) was shown to be involved in CM, but its specific mechanism is unclear. Reactive oxygen species (ROS) are increasingly regarded as important signaling molecules in several models of pain. However, studies about the role of ROS in the central sensitization of CM model are rare. We thus explored the specific process of SIRT1 involvement in the central sensitization of CM, focusing on the ROS pathway. Methods Inflammatory soup was repeatedly administered to male Sprague-Dawley rats to establish a CM model. The SIRT1 expression level in trigeminal nucleus caudalis (TNC) tissues was assessed by qRT-PCR and Western blotting analysis. The levels of ROS were detected by a Tissue Reactive Oxygen Detection Kit, DHE staining, and the fluorescence signal intensity of 8-OHdG. A ROS scavenger (tempol), a SIRT1 activator (SRT1720), a SIRT1 inhibitor (EX527), and a mitochondrial fission inhibitor (Mdivi-1) were used to investigate the specific molecular mechanisms involved. NMDAR2B, CGRP, ERK, and mitochondrial fission-related protein were evaluated by Western blotting, and the CGRP level in frozen sections of the TNC was detected via immunofluorescence staining. Results After repeated inflammatory soup infusion and successful establishment of the CM rat model, SIRT1 expression was found to be significantly reduced, accompanied by elevated ROS levels. Treatment with Tempol, SRT1720, or Mdivi-1 alleviated allodynia and reduced the increase in NMDAR2B phosphorylation and CGRP and ERK phosphorylation in the CM rat. In contrast, EX527 had the opposite effect in CM rat. SRT1720 and EX527 decreased and increased ROS levels, respectively, in CM rats, and tempol reversed the aggravating effect of EX527 in CM rats. Furthermore, the regulatory effect of SIRT1 on ROS may include the involvement of the mitochondrial fission protein DRP1. Conclusion The results indicate the importance of SIRT1 in CM may be due to its role in regulating the production of ROS, which are involved in modulating central sensitization in CM. These findings could lead to new ideas for CM treatment with the use of SIRT1 agonists and antioxidants.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yanyun Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dunke Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guangcheng Qin
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiying Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lixue Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|