1
|
Yi G, Li M, Zhou J, Li J, Song X, Li S, Liu J, Zhang H, Chen Z. Novel pH-responsive lipid nanoparticles deliver UA-mediated mitophagy and ferroptosis for osteoarthritis treatment. Mater Today Bio 2025; 32:101697. [PMID: 40225130 PMCID: PMC11986606 DOI: 10.1016/j.mtbio.2025.101697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Synovial inflammation plays a crucial role in osteoarthritis (OA) development, leading to chronic inflammation and cartilage destruction. Although targeting synovitis can alleviate OA, clinical outcomes have been disappointing due to poor drug targeting and joint cavity heterogeneity. This study presents pH-responsive lipid nanoparticles (LNPs@UA), loaded with Urolithin A (UA), as a potential OA treatment. LNPs@UA showed uniform particle size, low zeta potential, and effective mitochondria-targeting and pH-responsive capabilities. In vitro, LNPs@UA reduced reactive oxygen species (ROS), pro-inflammatory factors (IL-1β, IL-6, TNF-α), and promoted M2 macrophage polarization. It improved mitochondrial structure, enhanced autophagy, and inhibited ferroptosis. In vivo, LNPs@UA alleviated OA progression in an ACLT-induced OA mouse model. Transcriptomic analysis revealed inhibition of NF-κB signaling and activation of repair pathways. These results suggest LNPs@UA could offer a promising therapeutic approach for OA.
Collapse
Affiliation(s)
- Guoliang Yi
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
- Guizhou Medical University, Guiyang, 550004, China
| | - Min Li
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiayi Zhou
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jinxin Li
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Xizheng Song
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Siming Li
- Guizhou Medical University, Guiyang, 550004, China
- Department of Orthopedics, Guangzhou Red Cross Hospital, Guangzhou, 510220, China
| | - Jianghua Liu
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haowei Zhang
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zhiwei Chen
- Department of Orthopedics, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
2
|
Wang D, Qu X, Zhang Z, Zhou G. New developments in the role of ferroptosis in sepsis‑induced cardiomyopathy (Review). Mol Med Rep 2025; 31:118. [PMID: 40052561 PMCID: PMC11904766 DOI: 10.3892/mmr.2025.13483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/15/2025] [Indexed: 03/15/2025] Open
Abstract
Sepsis is a life‑threatening organ dysfunction disorder caused by dysfunctional host response to infection. Sepsis‑induced cardiomyopathy (SIC) is a common and serious complication of sepsis, and it is associated with increased mortality rates; however, its specific pathogenesis is still unclear. Ferroptosis, which is an iron‑dependent form of programmed cell death, is involved in the pathophysiology of SIC. Further study on the mechanism and therapeutic targets of ferroptosis in SIC may provide new strategies for clinical diagnosis and treatment of this condition. The present article reviews the mechanisms between SIC and ferroptosis, summarizes the progress in research of the involvement of ferroptosis in SIC and provides new potential strategies for further research and treatment in the future.
Collapse
Affiliation(s)
- Dingdeng Wang
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Xinguang Qu
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Zhaohui Zhang
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| | - Gaosheng Zhou
- Department of Critical Care Medicine, The First College of Clinical Medical Science, China Three Gorges University, Yichang Central People's Hospital, Yichang, Hubei 443003, P.R. China
| |
Collapse
|
3
|
Li D, Xie M, Zeng H, Yu J, Xu R, Wang Z, Huang Y, Yang Y, Sun Y. UPR mt alleviates bone cancer pain through the restoration of mitochondrial function. Exp Cell Res 2025; 448:114568. [PMID: 40273969 DOI: 10.1016/j.yexcr.2025.114568] [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: 01/31/2025] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
The mitochondrial unfolded protein response (UPRmt) is an intracellular retrograde signaling process that facilitates the restoration of mitochondrial homeostasis. Mitochondria are essential for neuronal signaling, and their dysfunction has been implicated as a significant mechanism in the development of chronic pain. Nevertheless, little is known about the exact function of UPRmt in bone cancer pain (BCP). This research intended to explore the connection between UPRmt and the progression of BCP. In BCP group, the ultrastructure of spinal cord mitochondria was disrupted, accompanied by a decline in ATP levels and a decrease in Mitochondrial membrane potential (MMP). Concurrently, mRNA and protein levels of UPRmt marker proteins (Atf5, Hsp60, LonP1, and ClpP) were upregulated, with the expression of Atf5, a key transcription factor of UPRmt, notably enhanced in spinal dorsal horn neurons. Nicotinamide riboside (NR)-mediated pharmacological augmentation of the UPRmt significantly alleviated BCP-induced nociceptive hypersensitivity, as demonstrated by elevated mechanical withdrawal thresholds and diminished spontaneous flinching behavior. Concomitant mitochondrial functional recovery was evidenced by restoration of MMP and normalization of ATP level. Notably, genetic knockdown of activating transcription factor 5 (Atf5) abolished both NR-induced UPRmt activation and the consequent protection against rotenone-mediated mitochondrial dysfunction. These findings establish UPRmt potentiation as an effective strategy for ameliorating mitochondrial dysfunction and attenuating BCP-associated nociception, proposing this pathway as a novel therapeutic target for clinical pain management.
Collapse
Affiliation(s)
- Dan Li
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingming Xie
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Haohao Zeng
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiacheng Yu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Rui Xu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhen Wang
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Yulin Huang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Yan Yang
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Yu'e Sun
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
4
|
Wu H, Diao H, Zhang F, Jiang W, Pan T, Bian Y. Organelle interplay in cardiovascular diseases: Mechanisms, pathogenesis, and therapeutic perspectives. Biomed Pharmacother 2025; 185:117978. [PMID: 40073746 DOI: 10.1016/j.biopha.2025.117978] [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: 10/31/2024] [Revised: 02/16/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of rising morbidity and mortality among humans worldwide; however, our approach to the pathogenesis, exploration, and management of CVDs still remains limited. As the heart consists of cardiomyocytes, cardiac fibroblasts, endothelial cells, smooth muscle cells, and several types of cells, different types of dysfunction in the interplay between organelles play an important damaging role, resulting in cardiac pathologies. The interplay between cellular organelles is intricate and vital for maintaining cellular homeostasis, as highlighted by multiple diseases linked to the dysfunction of both organelles. Many studies have revealed the potential mechanisms by which organelles communicate with each other and regulate the pathological processes of CVDs together. However, gaps remain in fully understanding the complexity of these interactions and translating these insights into therapeutic approaches. In this review, we summarized how the interplay between cellular organelles in the cardiomyocytes alters in various heart diseases. We find underexplored areas, such as the crucial signaling pathways governing organelle communication, and discuss their implications for disease future progression. Furthermore, we evaluate emerging potential medicines aimed at restoring organelle interactions. Finally, we propose future directions for researching to advance the development of novel medicines and therapies, addressing current gaps and providing a theoretical basis for improved clinical outcomes in CVDs.
Collapse
Affiliation(s)
- Han Wu
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Hongtao Diao
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Feng Zhang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Weitao Jiang
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Tengfei Pan
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yu Bian
- Department of Pharmacology (The State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China.
| |
Collapse
|
5
|
Lu J, Cai J, Zhou Z, Ma J, Han T, Lu N, Zhu L. Gel@CAT-L hydrogel mediates mitochondrial unfolded protein response to regulate reactive oxygen species and mitochondrial homeostasis in osteoarthritis. Biomaterials 2025; 321:123283. [PMID: 40222260 DOI: 10.1016/j.biomaterials.2025.123283] [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: 01/10/2025] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVE This study investigates the role of Gelatin-Catalase (Gel@CAT)-L hydrogel in mediating reactive oxygen species (ROS) production and maintaining mitochondrial homeostasis through SIRT3-mediated unfolded protein response (UPRmt), while exploring its involvement in the molecular mechanism of osteoarthritis (OA). METHODS Self-assembled Gel@CAT-L hydrogels were fabricated and characterized using transmission electron microscopy, mechanical testing, external release property evaluation, and oxygen production measurement. Biocompatibility was assessed via live/dead cell staining and CCK8 assays. An OA mouse model was established using destabilization of the medial meniscus (DMM) surgery. X-ray and micro-CT imaging were employed to evaluate the structural integrity of the mouse knee joints, while histological staining was used to assess cartilage degeneration. Immunohistochemistry was performed to analyze the expression of proteins including Col2a1, Aggrecan, MMP13, ADAMTS5, SIRT3, PINK1, and Parkin. Multi-omics analyses-encompassing high-throughput sequencing, proteomics, and metabolomics-were conducted to identify key genes and metabolic pathways targeted by Gel@CAT-L hydrogel intervention in OA. Immunofluorescence techniques were utilized to measure ROS levels, mitochondrial membrane potential, and the expression of SIRT3, PINK1, Parkin, LYSO, LC3B, Col2a1, and MMP13 in primary mouse chondrocytes and mouse knee joints. Flow cytometry was applied to quantify ROS-positive cells. RT-qPCR analysis was conducted to determine mRNA levels of Aggrecan, Col2a1, ADAMTS5, MMP13, SIRT3, mtDNA, HSP60, LONP1, CLPP, and Atf5 in primary mouse chondrocytes, mouse knee joints, and human knee joints. Western blotting was performed to measure protein expression levels of SIRT3, HSP60, LONP1, CLPP, and Atf5 in both primary mouse chondrocytes and mouse knee joints. Additionally, 20 samples each from the control (CON) and OA groups were collected for analysis. Hematoxylin and eosin staining was used to evaluate cartilage degeneration in human knee joints. The Mankin histological scoring system quantified the degree of cartilage degradation, while immunofluorescence analyzed SIRT3 protein expression in human knee joints. RESULTS In vitro experiments demonstrated that self-assembled Gel@CAT-L hydrogels exhibited excellent biodegradability and oxygen-releasing capabilities, providing a stable three-dimensional environment conducive to cell viability and proliferation while reducing ROS levels. Multi-omics analysis identified SIRT3 as a key regulatory gene in mitigating OA and revealed its central role in the UPRmt pathway. Furthermore, Gel@CAT-L was confirmed to regulate mitochondrial homeostasis. Both in vitro experiments and in vivo mouse model studies confirmed that Gel@CAT-L significantly reduced ROS levels and regulated mitochondrial autophagy by activating the SIRT3-mediated UPRmt pathway, thereby improving the pathological state of OA. Clinical trials indicated downregulation of SIRT3 and UPRmt-related proteins in OA patients. CONCLUSION Gel@CAT-L hydrogel activates SIRT3-mediated UPRmt to regulate ROS and mitochondrial homeostasis, providing potential therapeutic benefits for OA.
Collapse
Affiliation(s)
- Jiajia Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China
| | - Jiao Cai
- Department of Medical Administration, Shanghai Changzheng Hospital, Shanghai, 200434, China
| | - Zhibin Zhou
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China
| | - Jun Ma
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China; Department of Orthopaedic Trauma, Naval Medical Center of PLA, Naval Medical University, Shanghai, 200001, China
| | - Tianyu Han
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China.
| | - Nan Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| | - Lei Zhu
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China.
| |
Collapse
|
6
|
Chang X, Zhou S, Huang Y, Liu J, Wang Y, Guan X, Wu Q, Liu Z, Liu R. Zishen Huoxue decoction (ZSHX) alleviates ischemic myocardial injury (MI) via Sirt5-β-tubulin mediated synergistic mechanism of "mitophagy-unfolded protein response" and mitophagy. Chin J Nat Med 2025; 23:311-321. [PMID: 40122661 DOI: 10.1016/s1875-5364(25)60838-7] [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: 12/13/2023] [Revised: 04/27/2024] [Accepted: 05/06/2024] [Indexed: 03/25/2025]
Abstract
Zishen Huoxue decoction (ZSHX) enhances cardiomyocyte viability following hypoxic stress; however, its upstream therapeutic targets remain unclear. Network pharmacology and RNA sequencing analyses revealed that ZSHX target genes were closely associated with mitophagy and apoptosis in the mitochondrial pathway. In vitro, ZSHX inhibited pathological mitochondrial fission following hypoxic stress, regulated FUN14 domain-containing protein 1 (FUNDC1)-related mitophagy, and increased the levels of mitophagy lysosomes and microtubule-associated protein 1 light chain 3 beta II (LC3II)/translocase of outer mitochondrial membrane 20 (TOM20) expression while inhibiting the over-activated mitochondrial unfolded protein response. Additionally, ZSHX regulated the stability of beta-tubulin through Sirtuin 5 (SIRT5) and could modulate FUNDC1-related synergistic mechanisms of mitophagy and unfolded protein response in the mitochondria (UPRmt) via the SIRT5 and -β-tubulin axis. This targeting pathway may be crucial for cardiomyocytes to resist hypoxia. Collectively, these findings suggest that ZSHX can protect against cardiomyocyte injury via the SIRT5-β-tubulin axis, which may be associated with the synergistic protective mechanism of SIRT5-β-tubulin axis-related mitophagy and UPRmt on cardiomyocytes.
Collapse
Affiliation(s)
- Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053
| | - Siyuan Zhou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053
| | - Yu Huang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053
| | - Jinfeng Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053
| | - Yanli Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053
| | - Xuanke Guan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053
| | - Qiaomin Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053
| | - Zhiming Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053,.
| | - Ruxiu Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053,.
| |
Collapse
|
7
|
Huang W, Lou A, Wang J, Wang Y, Zhang W, Li J, Wang S, Geng S, Wang G, Li X. TMBIM1 ameliorates sepsis-induced cardiac dysfunction by promoting Parkin-mediated mitophagy. FASEB J 2025; 39:e70397. [PMID: 39937566 DOI: 10.1096/fj.202402599rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/13/2025]
Abstract
Myocardial dysfunction is a significant complication of sepsis that is associated with elevated mortality rates. Transmembrane BAX inhibitor motif containing 1 (TMBIM1), a stress-responsive protein, has garnered interest in the field of cardiovascular disease for its cardioprotective properties. Nevertheless, the role of TMBIM1 on sepsis-induced cardiac dysfunction (SICD) remains unknown. Here, our findings revealed a significant elevation in TMBIM1 expression within the myocardium following endotoxin challenge and further demonstrate the cardioprotective effects of TMBIM1 through adenovirus-mediated gene manipulation. Notably, lipopolysaccharide exposure markedly induced mitochondrial dysfunction in cardiomyocytes, which was effectively alleviated by TMBIM1 overexpression, while TMBIM1 knockdown exacerbated this dysfunction. Moreover, in cardiomyocytes subjected to endotoxin challenge, TMBIM1 was observed to interact with Parkin, facilitating its translocation from the cytosol to damaged mitochondria. This interaction enhanced the activation of mitophagy, thereby promoting the clearance of dysfunctional mitochondria and subsequently mitigating cellular injury. Hence, targeting TMBIM1 could be a novel therapeutic strategy for treating SICD.
Collapse
Affiliation(s)
- Weichang Huang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Critical Care Medicine, The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), Southern Medical University, Dongguan, China
| | - Anni Lou
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuegang Wang
- Department of Cardiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenyong Zhang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jierui Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuanghu Wang
- The Laboratory of Clinical Pharmacy, The People's Hospital of Lishui, Lishui, Zhejiang, China
| | - Shiyu Geng
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guozhen Wang
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Liu S, Faitg J, Tissot C, Konstantopoulos D, Laws R, Bourdier G, Andreux PA, Davey T, Gallart-Ayala H, Ivanisevic J, Singh A, Rinsch C, Marcinek DJ, D’Amico D. Urolithin A provides cardioprotection and mitochondrial quality enhancement preclinically and improves human cardiovascular health biomarkers. iScience 2025; 28:111814. [PMID: 40034121 PMCID: PMC11875685 DOI: 10.1016/j.isci.2025.111814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/04/2024] [Accepted: 01/10/2025] [Indexed: 03/05/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain the primary cause of global mortality. Nutritional interventions hold promise to reduce CVD risks in an increasingly aging population. However, few nutritional interventions are proven to support heart health and act mostly on blood lipid homeostasis rather than at cardiac cell level. Here, we show that mitochondrial quality dysfunctions are common hallmarks in human cardiomyocytes upon heart aging and in chronic conditions. Preclinically, the post-biotic and mitophagy activator, urolithin A (UA), reduced both systolic and diastolic cardiac dysfunction in models of natural aging and heart failure. At a cellular level, this was associated with a recovery of mitochondrial ultrastructural defects and mitophagy. In humans, UA supplementation for 4 months in healthy older adults significantly reduced plasma ceramides clinically validated to predict CVD risks. These findings extend and translate UA's benefits to heart health, making UA a promising nutritional intervention to support cardiovascular function as we age.
Collapse
Affiliation(s)
- Sophia Liu
- Department of Radiology, University of Washington Medical Center, Box 358050, Seattle, WA 98109, USA
| | - Julie Faitg
- Amazentis, EPFL Innovation Park, Lausanne, Switzerland
| | | | | | - Ross Laws
- Electron Microscopy Research Services, Newcastle University, Newcastle, UK
| | | | | | - Tracey Davey
- Electron Microscopy Research Services, Newcastle University, Newcastle, UK
| | - Hector Gallart-Ayala
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Julijana Ivanisevic
- Metabolomics Platform, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Anurag Singh
- Amazentis, EPFL Innovation Park, Lausanne, Switzerland
| | - Chris Rinsch
- Amazentis, EPFL Innovation Park, Lausanne, Switzerland
| | - David J. Marcinek
- Department of Radiology, University of Washington Medical Center, Box 358050, Seattle, WA 98109, USA
| | | |
Collapse
|
9
|
Zhang Z, Yu X, Li J, Shen X, Fu W, Liu Y, Dong X, Wang Z. Irisin Mitigates Doxorubicin-Induced Cardiotoxicity by Reducing Oxidative Stress and Inflammation via Modulation of the PERK-eIF2α-ATF4 Pathway. Drug Des Devel Ther 2025; 19:1067-1081. [PMID: 39974610 PMCID: PMC11837746 DOI: 10.2147/dddt.s492691] [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: 09/16/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
Purpose Doxorubicin (DOX), an anthracycline antibiotic, has limited clinical use due to its pronounced cardiotoxicity. Irisin, a myokine known for its metabolic regulation, has shown therapeutic effects on cardiovascular disease. This study investigates the potential cardioprotective function of irisin in reducing the cardiac injury induced by DOX. Methods In vitro, H9c2 cells were pretreated with irisin (20 nM) for 24 hours before exposure to DOX (1 μM). In vivo, C57BL/6 mice were administered DOX (5 mg/kg/week, i.p.) for 4 weeks, reaching a cumulative dose of 20 mg/kg. Irisin (1 mg/kg/ 3 days, i.p.) was administered to the mice both 7 days prior to and during DOX injection.Cardiac function was evaluated by echocardiography, and cardiac histology was assessed using HE, WGA, and Masson staining. Myocardial injury markers were quantified using ELISA, and apoptosis was analyzed via TUNEL staining. Oxidative stress was determined by measuring antioxidase activity, MDA and GSH levels, and DHE staining, while mitochondrial superoxide production was assessed using MitoSOX Red. Mitochondrial morphology and function evaluated using transmission electron microscopy and Seahorse analysis, respectively Inflammatory cytokines were quantified in serum and cell supernatants. The role of the PERK-eIF2α-ATF4 pathway mediated by irisin was investigated by Western blot. Using adeno-associated virus serotype-9 carrying mouse FNDC5 shRNA (AAV9-shFNDC5) further validated the protective role of irisin in DOX-induced myocardial injury. Results Irisin reduced DOX-induced cardiac dysfunction and fibrosis. Moreover, irisin mitigated oxidative stress and inflammation through inhibiting the PERK-eIF2α-ATF4 pathway activated by DOX, thus preserving mitochondrial function. While cardiac FNDC5 knockdown exacerbated DOX-induced heart injury and PERK-eIF2α-ATF4 activation, which was partially reversed by irisin. Conclusion Irisin mitigates oxidative stress and inflammation by modulating the PERK-eIF2α-ATF4 pathway, highlighting its potential as a prospective approach for combating DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Zilong Zhang
- Department of Cardiology, Cardiac and Pan - Vascular Medicine Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, People’s Republic of China
| | - Xiaolin Yu
- Department of Cardiology, Cardiac and Pan - Vascular Medicine Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, People’s Republic of China
| | - Jie Li
- Department of Cardiology, Cardiac and Pan - Vascular Medicine Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, People’s Republic of China
| | - Xin Shen
- Department of Cardiology, Cardiac and Pan - Vascular Medicine Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, People’s Republic of China
| | - Wenbo Fu
- Department of Cardiology, Cardiac and Pan - Vascular Medicine Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, People’s Republic of China
| | - Yongguo Liu
- Department of Cardiology, Cardiac and Pan - Vascular Medicine Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, People’s Republic of China
| | - Xiangyu Dong
- Department of Cardiology, Cardiac and Pan - Vascular Medicine Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, People’s Republic of China
| | - Zhao Wang
- Department of Cardiology, Cardiac and Pan - Vascular Medicine Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, People’s Republic of China
| |
Collapse
|
10
|
Li W, Li Y, Xiao L, Xie Z, Peng J, Huang W, Li X, Meng Y. Micheliolide attenuates sepsis-induced acute lung injury by suppressing mitochondrial oxidative stress and PFKFB3-driven glycolysis. J Transl Med 2025; 23:181. [PMID: 39953547 PMCID: PMC11829335 DOI: 10.1186/s12967-024-05906-0] [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: 08/02/2024] [Accepted: 11/19/2024] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Sepsis is a potentially fatal condition with a significant risk of death. Acute lung injury (ALI) is a life-threatening complication of sepsis, and the inflammatory response plays a critical role in sepsis-induced ALI. The protective effects of micheliolide (MCL) against renal fibrosis and leukemia have been demonstrated, but the precise underlying mechanisms remain unclear. METHODS In vitro, lipopolysaccharides (LPS) and interferon-gamma (IFN-γ) were used to stimulate RAW264.7 cells and bone marrow-derived macrophages (BMDMs) to investigate the protective effect of MCL on sepsis-induced ALI. Cecal ligation and puncture (CLP) models were constructed in mice to induce ALI in vivo. The expression of inflammatory factors, macrophage polarization markers, and the glycolysis-related enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) were measured in vivo. Mitochondrial function, oxidative stress, and mitochondrial-related proteins were evaluated in vitro. RESULTS MCL inhibited CLP-induced ALI, as evidenced by improvements in proinflammatory factor levels, lung wet/dry ratios, and histopathological findings. In vitro, MCL treatment significantly suppressed LPS + IFN-γ-induced M1-type polarization of RAW264.7 cells and BMDMs, as well as the production of inflammatory factors and oxidative stress. Mechanistic experiments revealed that MCL suppresses PFKFB3-driven glycolysis to reduce inflammation and activates the mitochondrial unfolded protein response (UPRmt) to alleviate mitochondrial stress. However, the therapeutic effect of MCL was diminished when PFKFB3 was overexpressed in cells. CONCLUSION This study is the first to demonstrate that MCL attenuates sepsis-induced ALI by reducing M1-type macrophage polarization. Its therapeutic effect is closely related to the suppression of oxidative stress and PFKFB3-driven glycolysis.
Collapse
Affiliation(s)
- Wenhan Li
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Yuhan Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Linjie Xiao
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Zhanzhan Xie
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Jun Peng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China
| | - Wenhui Huang
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Xu Li
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| | - Ying Meng
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| |
Collapse
|
11
|
Liu H, Xu C, Hu Q, Wang Y. Sepsis-induced cardiomyopathy: understanding pathophysiology and clinical implications. Arch Toxicol 2025; 99:467-480. [PMID: 39601874 DOI: 10.1007/s00204-024-03916-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
Sepsis is a life-threatening form of organ dysfunction resulting from a dysregulated response to infection. The complex pathogenesis of sepsis poses challenges because of the lack of reliable biomarkers for early identification and effective treatments. As sepsis progresses to severe forms, cardiac dysfunction becomes a major concern, often manifesting as ventricular dilation, a reduced ejection fraction, and a diminished contractile capacity, known as sepsis-induced cardiomyopathy (SIC). The absence of standardized diagnostic and treatment protocols for SIC leads to varied criteria being used across medical institutions and studies, resulting in significant outcome disparities. Despite the high prevalence of SIC, accurate statistical data are lacking. To understand how SIC affects sepsis prognosis, a thorough exploration of its pathophysiological mechanisms, including systemic factors and complex signalling within myocardial and immune cells, is required. Identifying the factors influencing SIC occurrence and progression is crucial and must be conducted within specific clinical contexts. In this review, the clinical manifestations, pathophysiological mechanisms, and treatment strategies for SIC are discussed, along with the clinical background. We aim to connect current practices with future research challenges, providing clear guidance for clinicians and researchers.
Collapse
Affiliation(s)
- Haoran Liu
- Emergency and Trauma College, Hainan Medical University, Haikou, People's Republic of China
| | - Chaoqun Xu
- School of Medicine, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, People's Republic of China
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, People's Republic of China
| | - Qin Hu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China
| | - Yang Wang
- Emergency Medicine Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, People's Republic of China.
| |
Collapse
|
12
|
Wang X, Ma C, Mi K, Cao X, Tan Y, Yuan H, Ren J, Liang X. Urolithin A attenuates Doxorubicin-induced cardiotoxicity by enhancing PINK1-regulated mitophagy via Ambra1. Chem Biol Interact 2025; 406:111363. [PMID: 39725191 DOI: 10.1016/j.cbi.2024.111363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/02/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Doxorubicin (Dox) is a widely used antineoplastics although its clinical usage is greatly limited by its cardiotoxicity. Several studies have depicted an essential role for dampened mitophagy and mitochondrial injury in Dox cardiotoxicity. However, preventative measure to alleviate Dox-evoked cardiotoxicity via targeting mitophagy and mitochondrial integrity remains elusive. Urolithin A (UA) is a newly identified mitophagy inducer with antioxidant and anti-apoptotic properties although its effect on Dox-induced cardiotoxicity is unknown. This study was designed to explore the effect of UA on Dox cardiotoxicity and mechanisms involved. Our results indicated that UA alleviated Dox-induced cardiac dysfunction exhibited by echocardiographic parameters and histological analyses, and partially relieved Dox-induced apoptosis in vitro and in vivo, and mitochondrial dysfunction including ΔΨm dissipation and ROS production in vitro. The ability of UA to facilitate restoration of mitophagy in mice and H9C2s underscored its advantageous effects, manifested as upregulation of mitophagy-related proteins, including p62, LC3, PINK1 and Parkin, as well as the co-location between LC3 and mitochondria. Incubation with 3 -MA nearly reversed the UA-evoked rise of mitophagy-related proteins, and inhibition of apoptosis. Given that knockdown of Ambra1 almost abolished UA-induced protective effect, the enhanced expression of Ambra1 owing to UA increased PINK1 levels by inhibiting its degradation via LONP1. Collectively, our results suggest that the cardioprotective properties of UA depend on the stimulation of PINK1-dependent mitophagy through promoting Ambra1 expression to inhibit PINK1 degradation by LONP1. This highlights UA's potential as a valuable treatment option and its importance in cardioprotective strategies against Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Chao Ma
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Keying Mi
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Xinran Cao
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Yinghua Tan
- Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China
| | - Haitao Yuan
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China.
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| | - Xinyue Liang
- Department of Cardiology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China; Department of Cardiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; JiNan Key Laboratory of Cardiovascular Disease, Jinan, China.
| |
Collapse
|
13
|
Wang Y, Weng L, Wu X, Du B. The role of programmed cell death in organ dysfunction induced by opportunistic pathogens. Crit Care 2025; 29:43. [PMID: 39856779 PMCID: PMC11761187 DOI: 10.1186/s13054-025-05278-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Sepsis is a life-threatening condition resulting from pathogen infection and characterized by organ dysfunction. Programmed cell death (PCD) during sepsis has been associated with the development of multiple organ dysfunction syndrome (MODS), impacting various physiological systems including respiratory, cardiovascular, renal, neurological, hematological, hepatic, and intestinal systems. It is well-established that pathogen infections lead to immune dysregulation, which subsequently contributes to MODS in sepsis. However, recent evidence suggests that sepsis-related opportunistic pathogens can directly induce organ failure by promoting PCD in parenchymal cells of each affected organ. This study provides an overview of PCD in damaged organ and the induction of PCD in host parenchymal cells by opportunistic pathogens, proposing innovative strategies for preventing organ failure in sepsis.
Collapse
Affiliation(s)
- Yangyanqiu Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Medical ICU, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Li Weng
- State Key Laboratory of Complex Severe and Rare Diseases, Medical ICU, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xunyao Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Clinical and Science Investigation Institute, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| | - Bin Du
- State Key Laboratory of Complex Severe and Rare Diseases, Medical ICU, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, 100730, China.
- State Key Laboratory of Complex Severe and Rare Diseases, Clinical and Science Investigation Institute, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
14
|
Alfei S, Zuccari G. Ellagic Acid: A Green Multi-Target Weapon That Reduces Oxidative Stress and Inflammation to Prevent and Improve the Condition of Alzheimer's Disease. Int J Mol Sci 2025; 26:844. [PMID: 39859559 PMCID: PMC11766176 DOI: 10.3390/ijms26020844] [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: 12/15/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Oxidative stress (OS), generated by the overrun of reactive species of oxygen and nitrogen (RONS), is the key cause of several human diseases. With inflammation, OS is responsible for the onset and development of clinical signs and the pathological hallmarks of Alzheimer's disease (AD). AD is a multifactorial chronic neurodegenerative syndrome indicated by a form of progressive dementia associated with aging. While one-target drugs only soften its symptoms while generating drug resistance, multi-target polyphenols from fruits and vegetables, such as ellagitannins (ETs), ellagic acid (EA), and urolithins (UROs), having potent antioxidant and radical scavenging effects capable of counteracting OS, could be new green options to treat human degenerative diseases, thus representing hopeful alternatives and/or adjuvants to one-target drugs to ameliorate AD. Unfortunately, in vivo ETs are not absorbed, while providing mainly ellagic acid (EA), which, due to its trivial water-solubility and first-pass effect, metabolizes in the intestine to yield UROs, or irreversible binding to cellular DNA and proteins, which have very low bioavailability, thus failing as a therapeutic in vivo. Currently, only UROs have confirmed the beneficial effect demonstrated in vitro by reaching tissues to the extent necessary for therapeutic outcomes. Unfortunately, upon the administration of food rich in ETs or ETs and EA, URO formation is affected by extreme interindividual variability that renders them unreliable as novel clinically usable drugs. Significant attention has therefore been paid specifically to multitarget EA, which is incessantly investigated as such or nanotechnologically manipulated to be a potential "lead compound" with protective action toward AD. An overview of the multi-factorial and multi-target aspects that characterize AD and polyphenol activity, respectively, as well as the traditional and/or innovative clinical treatments available to treat AD, constitutes the opening of this work. Upon focus on the pathophysiology of OS and on EA's chemical features and mechanisms leading to its antioxidant activity, an all-around updated analysis of the current EA-rich foods and EA involvement in the field of AD is provided. The possible clinical usage of EA to treat AD is discussed, reporting results of its applications in vitro, in vivo, and during clinical trials. A critical view of the need for more extensive use of the most rapid diagnostic methods to detect AD from its early symptoms is also included in this work.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano, 4, 16148 Genova, Italy
| | - Guendalina Zuccari
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano, 4, 16148 Genova, Italy
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genoa, Italy
| |
Collapse
|
15
|
Liu Y, Ouyang J, Zhang C, Niu P, Shang B, Yao G, Shi Y, Zou X. Dexmedetomidine improves clinical outcomes in sepsis-induced myocardial injury: a retrospective cohort study. Front Pharmacol 2025; 15:1529167. [PMID: 39881865 PMCID: PMC11774859 DOI: 10.3389/fphar.2024.1529167] [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: 11/16/2024] [Accepted: 12/31/2024] [Indexed: 01/31/2025] Open
Abstract
Background The efficacy of dexmedetomidine (DEX) in treating sepsis-induced myocardial injury (SIMI) remains unclear. In this study, we explored the relationship between DEX use and clinical outcomes of patients with SIMI, focusing on the dosage and treatment duration. Methods In this retrospective cohort analysis, we identified patients with SIMI from the Medical Information Mart for Intensive Care IV (MIMIC-IV) database and categorized them into the DEX and non-DEX groups based on intensive care unit treatment. The baseline bias was reduced through propensity score matching (PSM). The primary outcome was 28-day mortality, whereas the secondary outcomes were in-hospital mortality and mortality rates at 7 days, 90 days, and 1 year. The association between DEX use and in-hospital mortality was assessed using Kaplan-Meier analysis and Cox proportional hazards models. Results After PSM, 373 patients in the DEX group were matched with 579 patients in the non-DEX group to achieve a balanced distribution of the covariates. The Cox regression model demonstrated a significant reduction in the 28-day mortality associated with DEX use, yielding a hazard ratio (HR) of 0.61 (95% confidence interval [CI]: 0.47-0.78, P < 0.001). In-hospital mortality also significantly decreased (HR = 0.43, 95% CI: 0.33-0.57, P < 0.001). Lower mortality rates were observed at 7 days, 90 days, and 1 year. DEX doses >0.4 μg/kg/h, particularly in the range of 0.400-0.612 μg/kg/h, total doses >3.113 mg during hospitalization, and treatment durations exceeding 72 h were associated with improved mortality risk at all intervals. Regarding DEX efficacy at 28 days, our subgroup analyses indicated a significant interaction between the Sequential Organ Failure Assessment score and invasive mechanical ventilation. Conclusion DEX administration was associated with improved in-hospital mortality and reduced mortality rates at 7 days, 28 days, 90 days, and 1 year in patients with SIMI. These findings require validation in future studies.
Collapse
Affiliation(s)
- Yuan Liu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianjie Ouyang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Cuicui Zhang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Pingping Niu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Baoling Shang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Gengzhen Yao
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Yongyong Shi
- Department of Anesthesiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guizhou Hospital, Guiyang, China
| | - Xu Zou
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Cardiology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Mao K, Lin F, Pan Y, Lu Z, Luo B, Zhu Y, Fang J, Ye J. Identification of mitophagy-related gene signatures for predicting delayed graft function and renal allograft loss post-kidney transplantation. Transpl Immunol 2024; 87:102148. [PMID: 39549926 DOI: 10.1016/j.trim.2024.102148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/10/2024] [Accepted: 11/10/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Ischemia-reperfusion injury (IRI) is an unavoidable consequence post-kidney transplantation, which inevitably leads to kidney damage. Numerous studies have demonstrated that mitophagy is implicated in human cancers. However, the function of mitophagy in kidney transplantation remains poorly understood. This study aims to develop mitophagy-related gene (MRGs) signatures to predict delayed graft function (DGF) and renal allograft loss post-kidney transplantation. METHODS Differentially expressed genes (DEGs) were identified and intersected with the MRGs to obtain mitophagy-related DEGs (MRDEGs). Functional enrichment analyses were conducted. Subsequently, random forest and SVM-RFE machine learning were employed to identify hub genes. The DGF diagnostic prediction signature was constructed using LASSO regression analysis. The renal allograft prognostic prediction signature was developed through univariate Cox and LASSO regression analysis. In addition, ROC curves, immunological characterization, correlation analysis, and survival analysis were performed. RESULTS Nineteen MRDEGs were obtained by intersecting 61 DEGs with 4897 MRGs. Seven hub genes were then identified through machine learning. Subsequently, a five-gene DGF diagnostic prediction signature was established, with ROC curves indicating its high diagnostic value for DGF. Immune infiltration analysis revealed that many immune cells were more abundant in the DGF group compared to the Immediate Graft Function (IGF) group. A two-gene prognostic signature was developed, which accurately predicted renal allografts prognosis. CONCLUSIONS The mitophagy-related gene signatures demonstrated high predictive accuracy for DGF and renal allograft loss. Our study may provide new perspectives on prognosis and treatment strategies post-kidney transplantation.
Collapse
Affiliation(s)
- Kaifeng Mao
- Department of Kidney Transplantation, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China; Division of Urology, Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen City, Guangdong Province, China
| | - Fenwang Lin
- Department of Kidney Transplantation, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China
| | - Yige Pan
- Division of Nephrology, Department of Nursing, The University of Hong Kong-Shenzhen Hospital, Shenzhen City, Guangdong Province, China
| | - Zhenquan Lu
- Division of Urology, Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen City, Guangdong Province, China
| | - Bingfeng Luo
- Division of Urology, Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen City, Guangdong Province, China
| | - Yifei Zhu
- Division of Urology, Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen City, Guangdong Province, China
| | - Jiaqi Fang
- Division of Urology, Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen City, Guangdong Province, China
| | - Junsheng Ye
- Department of Kidney Transplantation, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
| |
Collapse
|
17
|
Li X, Sun H, Zhang L, Liang H, Zhang B, Yang J, Peng X, Sun J, Zhou Y, Zhai M, Jiang L, Zhu H, Duan W. GDF15 attenuates sepsis-induced myocardial dysfunction by inhibiting cardiomyocytes ferroptosis via the SOCS1/GPX4 signaling pathway. Eur J Pharmacol 2024; 982:176894. [PMID: 39147013 DOI: 10.1016/j.ejphar.2024.176894] [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: 03/11/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Sepsis is a systemic inflammatory response syndrome triggered by infection, presenting with symptoms such as fever, increased heart rate, and low blood pressure. In severe cases, it can lead to multiple organ dysfunction, posing a life-threatening risk. Sepsis-induced cardiomyopathy (SIC) is a critical factor in the poor prognosis of septic patients, leading to myocardial dysfunction characterized by cell death, inflammation, and diminished cardiac function. Ferroptosis, an iron-dependent form of programmed cell death, is a key mechanism causing cardiomyocyte damage in SIC. Growth differentiation factor 15 (GDF15), a member of the TGF-β superfamily, is associated with various cardiovascular diseases and can inhibit oxidative stress, reduce reactive oxygen species (ROS), and suppress ferroptosis. Elevated serum GDF15 levels in sepsis are correlated with organ injuries, suggesting its potential as a therapeutic target. However, its role and mechanisms in SIC remain unclear. Glutathione peroxidase 4 (GPX4), the only enzyme capable of reducing lipid peroxides within cells, protects cells by reducing lipid peroxidation levels and inhibiting ferroptosis. Investigating the regulatory factors of GPX4 may provide a theoretical basis for SIC treatment. In this study, a mouse SIC model revealed that elevated GDF15 exerts a protective effect. Antagonizing GDF15 exacerbates myocardial damage. Through transcriptomic analysis and other methods, we confirmed that GDF15 inhibits the expression of SOCS1 by activating the ALK5-SMAD2/3 pathway, thereby activates the JAK2/STAT3 pathway, promotes the transcription of GPX4, inhibits ferroptosis in cardiomyocytes, and plays a myocardial protective role in SIC.
Collapse
Affiliation(s)
- Xiayun Li
- College of Life Sciences, Northwest University, Xi'an, 710069, China; Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - He Sun
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Liyun Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Hongliang Liang
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA, 94305, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, 94305, USA
| | - Bin Zhang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China; Department of Surgery, The 954th Hospital of the Chinese People's Liberation Army, Shannan, 856100, China
| | - Jiachang Yang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Xiangyan Peng
- School of Medicine, Northwest University, Xi'an, 710069, China
| | - Jingwei Sun
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Yang Zhou
- College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Mengen Zhai
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China
| | - Liqing Jiang
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| | - Hanzhao Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| | - Weixun Duan
- Department of Cardiovascular Surgery, Xijing Hospital, The Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
18
|
Wu Y, Yang Y, Qin X, Zhang Z, Ullah M, Li Y, Zhang Z. Unfolded proteins in the mitochondria activate HRI and inhibit mitochondrial protein translation. Cell Signal 2024; 123:111353. [PMID: 39168261 DOI: 10.1016/j.cellsig.2024.111353] [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: 03/22/2024] [Revised: 08/04/2024] [Accepted: 08/17/2024] [Indexed: 08/23/2024]
Abstract
The mitochondrial unfolded protein response (UPRmt) is triggered through eIF2α phosphorylation in mammals. However, the mechanisms of UPRmt activation and the influence of eIF2α phosphorylation on mitochondrial protein translation remain unclear. In this study, we confirmed that the UPRmt is a rapid and specific stress response that occurs through pharmacological induction of eIF2α phosphorylation, along with the phosphorylation of eIF2α, ATF4, and CHOP. Moreover, with the upregulation of the expression of some chaperones, cytochrome P450 enzymes, and DDIT4, as determined by RNA-Seq and ribosome profiling, eIF2α phosphorylation was found to be essential for the expression of ATF4 and CHOP, after which ATF4 trafficked into the nucleus and initiated CHOP expression. In addition, the generation of ROS and mitochondrial morphology were not affected by the GTPP-induced UPRmt. Furthermore, we investigated the mechanism by which HRI kinase-mediated UPRmt is induced by mitochondrial unfolded proteins via CRISPR-Cas9 technology, mitochondrial recruitment of HRI and interaction with other proteins. Moreover, we confirmed that mitochondrial protein translation and mitochondrial protein import were inhibited through eIF2α phosphorylation with the accumulation of unfolded mitochondrial proteins. These findings reveal the molecular mechanism of the UPRmt and its impact on cellular protein translation, which will offer novel insights into the functions of the UPRmt, including its implications for human disease and pathobiology.
Collapse
Affiliation(s)
- Yongshu Wu
- College of Animal Science and Technology College of Veterinary Medicine/Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province/Zhejiang Provincial Engineering Laboratory for Animal Health Inspection and Internet Technology/Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management/China-Australia Joint Laboratory for Animal Health Big Data Analytics, Zhejiang A&F University, Hangzhou 311300, China
| | - Yang Yang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China
| | - Xiaodong Qin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China
| | - Zhixiong Zhang
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China
| | - Munib Ullah
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou 730046, Gansu, China
| | - Yanmin Li
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan 610041, China.
| | - Zhidong Zhang
- College of Animal Science and Veterinary Medicine, Southwest Minzu University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
19
|
Xiong X, Hou J, Zheng Y, Jiang T, Zhao X, Cai J, Huang J, He H, Xu J, Qian S, Lu Y, Wang X, Wang W, Ye Q, Zhou S, Lian M, Xiao J, Song W, Xie C. NAD +-boosting agent nicotinamide mononucleotide potently improves mitochondria stress response in Alzheimer's disease via ATF4-dependent mitochondrial UPR. Cell Death Dis 2024; 15:744. [PMID: 39394148 PMCID: PMC11470026 DOI: 10.1038/s41419-024-07062-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/01/2024] [Accepted: 09/06/2024] [Indexed: 10/13/2024]
Abstract
Extensive studies indicate that mitochondria dysfunction is pivotal for Alzheimer's disease (AD) pathogenesis; while cumulative evidence suggests that increased mitochondrial stress response (MSR) may mitigate neurodegeneration in AD, explorations to develop a MSR-targeted therapeutic strategy against AD are scarce. We combined cell biology, molecular biology, and pharmacological approaches to unravel a novel molecular pathway by which NAD+-boosting agent nicotinamide mononucleotide (NMN) regulates MSR in AD models. Here, we report dyshomeostasis plasma UPRmt-mitophagy-mediated MSR profiles in AD patient samples. NMN restores NAD+ metabolic profiles and improves MSR through the ATF4-dependent UPRmt pathway in AD-related cross-species models. At the organismal level, NAD+ repletion with NMN supplementation ameliorates mitochondrial proteotoxicity, decreases hippocampal synaptic disruption, decreases neuronal loss, and brain atrophy in mice model of AD. Remarkably, omics features of the hippocampus with NMN show that NMN leads to transcriptional changes of genes and proteins involved in MSR characteristics, principally within the astrocyte unit rather than microglia and oligodendrocytes. In brief, our work provides evidence that MSR has an active role in the pathogenesis of AD, as reducing mitochondrial homeostasis via atf4 depletion in AD mice aggravates the hallmarks of the disease; conversely, bolstering mitochondrial proteostasis by NMN decreases protein aggregation, restores memory performance, and delays disease progression, ultimately translating to increased healthspan.
Collapse
Affiliation(s)
- Xi Xiong
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jialong Hou
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Zheng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Tao Jiang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuemiao Zhao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinlai Cai
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiani Huang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haijun He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiaxue Xu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuangjie Qian
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yao Lu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Neurology, Yuhuan City People's Hospital, Taizhou, China
| | - XinShi Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wenwen Wang
- The Center of Traditional Chinese Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qianqian Ye
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shuoting Zhou
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengjia Lian
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, China
| | - Jian Xiao
- Oujiang Laboratory, Wenzhou, Zhejiang, China.
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China.
| | - Weihong Song
- Oujiang Laboratory, Wenzhou, Zhejiang, China.
- Key Laboratory Of Alzheimer's Disease Of Zhejiang Province, Institute Of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Chenglong Xie
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory, Wenzhou, Zhejiang, China.
- Key Laboratory Of Alzheimer's Disease Of Zhejiang Province, Institute Of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Department of Geriatrics, Geriatric Medical Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
20
|
Yin L, Yuan L, Luo Z, Tang Y, Lin X, Wang S, Liang P, Huang L, Jiang B. COX-2 optimizes cardiac mitochondrial biogenesis and exerts a cardioprotective effect during sepsis. Cytokine 2024; 182:156733. [PMID: 39128194 DOI: 10.1016/j.cyto.2024.156733] [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: 06/26/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Septic cardiomyopathy is a component of multiple organ dysfunction in sepsis. Mitochondrial dysfunction plays an important role in septic cardiomyopathy. Studies have shown that cyclooxygenase-2 (COX-2) had a protective effect on the heart, and prostaglandin E2 (PGE2), the downstream product of COX-2, was increasingly recognized to have a protective effect on mitochondrial function. OBJECTIVE This study aims to demonstrate that COX-2/PGE2 can protect against septic cardiomyopathy by regulating mitochondrial function. METHODS Cecal ligation and puncture (CLP) was used to establish a mouse model of sepsis and RAW264.7 macrophages and H9C2 cells were used to simulate sepsis in vitro. The NS-398 and celecoxib were used to inhibit the activity of COX-2. ZLN005 and SR18292 were used to activate or inhibit the PGC-1α activity. The mitochondrial biogenesis was examined through the Mitotracker Red probe, mtDNA copy number, and ATP content detection. RESULTS The experimental data suggested that COX-2 inhibition attenuated PGC-1α expression thus decreasing mitochondrial biogenesis, whereas increased PGE2 could promote mitochondrial biogenesis by activating PGC-1α. The results also showed that the effect of COX-2/PGE2 on PGC-1α was mediated by the activation of cyclic adenosine monophosphate (cAMP) response element binding protein (CREB). Finally, the effect of COX-2/PGE2 on the heart was also verified in the septic mice. CONCLUSION Collectively, these results suggested that COX-2/PGE2 pathway played a cardioprotective role in septic cardiomyopathy through improving mitochondrial biogenesis, which has changed the previous understanding that COX-2/PGE2 only acted as an inflammatory factor.
Collapse
Affiliation(s)
- Leijing Yin
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, PR China; Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - Ludong Yuan
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, PR China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan Province, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan Province, PR China
| | - Zhengyang Luo
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, PR China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan Province, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan Province, PR China
| | - Yuting Tang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, PR China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan Province, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan Province, PR China
| | - Xiaofang Lin
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, PR China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan Province, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan Province, PR China
| | - Shuxin Wang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, PR China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan Province, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan Province, PR China
| | - Pengfei Liang
- Department of Burns and Plastic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province, PR China
| | - Lingjin Huang
- Department of Cardiothoracic Surgery, Xiangya Hospital Central South University, Changsha, PR China.
| | - Bimei Jiang
- Department of Pathophysiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, PR China; Sepsis Translational Medicine Key Lab of Hunan Province, Central South University, Changsha, Hunan Province, PR China; National Medicine Functional Experimental Teaching Center, Central South University, Changsha, Hunan Province, PR China.
| |
Collapse
|
21
|
Chen R, Zheng A, Wang Y, Guo L, Dou H, Lu L, Rafiq M, Li P, Chen X, Xiao Q. Salvianolic acid B improves mitochondrial dysfunction of septic cardiomyopathy via enhancing ATF5-mediated mitochondrial unfolded protein response. Toxicol Appl Pharmacol 2024; 491:117072. [PMID: 39153513 DOI: 10.1016/j.taap.2024.117072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
AIMS Septic cardiomyopathy is characterized by impaired contractile function and mitochondrial activity dysregulation. Salvianolic acid B (Sal B) is a potent therapeutic compound derived from the traditional Chinese medicine Salvia miltiorrhiza. This study explored the protective effects of Sal B on septic heart injury, emphasizing the mitochondrial unfolded protein response (UPRmt). MATERIALS AND METHODS An in vivo mouse model of lipopolysaccharide (LPS)-induced heart injury was utilized to assess Sal B's protective role in septic cardiomyopathy. Additionally, cell models stimulated by LPS were developed to investigate the mechanisms of Sal B on UPRmt. Quantitative polymerase chain reaction, western blotting, immunohistochemistry, and immunofluorescence were employed for molecular analysis. RESULTS Sal B, administered at doses of 10, 30, and 60 mg/kg, demonstrated protective effects on cardiac contractile function, reduced heart inflammation, and mitigated cardiac injury in LPS-exposed mice. In cardiomyocytes, LPS induced apoptosis, elevated mitochondrial ROS levels, promoted mitochondrial fission, and decreased mitochondrial membrane potential, all of which were alleviated by Sal B. Mechanistically, Sal B was found to induce UPRmt both in vivo and in vitro. ATF5, identified as a UPRmt activator, was modulated by LPS and Sal B, resulting in increased ATF5 expression and its translocation from the cytosol to the nucleus. ATF5-siRNA delivery reversed UPRmt upregulation, exacerbating mitochondrial dysfunction in LPS-stimulated cardiomyocytes and counteracting the mitochondrial function enhancement in Sal B-treated cardiomyocytes. CONCLUSIONS This study provides evidence that Sal B confers cardiac protection by enhancing UPRmt, highlighting its potential as a therapeutic approach for mitigating mitochondrial dysfunction in septic cardiomyopathy.
Collapse
Affiliation(s)
- Renshan Chen
- Guangzhou Hospital of Integrated Traditional and Western Medicine, 87 Yingbin Avenue, Guangzhou 510800, PR China
| | - Anran Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China; Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Yunjing Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China; Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Liyou Guo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China; Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Huaqian Dou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China; Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Liangyan Lu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China; Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Muhammad Rafiq
- Department of Biosciences, COMSATS University Islamabad, Park Road, Tarlai Kalan, Islamabad 45550, Pakistan
| | - Peihua Li
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China; Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China
| | - Xiuhui Chen
- Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Department of Pharmacy & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, 68 South Xihu Third Road, Shilong Town, Dongguan 523000, China.
| | - Qing Xiao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China; Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, 1 Xinzao Road, Panyu District, Guangzhou 511436, PR China.
| |
Collapse
|
22
|
Chang X, Wu D, Gao X, Lin J, Tan Y, Wang J, Zhu H, Zhou H. BuyangHuanwu Decoction alleviates Endothelial Cell Apoptosis and Coronary Microvascular Dysfunction via Regulation of the MAPKK4/p38 Signaling Axis. Int J Med Sci 2024; 21:2464-2479. [PMID: 39439466 PMCID: PMC11492876 DOI: 10.7150/ijms.98183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 09/03/2024] [Indexed: 10/25/2024] Open
Abstract
MAPKK4 has been implicated in the pathological mechanisms underlying myocardial and vascular injury, specifically influencing endothelial cell damage and programmed cell death via subcellular pathways. Nevertheless, the regulatory role of MAPKK4 in coronary microvascular injury following myocardial infarction remains unconfirmed, and the exploration of targeted mitochondrial protective therapeutic agents remains unaddressed. In light of this gap, we established a MAPKK4 gene-modified mouse model of ischemia-reperfusion injury and employed Buyang Huanwu decoction (BYHW), a traditional cardiovascular therapeutic formula, to assess its efficacy in treating coronary microvascular injury post-ischemia-reperfusion. The study aimed to elucidate the mechanism by which BYHW mitigates coronary microvascular injury induced by ischemia-reperfusion through the attenuation of endothelial cell apoptosis. Experimental outcomes revealed that high-dose BYHW significantly ameliorated coronary microvascular injury post-ischemia-reperfusion, restoring the structural integrity of the coronary microvasculature and reducing inflammation and oxidative stress. Contrarily, in transgenic mice overexpressing MAPKK4, BYHW intervention failed to attenuate microvascular inflammation and oxidative stress. To further investigate, we simulated hypoxia/reoxygenation injury in vascular endothelial cells using a MAPKK4-related cellular gene modification model. The results indicated that BYHW attenuates inflammatory damage and enhances the viability of vascular endothelial cells following hypoxic stress, inhibiting apoptosis via the mitochondrial pathway. However, overexpression of MAPKK4/p38 negated the therapeutic effects of BYHW, showing no impact on endothelial cell apoptosis and oxidative stress under hypoxic conditions. Molecular interaction studies confirmed that the active components of BYHW, Astragaloside IV and Ligustrazine, interact with the MAPKK4/P38 axis. In vitro experiments further suggested that the interaction between MAPKK4 and P38 play a crucial role in the ability of BYHW to inhibit apoptosis in coronary microvascular endothelial cells. Therapeutically, MAPKK4 may potentiate the apoptotic pathway in microvascular endothelial cells by modulating downstream P38 expression and phosphorylation, thereby exacerbating ischemia-reperfusion-induced coronary microvascular endothelial injury. From an in vivo perspective, the transgenic overexpression of MAPKK4 and P38 inhibited the microvascular protective effects of BYHW. These findings collectively underscore the significance of the MAPKK4-P38 axis in the protection of coronary microvascular endothelial cells.
Collapse
Affiliation(s)
- Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Dan Wu
- Division of Vascular Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510800, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Disease, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xin Gao
- Outpatient Department of the Sixth Medical Center of the PLA General Hospital, China
| | - Jianguo Lin
- The First Affliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Tan
- Beijing University of Chinese Medicine, Beijing, 100028, China
| | - Junyan Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China
| | - Hang Zhu
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
23
|
Hu D, Sheeja Prabhakaran H, Zhang YY, Luo G, He W, Liou YC. Mitochondrial dysfunction in sepsis: mechanisms and therapeutic perspectives. Crit Care 2024; 28:292. [PMID: 39227925 PMCID: PMC11373266 DOI: 10.1186/s13054-024-05069-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: 05/14/2024] [Accepted: 08/17/2024] [Indexed: 09/05/2024] Open
Abstract
Sepsis is a severe medical condition characterized by a systemic inflammatory response, often culminating in multiple organ dysfunction and high mortality rates. In recent years, there has been a growing recognition of the pivotal role played by mitochondrial damage in driving the progression of sepsis. Various factors contribute to mitochondrial impairment during sepsis, encompassing mechanisms such as reactive nitrogen/oxygen species generation, mitophagy inhibition, mitochondrial dynamics change, and mitochondrial membrane permeabilization. Damaged mitochondria actively participate in shaping the inflammatory milieu by triggering key signaling pathways, including those mediated by Toll-like receptors, NOD-like receptors, and cyclic GMP-AMP synthase. Consequently, there has been a surge of interest in developing therapeutic strategies targeting mitochondria to mitigate septic pathogenesis. This review aims to delve into the intricate mechanisms underpinning mitochondrial dysfunction during sepsis and its significant impact on immune dysregulation. Moreover, we spotlight promising mitochondria-targeted interventions that have demonstrated therapeutic efficacy in preclinical sepsis models.
Collapse
Affiliation(s)
- Dongxue Hu
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Harshini Sheeja Prabhakaran
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Yuan-Yuan Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China.
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 119077, Singapore.
| |
Collapse
|
24
|
Pu X, Zhang Q, Liu J, Wang Y, Guan X, Wu Q, Liu Z, Liu R, Chang X. Ginsenoside Rb1 ameliorates heart failure through DUSP-1-TMBIM-6-mediated mitochondrial quality control and gut flora interactions. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155880. [PMID: 39053246 DOI: 10.1016/j.phymed.2024.155880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/04/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND There is currently no specific therapeutic drug available for heart failure in clinical practice. Numerous studies have validated the efficacy of Ginsenoside Rb1, an active component found in various herbal remedies used for heart failure treatment, in effectively ameliorating myocardial ischemia. However, the precise mechanism of action and molecular targets of Ginsenoside Rb1 remain unclear. PURPOSE This study aims to explore the molecular mechanisms through which Ginsenoside Rb1 synergistically modulates the gut flora and mitochondrial quality control network in heart failure by targeting the DUSP-1-TMBIM-6-VDAC1 axis. STUDY DESIGN This study utilized DUSP-1/VDAC1 knockout (DUSP-1-/-/VDAC1-/-) and DUSP-1/VDAC1 transgenic (DUSP-1+/+/VDAC1+/+) mouse models of heart failure, established through Transverse Aortic Constriction (TAC) surgery and genetic modification techniques. The mice were subsequently subjected to treatment with Ginsenoside Rb1. METHODS A series of follow-up multi-omics analyses were conducted, including assessments of intestinal flora, gene transcription sequencing, single-cell databases, and molecular biology assays of primary cardiomyocytes, to investigate the mechanism of action of Ginsenoside Rb1. RESULTS Ginsenoside Rb1 was found to have multiple regulatory mechanisms on mitochondria. Notably, DUSP-1 was discovered to be a crucial molecular target of Ginsenoside Rb1, controlling both intestinal flora and mitochondrial function. The regulatory effects of DUSP-1 on inflammation and mitochondrial quality control were mediated by changes in TMBIM-6 and VDAC1. Furthermore, NLRP3-mediated inflammatory responses were found to interact with mitochondrial quality control, exacerbating myocardial injury under stress conditions. Ginsenoside Rb1 modulated the DUSP-1-TMBIM-6-VDAC1 axis, inhibited the release of pro-inflammatory factors, altered the structural composition of the gut flora, and protected impaired heart function. These effects indirectly influenced the crosstalk between inflammation, mitochondria, and gut flora. CONCLUSION The DUSP-1-TMBIM-6-VDAC1 axis, an upstream pathway regulated by Ginsenoside Rb1, is a profound mechanism through which Ginsenoside Rb1 improves cardiac function in heart failure by modulating inflammation, mitochondria, and gut flora.
Collapse
Affiliation(s)
- Xiangyi Pu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Qin Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jinfeng Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yanli Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xuanke Guan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Qiaomin Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Zhiming Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Ruxiu Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
25
|
Han D, Su T, Wang M, Zhang R, Xu H, Chu R, Zhu Z, Shen Y, Wang N, He S, Wang Y, Han Y, Wang Q. JAK2 inhibitor protects the septic heart through enhancing mitophagy in cardiomyocytes. Biomed Pharmacother 2024; 178:117279. [PMID: 39121587 DOI: 10.1016/j.biopha.2024.117279] [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: 03/31/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024] Open
Abstract
Sepsis-induced myocardial dysfunction (SIMD) is a severe complication in sepsis, manifested as myocardial systolic dysfunction, which is associated with poor prognosis and higher mortality. Mitophagy, a self-protective mechanism maintaining cellular homeostasis, plays an indispensable role in cardioprotection. This study aimed to unveil the cardioprotective effects of Baricitinib on LPS-induced myocardial dysfunction and its effect on mitophagy. Herein, we demonstrated that LPS induced severe myocardial dysfunction and initiated mitophagy in septic mice hearts. Despite the initiation of mitophagy, a significant number of apoptotic cells and damaged mitochondria persisted in the myocardium, and myocardial energy metabolism remained impaired, indicating that the limited mitophagy was insufficient to mitigate LPS-induced damage. The JAK2-AKT-mTOR signaling pathway is activated in LPS-induced cardiomyocytes and in the hearts of septic mice. Baricitinib administration remarkably improved cardiac function, suppressed systemic inflammatory response, attenuated histopathological changes, inhibited cardiac cell apoptosis and alleviated myocardial damage in septic mice. Furthermore, Baricitinib treatment significantly enhanced PINK1-Parkin-mediated mitophagy, increased autophagosomes, decreased impaired mitochondria, and restored myocardial energy metabolism. Mechanically, the limited mitophagy in septic myocardium was associated with increased p-ULK1 (Ser757), which was regulated by p-mTOR. Baricitinib reduced p-ULK1 (Ser757) and enhanced mitophagy by inhibiting the JAK2-AKT-mTOR signaling pathway. Inhibition of mitophagy with Mdivi-1 reversed the cardiac protective and anti-inflammatory effects of Baricitinib in septic mice. These findings suggest that Baricitinib attenuates SIMD by enhancing mitophagy in cardiomyocytes via the JAK2-AKT-mTOR signaling pathway, providing a novel mechanistic and therapeutic insight into the SIMD.
Collapse
Affiliation(s)
- Dafei Han
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Tiantian Su
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Mingzhu Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Renhao Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Huihui Xu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Rui Chu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Zhenduo Zhu
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China
| | - Yawei Shen
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Nan Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Shufang He
- Department of Anesthesiology and Perioperative Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| | - Yongsheng Wang
- Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, China.
| | - Yongsheng Han
- Department of Emergency Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qingtong Wang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, China; Department of Cardiology, The Third Affiliated Hospital of Anhui Medical University (The First People's Hospital of Hefei), Hefei, China.
| |
Collapse
|
26
|
Cui Y, Zhang Y, Dai S, Wan S, Guan H, Wang D, Jin B, Xiao W, Liu F. The mechanism of 14-3-3η in thyroxine induced mitophagy in cardiomyocytes. Mol Cell Endocrinol 2024; 590:112271. [PMID: 38759835 DOI: 10.1016/j.mce.2024.112271] [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: 07/02/2023] [Revised: 04/24/2024] [Accepted: 05/14/2024] [Indexed: 05/19/2024]
Abstract
Hyperthyroidism is becoming increasingly important as an independent risk factor for cardiovascular disease, eventually resulting in cardiac hypertrophy and heart failure. The 14-3-3 protein family subtypes regulate many cellular processes in eukaryotes by interacting with a diverse array of client proteins. Considering that the 14-3-3η protein protects cardiomyocytes by affecting mitochondrial function, exploring the biological influence and molecular mechanisms by which 14-3-3η alleviates the cardiac hypertrophy of hyperthyroidism is imperative. In vivo and in vitro, RT-PCR, Western blot, and Mitochondrial tracking assay were performed to understand the molecular mechanism of thyroxine-induced cardiomyocyte hypertrophy. HE staining, transmission electron microscopy, and immunofluorescence were used to observe intuitively changes of hearts and cardiomyocytes. The in vivo and in vitro results indicated that overexpression of the 14-3-3η ameliorated thyroxine-induced cardiomyocyte hypertrophy, whereas knockdown of the 14-3-3η protein aggravated thyroxine-induced cardiomyocyte hypertrophy. Additionally, overexpression of the 14-3-3η protein reduces thyroxine-induced mitochondrial damage and mitophagy in cardiomyocytes. Overexpression of 14-3-3η protein improves excessive mitophagy in the myocardium caused by thyroxine and thus prevents cardiac hypertrophy.
Collapse
Affiliation(s)
- Yalan Cui
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi, 541004, China; Clinical Pathology Department, The Second People's Hospital of China Three Gorges University, Yichang, Hubei, 443600, China
| | - Yan Zhang
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Songsong Dai
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Sha Wan
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Heng Guan
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Decai Wang
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Site of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Beifang Jin
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Wenping Xiao
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi, 541004, China
| | - Fang Liu
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi, 541004, China; Center of Diabetic Systems Medicine, Guangxi Key Laboratory of Excellence, Guilin Medical University, Guilin, Guangxi, 541004, China.
| |
Collapse
|
27
|
Chang X, Zhou S, Liu J, Wang Y, Guan X, Wu Q, Liu Z, Liu R. Zishenhuoxue decoction-induced myocardial protection against ischemic injury through TMBIM6-VDAC1-mediated regulation of calcium homeostasis and mitochondrial quality surveillance. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155331. [PMID: 38870748 DOI: 10.1016/j.phymed.2023.155331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 12/07/2023] [Accepted: 12/30/2023] [Indexed: 06/15/2024]
Abstract
BACKGROUND Zishenhuoxue decoction (ZSHX), a Chinese herbal medicine, exhibits myocardial and vascular endothelial protective properties. The intricate regulatory mechanisms underlying myocardial ischemic injury and its association with dysfunctional mitochondrial quality surveillance (MQS) remain elusive. HYPOTHESIS/PURPOSE To study the protective effect of ZSHX on ischemic myocardial injury in mice using a TMBIM6 gene-modified animal model and mitochondrial quality control-related experiments. STUDY DESIGN Using model animals and myocardial infarction surgery-induced ischemic myocardial injury TMBIM6 gene-modified mouse models, the pharmacological activity of ZSHX in inhibiting ischemic myocardial injury and mitochondrial homeostasis disorder in vivo was tested. METHODS Our focal point entailed scrutinizing the impact of ZSHX on ischemic myocardial impairment through the prism of TMBIM6. This endeavor was undertaken utilizing mice characterized by heart-specific TMBIM6 knockout (TMBIM6CKO) and their counterparts, the TMBIM6 transgenic (TMBIM6TG) and VDAC1 transgenic (VDAC1TG) mice. RESULTS ZSHX demonstrated dose-dependent effectiveness in mitigating ischemic myocardial injury and enhancing mitochondrial integrity. TMBIM6CKO hindered ZSHX's cardio-therapeutic and mitochondrial protective effects, while ZSHX's benefits persisted in TMBIM6TG mice. TMBIM6CKO also blocked ZSHX's regulation of mitochondrial function in HR-treated cardiomyocytes. Hypoxia disrupted the MQS in cardiomyocytes, including calcium overload, excessive fission, mitophagy issues, and disrupted biosynthesis. ZSHX counteracted these effects, thereby normalizing MQS and inhibiting calcium overload and cardiomyocyte necroptosis. Our results also showed that hypoxia-induced TMBIM6 blockade resulted in the over-activation of VDAC1, a major mitochondrial calcium uptake pathway, while ZSHX could increase the expression of TMBIM6 and inhibit VDAC1-mediated calcium overload and MQS abnormalities. CONCLUSIONS Our findings suggest that ZSHX regulates mitochondrial calcium homeostasis and MQS abnormalities through a TMBIM6-VDAC1 interaction mechanism, which helps to treat ischemic myocardial injury and provides myocardial protection. This study also offers insights for the clinical translation and application of mitochondrial-targeted drugs in cardiomyocytess.
Collapse
Affiliation(s)
- Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange, Xicheng District, Beijing 100053, China
| | - Siyuan Zhou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange, Xicheng District, Beijing 100053, China
| | - Jinfeng Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange, Xicheng District, Beijing 100053, China
| | - Yanli Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange, Xicheng District, Beijing 100053, China
| | - Xuanke Guan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange, Xicheng District, Beijing 100053, China
| | - Qiaomin Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange, Xicheng District, Beijing 100053, China
| | - Zhiming Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange, Xicheng District, Beijing 100053, China.
| | - Ruxiu Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange, Xicheng District, Beijing 100053, China.
| |
Collapse
|
28
|
Hong WL, Huang H, Zeng X, Duan CY. Targeting mitochondrial quality control: new therapeutic strategies for major diseases. Mil Med Res 2024; 11:59. [PMID: 39164792 PMCID: PMC11337860 DOI: 10.1186/s40779-024-00556-1] [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: 09/09/2023] [Accepted: 07/13/2024] [Indexed: 08/22/2024] Open
Abstract
Mitochondria play a crucial role in maintaining the normal physiological state of cells. Hence, ensuring mitochondrial quality control is imperative for the prevention and treatment of numerous diseases. Previous reviews on this topic have however been inconsistencies and lack of systematic organization. Therefore, this review aims to provide a comprehensive and systematic overview of mitochondrial quality control and explore the possibility of targeting the same for the treatment of major diseases. This review systematically summarizes three fundamental characteristics of mitochondrial quality control, including mitochondrial morphology and dynamics, function and metabolism, and protein expression and regulation. It also extensively examines how imbalances in mitochondrial quality are linked to major diseases, such as ischemia-hypoxia, inflammatory disorders, viral infections, metabolic dysregulations, degenerative conditions, and tumors. Additionally, the review explores innovative approaches to target mitochondrial quality control, including using small molecule drugs that regulate critical steps in maintaining mitochondrial quality, nanomolecular materials designed for precise targeting of mitochondria, and novel cellular therapies, such as vesicle therapy and mitochondrial transplantation. This review offers a novel perspective on comprehending the shared mechanisms underlying the occurrence and progression of major diseases and provides theoretical support and practical guidance for the clinical implementation of innovative therapeutic strategies that target mitochondrial quality control for treating major diseases.
Collapse
Affiliation(s)
- Wei-Long Hong
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - He Huang
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Xue Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Chen-Yang Duan
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
29
|
Dong X, Zhuang HW, Wen RJ, Huang YS, Liang BX, Li H, Xian SX, Li C, Wang LJ, Wang JY. Xinyang tablet alleviated cardiac dysfunction in a cardiac pressure overload model by regulating the receptor-interacting serum/three-protein kinase 3/FUN14 domain containing 1-mediated mitochondrial unfolded protein response and mitophagy. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118152. [PMID: 38614260 DOI: 10.1016/j.jep.2024.118152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xinyang tablet (XYT) has been used for heart failure (HF) for over twenty years in clinical practice, but the underlying molecular mechanism remains poorly understood. AIMS OF THE STUDY In the present study, we aimed to explore the protective effects of XYT in HF in vivo and in vitro. MATERIALS AND METHODS Transverse aortic constriction was performed in vivo to establish a mouse model of cardiac pressure overload. Echocardiography, tissue staining, and real-time quantitative PCR (qPCR) were examined to evaluate the protective effects of XYT on cardiac function and structure. Adenosine 5'-triphosphate production, reactive oxygen species staining, and measurement of malondialdehyde and superoxide dismutase was used to detect mitochondrial damage. Mitochondrial ultrastructure was observed by transmission electron microscope. Immunofluorescence staining, qPCR, and Western blotting were performed to evaluate the effect of XYT on the mitochondrial unfolded protein response and mitophagy, and to identify its potential pharmacological mechanism. In vitro, HL-1 cells and neonatal mouse cardiomyocytes were stimulated with Angiotensin II to establish the cell model. Western blotting, qPCR, immunofluorescence staining, and flow cytometry were utilized to determine the effects of XYT on cardiomyocytes. HL-1 cells overexpressing receptor-interacting serum/three-protein kinase 3 (RIPK3) were generated by transfection of RIPK3-overexpressing lentiviral vectors. Cells were then co-treated with XYT to determine the molecular mechanisms. RESULTS In the present study, XYT was found to exerta protective effect on cardiac function and structure in the pressure overload mice. And it was also found XYT reduced mitochondrial damage by enhancing mitochondrial unfolded protein response and restoring mitophagy. Further studies showed that XYT achieved its cardioprotective role through regulating the RIPK3/FUN14 domain containing 1 (FUNDC1) signaling. Moreover, the overexpression of RIPK3 successfully reversed the XYT-induced protective effects and significantly attenuated the positive effects on the mitochondrial unfolded protein response and mitophagy. CONCLUSIONS Our findings indicated that XYT prevented pressure overload-induced HF through regulating the RIPK3/FUNDC1-mediated mitochondrial unfolded protein response and mitophagy. The information gained from this study provides a potential strategy for attenuating mitochondrial damage in the context of pressure overload-induced heart failure using XYT.
Collapse
Affiliation(s)
- Xin Dong
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Hao-Wen Zhuang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Rui-Jia Wen
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yu-Sheng Huang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bing-Xue Liang
- Chongqing College of Traditional Chinese Medicine, Chongqing, 400000, China
| | - Huan Li
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Shao-Xiang Xian
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Chun Li
- Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Ling-Jun Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| | - Jun-Yan Wang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China; Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Chronic Heart Failure, Guangzhou, 510405, China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
30
|
Alghusen IM, Carman MS, Wilkins HM, Strope TA, Gimore C, Fedosyuk H, Shawa J, Ephrame SJ, Denson AR, Wang X, Swerdlow RH, Slawson C. O-GlcNAc impacts mitophagy via the PINK1-dependent pathway. Front Aging Neurosci 2024; 16:1387931. [PMID: 39175808 PMCID: PMC11339348 DOI: 10.3389/fnagi.2024.1387931] [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: 02/18/2024] [Accepted: 07/11/2024] [Indexed: 08/24/2024] Open
Abstract
Background The accumulation of dysfunctional mitochondria is an early feature of Alzheimer's disease (AD). The impaired turnover of damaged mitochondria increases reactive oxygen species production and lowers ATP generation, leading to cellular toxicity and neurodegeneration. Interestingly, AD exhibits a disruption in the global post-translational modification β-N-acetylglucosamine (O-GlcNAc). O-GlcNAc is a ubiquitous single sugar modification found in the nuclear, cytoplasmic, and mitochondrial proteins. Cells maintain a homeostatic level of O-GlcNAc by cycling the addition and removal of the sugar by O-GlcNAc transferase (OGT) or O-GlcNAcase (OGA), respectively. Methods We used patient-derived induced pluripotent stem cells, a transgenic mouse model of AD, SH-SY5Y neuroblastoma cell lines to examine the effect of sustained O-GlcNAcase inhibition by Thiamet-G (TMG) or OGT deficiency on mitophagy using biochemical analyses. Results Here, we established an essential role for O-GlcNAc in regulating mitophagy (mitochondria-selective autophagy). Stimulating mitophagy using urolithin A (UA) decreases cellular O-GlcNAc and elevates mitochondrial O-GlcNAc. Sustained elevation in O-GlcNAcylation via pharmacologically inhibiting OGA using Thiamet-G (TMG) increases the mitochondrial level of mitophagy protein PTEN-induced kinase 1 (PINK1) and autophagy-related protein light chain 3 (LC3). Moreover, we detected O-GlcNAc on PINK1 and TMG increases its O-GlcNAcylation level. Conversely, decreasing cellular O-GlcNAcylation by knocking down OGT decreases both PINK1 protein expression and LC3 protein expression. Mitochondria isolated from CAMKII-OGT-KO mice also had decreased PINK1 and LC3. Moreover, human brain organoids treated with TMG showed significant elevation in LC3 compared to control. However, TMG-treated AD organoids showed no changes in LC3 expression. Conclusion Collectively, these data demonstrate that O-GlcNAc plays a crucial role in the activation and progression of mitophagy, and this activation is disrupted in AD.
Collapse
Affiliation(s)
- Ibtihal M. Alghusen
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Marisa S. Carman
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Heather M. Wilkins
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Taylor A. Strope
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Caleb Gimore
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Halyna Fedosyuk
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Jad Shawa
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sophiya John Ephrame
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Aspin R. Denson
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Xiaowan Wang
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Russell H. Swerdlow
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| | - Chad Slawson
- School of Medicine, Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, United States
- University of Kansas Alzheimer’s Disease Research Center, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
31
|
Deng J, Wang D, Shi Y, Lin L, Gao W, Sun Y, Song X, Li Y, Li J. Mitochondrial unfolded protein response mechanism and its cardiovascular protective effects. Biomed Pharmacother 2024; 177:116989. [PMID: 38959609 DOI: 10.1016/j.biopha.2024.116989] [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: 01/16/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 07/05/2024] Open
Abstract
The mitochondrial unfolded protein response (UPRmt) is a cytoprotective response in response to cellular stress that is activated in response to mitochondrial stress to maintain intra-protein homeostasis, thereby protecting the cell from a variety of stimuli. The activation of this response has been linked to cardiovascular diseases. Here, we reviewed the current understanding of UPRmt and discussed its specific molecular mechanism, mainly in mammals, as well as addressing its protective role against cardiovascular diseases, so as to provide direction for further research on UPRmt and therapies targeting cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Jinlan Deng
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Danyang Wang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanmei Shi
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Lin
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Weihan Gao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiayinan Song
- Chinese University of Traditional Chinese Medicine,Beijing University of Chinese Medicine, Chaoyang, China
| | - Yunlun Li
- Department of Cardiology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie Li
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
32
|
Li J, Zhang D, Zhang Y, Ge J, Yang C. Mitochondria-specific antioxidant MitoTEMPO alleviates senescence of bone marrow mesenchymal stem cells in ovariectomized rats. J Cell Physiol 2024; 239:e31323. [PMID: 38801103 DOI: 10.1002/jcp.31323] [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: 12/20/2023] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
Senescence in bone marrow mesenchymal stem cells (BMSCs), triggered by excessive oxidative stress, plays a crucial role in the onset of postmenopausal osteoporosis. Recent studies underscore the importance of mitochondrial rehabilitation and quality control as key determinants in the modulation of oxidative stress and cellular senescence. MitoTEMPO, a mitochondria-targeted antioxidant, has been shown to mitigate the heightened levels of reactive oxygen species (ROS). In our research, we observed that BMSCs from ovariectomized (OVX) rats displayed premature senescence, which was attributed to combined mitochondrial and lysosomal dysfunction, a condition that worsens with extended estrogen deprivation. Treatment with MitoTEMPO effectively reversed these effects, reinstating lysosomal functionality and suppressing the mitochondrial unfolded protein response (UPRmt). Subsequent in vivo experiments corroborated these observations, revealing that MitoTEMPO administration in OVX rats curtailed trabecular bone loss and reduced the expression of p53, HSP60, and CLPP in the trabecular bone region of the proximal tibia. Overall, our findings suggest that MitoTEMPO holds promise as a therapeutic agent to counteract senescence in OVX-BMSCs, offering a potential strategy for treating postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Dahe Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuxin Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jing Ge
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chi Yang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
He X, Wu Z, Jiang J, Xu W, Yuan A, Liao F, Ding S, Pu J. Urolithin A Protects against Hypoxia-Induced Pulmonary Hypertension by Inhibiting Pulmonary Arterial Smooth Muscle Cell Pyroptosis via AMPK/NF-κB/NLRP3 Signaling. Int J Mol Sci 2024; 25:8246. [PMID: 39125817 PMCID: PMC11311380 DOI: 10.3390/ijms25158246] [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: 05/29/2024] [Revised: 07/16/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Recent studies confirmed that pyroptosis is involved in the progression of pulmonary hypertension (PH), which could promote pulmonary artery remodeling. Urolithin A (UA), an intestinal flora metabolite of ellagitannins (ETs) and ellagic acid (EA), has been proven to possess inhibitory effects on pyroptosis under various pathological conditions. However, its role on PH remained undetermined. To investigate the potential of UA in mitigating PH, mice were exposed to hypoxia (10% oxygen, 4 weeks) to induce PH, with or without UA treatment. Moreover, in vitro experiments were carried out to further uncover the underlying mechanisms. The in vivo treatment of UA suppressed the progression of PH via alleviating pulmonary remodeling. Pyroptosis-related genes were markedly upregulated in mice models of PH and reversed after the administration of UA. In accordance with that, UA treatment significantly inhibited hypoxia-induced pulmonary arterial smooth muscle cell (PASMC) pyroptosis via the AMPK/NF-κB/NLRP3 pathway. Our results revealed that UA treatment effectively mitigated PH progression through inhibiting PASMC pyroptosis, which represents an innovative therapeutic approach for PH.
Collapse
Affiliation(s)
- Xinjie He
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (X.H.); (Z.W.); (J.J.); (W.X.); (A.Y.); (F.L.)
| | - Zhinan Wu
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (X.H.); (Z.W.); (J.J.); (W.X.); (A.Y.); (F.L.)
| | - Jinyao Jiang
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (X.H.); (Z.W.); (J.J.); (W.X.); (A.Y.); (F.L.)
| | - Wenyi Xu
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (X.H.); (Z.W.); (J.J.); (W.X.); (A.Y.); (F.L.)
| | - Ancai Yuan
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (X.H.); (Z.W.); (J.J.); (W.X.); (A.Y.); (F.L.)
| | - Fei Liao
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (X.H.); (Z.W.); (J.J.); (W.X.); (A.Y.); (F.L.)
| | - Song Ding
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (X.H.); (Z.W.); (J.J.); (W.X.); (A.Y.); (F.L.)
- Department of Cardiology, Punan Branch of Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Jun Pu
- Department of Cardiology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China; (X.H.); (Z.W.); (J.J.); (W.X.); (A.Y.); (F.L.)
| |
Collapse
|
34
|
L’Abbate S, Kusmic C. The Protective Effect of Flavonoids in the Diet on Autophagy-Related Cardiac Impairment. Nutrients 2024; 16:2207. [PMID: 39064651 PMCID: PMC11279826 DOI: 10.3390/nu16142207] [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: 06/25/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
The compounds known as flavonoids, commonly found in fruits, vegetables, legumes, medicinal herbs, chocolate, and coffee and tea beverages, have been extensively researched for their impact on cardiovascular health. Flavonoids, with their demonstrated potential, have shown promising effects in regulating blood vessel function and apoptotic processes, as well as in improving lipid profiles. While their powerful antioxidant properties were initially thought to be the main reason behind these effects, recent studies have uncovered new insights into the positive effects of flavonoids on cardiovascular health, and researchers have now identified several signaling pathways and mechanisms that also play a role. Of particular interest are the studies that have highlighted the role of autophagy in maintaining the physiological functions of cardiomyocytes and protecting them from harm. Recent publications have linked the dysregulation of autophagic processes with the development of cardiomyopathies, heart failure, and other cardiovascular diseases. This review aims to present the latest, novel findings from preclinical research regarding the potential beneficial effects of flavonoids on various heart conditions associated with altered autophagy processes.
Collapse
Affiliation(s)
| | - Claudia Kusmic
- Istituto di Fisiologia Clinica, Consiglio Nazionale delle Ricerche (CNR), 56124 Pisa, Italy;
| |
Collapse
|
35
|
Pei H, Qu J, Chen J, Zhao G, Lu Z. S100A9 as a Key Myocardial Injury Factor Interacting with ATP5 Exacerbates Mitochondrial Dysfunction and Oxidative Stress in Sepsis-Induced Cardiomyopathy. J Inflamm Res 2024; 17:4483-4503. [PMID: 39006491 PMCID: PMC11246037 DOI: 10.2147/jir.s457340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/22/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose Sepsis-induced cardiomyopathy (SICM) is a prevalent cardiac dysfunction caused by sepsis. Mitochondrial dysfunction is a crucial pathogenic factor associated with adverse cardiovascular adverse events; however, research on SICM remains insufficient. Methods To investigate the factors contributing to the pathological progression of SICM, we performed a comprehensive analysis of transcriptomic data from the GEO database using bioinformatics and machine learning techniques. CRISPR-Cas9 S100A9 knockout mice and primary cardiomyocytes were exposed to lipopolysaccharide to simulate SICM. Transcriptome analysis and mass spectrometry of primary cardiomyocytes were used to determine the potential pathogenic mechanisms of S100A9. The mitochondrial ultrastructure and mitochondrial membrane potential (MMP) were detected using transmission electron microscopy and flow cytometry, respectively. Pink1/Parkin and Drp1 proteins were detected using Western blotting to evaluate mitochondrial autophagy and division. The mtDNA and mRNA levels of mitochondrial transcription factors and synthases were evaluated using real-time polymerase chain reaction. Results Bioinformatics analysis identified 12 common differentially expressed genes, including SERPINA3N, LCN2, MS4A6D, LRG1, OSMR, SOCS3, FCGR2b, S100A9, S100A8, CASP4, ABCA8A, and NFKBIZ. Significant S100A9 upregulation was closely associated with myocardial injury exacerbation and cardiac function deterioration. GSEA revealed that myocardial contractile function, oxidative stress, and mitochondrial function were significantly affected by S100A9. Knocking out S100A9 alleviates the inflammatory response and mitochondrial dysfunction. The interaction of S100A9 with ATP5 enhanced mitochondrial division and autophagy, inhibited MMP and ATP synthesis, and induced oxidative stress, which are related to the Nlrp3-Nfkb-Caspase1 and Drp1-Pink1-Parkin signaling pathways. The expression of mitochondrial transcription factors (TFAM and TFBM) and ATP synthetases (ATP6 and ATP8, as well as COX1, COX2, and COX3) was further suppressed by S100A9 in SICM. Targeted S100A9 inhibition by paquinimod partially reversed myocardial mitochondrial dysfunction and oxidative stress. Conclusion The interaction of S100A9 with ATP5 exacerbates myocardial damage in sepsis by inducing mitochondrial dysfunction and oxidative stress.
Collapse
Affiliation(s)
- Hui Pei
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Jie Qu
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Jianming Chen
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - Guangju Zhao
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
| | - ZhongQiu Lu
- Emergency Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, People’s Republic of China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, People’s Republic of China
| |
Collapse
|
36
|
Zhang F, Ye Z, Ran Y, Liu C, Zhang M, Xu X, Song F, Yao L. Ruthenium red alleviates post-resuscitation myocardial dysfunction by upregulating mitophagy through inhibition of USP33 in a cardiac arrest rat model. Eur J Pharmacol 2024; 974:176633. [PMID: 38703975 DOI: 10.1016/j.ejphar.2024.176633] [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: 08/22/2023] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Cardiac arrest (CA) remains a leading cause of death, with suboptimal survival rates despite efforts involving cardiopulmonary resuscitation and advanced life-support technology. Post-resuscitation myocardial dysfunction (PRMD) is an important determinant of patient outcomes. Myocardial ischemia/reperfusion injury underlies this dysfunction. Previous reports have shown that ruthenium red (RR) has a protective effect against cardiac ischemia-reperfusion injury; however, its precise mechanism of action in PRMD remains unclear. This study investigated the effects of RR on PRMD and analyzed its underlying mechanisms. Ventricular fibrillation was induced in rats, which were then subjected to cardiopulmonary resuscitation to establish an experimental CA model. At the onset of return of spontaneous circulation, RR (2.5 mg/kg) was administered intraperitoneally. Our study showed that RR improved myocardial function and reduced the production of oxidative stress markers such as malondialdehyde (MDA), glutathione peroxidase (GSSG), and reactive oxygen species (ROS) production. RR also helped maintain mitochondrial structure and increased ATP and GTP levels. Additionally, RR effectively attenuated myocardial apoptosis. Furthermore, we observed downregulation of proteins closely related to mitophagy, including ubiquitin-specific protease 33 (USP33) and P62, whereas LC3B (microtubule-associated protein light chain 3B) was upregulated. The upregulation of mitophagy may play a critical role in reducing myocardial injury. These results demonstrate that RR may attenuate PRMD by promoting mitophagy through the inhibition of USP33. These effects are likely mediated through diverse mechanisms, including antioxidant activity, apoptosis suppression, and preservation of mitochondrial integrity and energy metabolism. Consequently, RR has emerged as a promising therapeutic approach for addressing post-resuscitation myocardial dysfunction.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China; Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, 510000, China
| | - Zhou Ye
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China; Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, 510000, China
| | - Yingqi Ran
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Cong Liu
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Mingtao Zhang
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Xiangchang Xu
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China
| | - Fengqing Song
- Department of Emergency Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China; Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-sen University, Guangzhou, 510000, China.
| | - Lan Yao
- Department of Emergency Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, 519000, China.
| |
Collapse
|
37
|
Du Y, Li J, Dai Z, Chen Y, Zhao Y, Liu X, Xia T, Zhu P, Wang Y. Pyruvate kinase M2 sustains cardiac mitochondrial quality surveillance in septic cardiomyopathy by regulating prohibitin 2 abundance via S91 phosphorylation. Cell Mol Life Sci 2024; 81:254. [PMID: 38856931 PMCID: PMC11335292 DOI: 10.1007/s00018-024-05253-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 04/11/2024] [Accepted: 04/20/2024] [Indexed: 06/11/2024]
Abstract
The endogenous mitochondrial quality control (MQC) system serves to protect mitochondria against cellular stressors. Although mitochondrial dysfunction contributes to cardiac damage during many pathological conditions, the regulatory signals influencing MQC disruption during septic cardiomyopathy (SC) remain unclear. This study aimed to investigate the involvement of pyruvate kinase M2 (PKM2) and prohibitin 2 (PHB2) interaction followed by MQC impairment in the pathogenesis of SC. We utilized LPS-induced SC models in PKM2 transgenic (PKM2TG) mice, PHB2S91D-knockin mice, and PKM2-overexpressing HL-1 cardiomyocytes. After LPS-induced SC, cardiac PKM2 expression was significantly downregulated in wild-type mice, whereas PKM2 overexpression in vivo sustained heart function, suppressed myocardial inflammation, and attenuated cardiomyocyte death. PKM2 overexpression relieved sepsis-related mitochondrial damage via MQC normalization, evidenced by balanced mitochondrial fission/fusion, activated mitophagy, restored mitochondrial biogenesis, and inhibited mitochondrial unfolded protein response. Docking simulations, co-IP, and domain deletion mutant protein transfection experiments showed that PKM2 phosphorylates PHB2 at Ser91, preventing LPS-mediated PHB2 degradation. Additionally, the A domain of PKM2 and the PHB domain of PHB2 are required for PKM2-PHB2 binding and PHB2 phosphorylation. After LPS exposure, expression of a phosphorylation-defective PHB2S91A mutant negated the protective effects of PKM2 overexpression. Moreover, knockin mice expressing a phosphorylation-mimetic PHB2S91D mutant showed improved heart function, reduced inflammation, and preserved mitochondrial function following sepsis induction. Abundant PKM2 expression is a prerequisite to sustain PKM2-PHB2 interaction which is a key element for preservation of PHB2 phosphorylation and MQC, presenting novel interventive targets for the treatment of septic cardiomyopathy.
Collapse
Affiliation(s)
- Yingzhen Du
- The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China
| | - Jialei Li
- School of Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhe Dai
- School of Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuxin Chen
- School of Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yao Zhao
- School of Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoman Liu
- School of Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Tian Xia
- Department of Clinical Laboratory Medicine, The First Medical Centre, Medical School of Chinese People's Liberation Army, Beijing, China
- Xianning Medical College, Hubei University of Science & Technology, Xianning, China
| | - Pingjun Zhu
- The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China.
| | - Yijin Wang
- The Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, China.
| |
Collapse
|
38
|
Wu J, Yang Y, Lin D, Wang Z, Ma J. SIRT3 and RORα are two prospective targets against mitophagy during simulated ischemia/reperfusion injury in H9c2 cells. Heliyon 2024; 10:e30568. [PMID: 38784556 PMCID: PMC11112282 DOI: 10.1016/j.heliyon.2024.e30568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Autophagy during myocardial ischemia/reperfusion (MI/R) exacerbates cardiomyocyte injury. Melatonin (Mel) alleviates myocardial damage by regulating mitochondrial function and mitophagy, but the role of mitophagy in melatonin-induced cardioprotection remains unclear. This study aimed to explore the roles of sirtuin3 (SIRT3) and retinoid-related orphan nuclear receptor-α (RORα) in mitophagy during simulated ischemia reperfusion (SIR) in H9c2 cells. Our data showed that mitophagy was excessively activated after SIR injury, which was consistent with reduced cell survival, enhanced oxidative responses and mitochondrial dysfunction in H9c2 myocytes. Melatonin greatly enhanced cell viability, reduced oxidative stress and improved mitochondrial function. The effects of melatonin protection were involved in excessive mitophagy inhibition, as demonstrated by the reduced levels of mitophagy-linked proteins, including Parkin, Beclin1, NIX and BNIP3, and the LC3 II/LC3 I ratio and elevations in p62. Additionally, the decreases in SIRT3 and RORα in H9c2 myocytes after SIR were reversed by melatonin, and the above effects of melatonin were eliminated by small interfering RNA (siRNA)-mediated knockdown of SIRT3 and RORα. In brief, SIRT3 and RORα are two prospective targets in the cardioprotection of melatonin against mitophagy during SIR in H9c2 myocytes.
Collapse
Affiliation(s)
- Jinjing Wu
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Yanli Yang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Duomao Lin
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Zhaoqi Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| | - Jun Ma
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University-Beijing Institute of Heart Lung and Blood Vessel Diseases, China
| |
Collapse
|
39
|
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
|
40
|
Lv Y, Yu Z, Zhang P, Zhang X, Li H, Liang T, Guo Y, Cheng L, Peng F. The structure and function of FUN14 domain-containing protein 1 and its contribution to cardioprotection by mediating mitophagy. Front Pharmacol 2024; 15:1389953. [PMID: 38828457 PMCID: PMC11140143 DOI: 10.3389/fphar.2024.1389953] [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: 02/22/2024] [Accepted: 04/29/2024] [Indexed: 06/05/2024] Open
Abstract
Cardiovascular disease (CVD) is a serious public health risk, and prevention and treatment efforts are urgently needed. Effective preventive and therapeutic programs for cardiovascular disease are still lacking, as the causes of CVD are varied and may be the result of a multifactorial combination. Mitophagy is a form of cell-selective autophagy, and there is increasing evidence that mitophagy is involved in cardioprotective processes. Recently, many studies have shown that FUN14 domain-containing protein 1 (FUNDC1) levels and phosphorylation status are highly associated with many diseases, including heart disease. Here, we review the structure and functions of FUNDC1 and the path-ways of its mediated mitophagy, and show that mitophagy can be effectively activated by dephosphorylation of Ser13 and Tyr18 sites, phosphorylation of Ser17 site and ubiquitination of Lys119 site in FUNDC1. By effectively activating or inhibiting excessive mitophagy, the quality of mitochondria can be effectively controlled. The main reason is that, on the one hand, improper clearance of mitochondria and accumulation of damaged mitochondria are avoided, and on the other hand, excessive mitophagy causing apoptosis is avoided, both serving to protect the heart. In addition, we explore the possible mechanisms by which FUNDC1-mediated mitophagy is involved in exercise preconditioning (EP) for cardioprotection. Finally, we also point out unresolved issues in FUNDC1 and its mediated mitophagy and give directions where further research may be needed.
Collapse
Affiliation(s)
- Yuhu Lv
- College of Physical Education, Guangdong University of Education, Guangzhou, China
- Research Center for Adolescent Sports and Health Promotion of Guangdong Province, Guangzhou, China
| | - Zhengze Yu
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Peiwen Zhang
- College of Nursing and Rehabilitation, Xi an FanYi University, Xi’an, China
| | - Xiqian Zhang
- College of Physical Education, Guangdong University of Education, Guangzhou, China
- Research Center for Adolescent Sports and Health Promotion of Guangdong Province, Guangzhou, China
| | - Huarui Li
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Ting Liang
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Yanju Guo
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Lin Cheng
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| | - Fenglin Peng
- College of Physical Education and Health, Guangxi Normal University, Guilin, China
| |
Collapse
|
41
|
Fang X, Zhang Y, Wu H, Wang H, Miao R, Wei J, Zhang Y, Tian J, Tong X. Mitochondrial regulation of diabetic endothelial dysfunction: Pathophysiological links. Int J Biochem Cell Biol 2024; 170:106569. [PMID: 38556159 DOI: 10.1016/j.biocel.2024.106569] [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: 12/07/2023] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Micro- and macrovascular complications frequently occur in patients with diabetes, with endothelial dysfunction playing a key role in the development and progression of the complications. For the early diagnosis and optimal treatment of vascular complications associated with diabetes, it is imperative to comprehend the cellular and molecular mechanisms governing the function of diabetic endothelial cells. Mitochondria function as crucial sensors of environmental and cellular stress regulating endothelial cell viability, structural integrity and function. Impaired mitochondrial quality control mechanisms and mitochondrial dysfunction are the main features of endothelial damage. Hence, targeted mitochondrial therapy is considered promising novel therapeutic options in vascular complications of diabetes. In this review, we focus on the mitochondrial functions in the vascular endothelial cells and the pathophysiological role of mitochondria in diabetic endothelial dysfunction, aiming to provide a reference for related drug development and clinical diagnosis and treatment.
Collapse
Affiliation(s)
- Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Haoran Wu
- Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Han Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate College, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiahua Wei
- Graduate College, Changchun University of Chinese Medicine, Jilin 130117, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Xiaolin Tong
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
42
|
Zhou QY, Ren C, Li JY, Wang L, Duan Y, Yao RQ, Tian YP, Yao YM. The crosstalk between mitochondrial quality control and metal-dependent cell death. Cell Death Dis 2024; 15:299. [PMID: 38678018 PMCID: PMC11055915 DOI: 10.1038/s41419-024-06691-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: 10/02/2023] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/29/2024]
Abstract
Mitochondria are the centers of energy and material metabolism, and they also serve as the storage and dispatch hubs of metal ions. Damage to mitochondrial structure and function can cause abnormal levels and distribution of metal ions, leading to cell dysfunction and even death. For a long time, mitochondrial quality control pathways such as mitochondrial dynamics and mitophagy have been considered to inhibit metal-induced cell death. However, with the discovery of new metal-dependent cell death including ferroptosis and cuproptosis, increasing evidence shows that there is a complex relationship between mitochondrial quality control and metal-dependent cell death. This article reviews the latest research results and mechanisms of crosstalk between mitochondrial quality control and metal-dependent cell death in recent years, as well as their involvement in neurodegenerative diseases, tumors and other diseases, in order to provide new ideas for the research and treatment of related diseases.
Collapse
Affiliation(s)
- Qi-Yuan Zhou
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Chao Ren
- Department of Pulmonary and Critical Care Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, China
| | - Jing-Yan Li
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Lu Wang
- Department of Critical Care Medicine, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu Duan
- Department of Critical Care Medicine, Affiliated Chenzhou Hospital (the First People's Hospital of Chenzhou), Southern Medical University, Chenzhou, 423000, China
| | - Ren-Qi Yao
- Department of General Surgery, the First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| | - Ying-Ping Tian
- Department of Emergency, the Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Yong-Ming Yao
- Medical Innovation Research Division, Translational Medicine Research Center and the Fourth Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
43
|
Chen Y, Cao W, Li B, Qiao X, Wang X, Yang G, Li S. The potential role of hydrogen sulfide in regulating macrophage phenotypic changes via PINK1/parkin-mediated mitophagy in sepsis-related cardiorenal syndrome. Immunopharmacol Immunotoxicol 2024; 46:139-151. [PMID: 37971696 DOI: 10.1080/08923973.2023.2281901] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE Sepsis is one of major reasons of cardiorenal syndrome type 5 (CRS-5), resulting in irreversible tissue damage and organ dysfunction. Macrophage has been demonstrated to play key role in the pathophysiology of sepsis, highlighting the need to identify therapeutic targets for modulating macrophage phenotype in sepsis. METHODS AND RESULTS In this study, a rapid-releasing hydrogen sulfide (H2S) donor NaSH, and a slow-releasing H2S compound S-propargyl-cysteine (SPRC) which is derived from garlic, have been studied for the immune-regulatory effects on macrophages. The NaSH and SPRC showed the potential to protect the heart and kidney from tissue injury induced by LPS. The immunohistochemistry of F4/80+ revealed that the infiltration of macrophages in the heart and kidney tissues of LPS-treated mice was reduced by NaSH and SPRC. In addition, in the LPS-triggered inflammatory cascade of RAW264.7 macrophage cells, NaSH and SPRC exhibited significantly inhibitory effects on the secretion of inflammatory cytokines, production of reactive oxygen species (ROS), and regulation of the macrophage phenotype from M1-like to M2-like. Moreover, autophagy, a crucial process involved in the elimination of impaired proteins and organelles during oxidative stress and immune response, was induced by NaSH and SPRC in the presence of LPS stimulation. Consequently, there was an increase in the number of mitochondria and an improvement in mitochondrial membrane potential. This process was mainly mediated by PINK1/Parkin pathway mediated mitophagy. DISCUSSION These results demonstrated that the immunoregulatory effects of H2S donors were through the PINK1/Parkin-mediated mitophagy pathway. Overall, our study provided a new therapeutic direction in LPS-induced cardiorenal injury.
Collapse
Affiliation(s)
- Yuxuan Chen
- Department of Cell Biology, Shandong University, Jinan, China
- Shandong Institute of Endocrinology and Metabolic Diseases, Shandong First Medical University, Jinan, China
- Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Wei Cao
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Bin Li
- Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaofei Qiao
- Department of Cell Biology, Shandong University, Jinan, China
| | - Xiangdong Wang
- Department of Cell Biology, Shandong University, Jinan, China
| | - Guang Yang
- Department of Joint Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Siying Li
- Department of Cell Biology, Shandong University, Jinan, China
- Shandong Institute of Endocrinology and Metabolic Diseases, Shandong First Medical University, Jinan, China
- Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Key Laboratory of Cardiovascular Disease Proteomics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
44
|
Cao Y, Chen X, Zhu Z, Luo Z, Hao Y, Yang X, Feng J, Zhang Z, Hu J, Jian Y, Zhu J, Liang W, Chen Z. STING contributes to lipopolysaccharide-induced tubular cell inflammation and pyroptosis by activating endoplasmic reticulum stress in acute kidney injury. Cell Death Dis 2024; 15:217. [PMID: 38485717 PMCID: PMC10940292 DOI: 10.1038/s41419-024-06600-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/18/2024]
Abstract
Recently, innate immunity and inflammation were recognized as the key factors for acute kidney injury (AKI) caused by sepsis, which is closely related to high mortality. Stimulator of interferon genes (STING) has emerged as a critical component of innate immune and inflammatory responses. However, the role of STING in the pathogenesis of septic AKI remains unclear. This study demonstrated that the STING was significantly activated in tubular cells induced by lipopolysaccharide (LPS) in vivo and in vitro. Tubule-specific STING knockout attenuated LPS-induced renal dysfunction and pathological changes. Mechanistically, the STING pathway promotes NOD-like receptor protein 3 (NLRP3) activation. STING triggers endoplasmic reticulum (ER) stress to induce mitochondrial reactive oxygen species (mtROS) overproduction, enhancing thioredoxin-interacting protein activation and association with NLRP3. Eventually, the NLRP3 inflammasome leads to tubular cell inflammation and pyroptosis. This study revealed the STING-regulated network and further identified the STING/ER stress/mtROS/NLRP3 inflammasome axis as an emerging pathway contributing to tubular damage in LPS-induced AKI. Hence, targeting STING may be a promising therapeutic strategy for preventing septic AKI.
Collapse
Affiliation(s)
- Yun Cao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
- Department of Nephrology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical College), Haikou, China
| | - Xinghua Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zijing Zhu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zilv Luo
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiqun Hao
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xueyan Yang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Feng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yonghong Jian
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiefu Zhu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
45
|
Zuo RK, Wang C, Yu ZY, Shi HM, Song XK, Zhou SD, Ma NN, Chang GJ, Shen XZ. A high concentrate diet inhibits forkhead box protein A2 expression, and induces oxidative stress, mitochondrial dysfunction and mitochondrial unfolded protein response in the liver of dairy cows. Microb Pathog 2024; 188:106570. [PMID: 38341108 DOI: 10.1016/j.micpath.2024.106570] [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: 11/19/2023] [Revised: 01/11/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
High-concentrate diet induce subacute ruminal acidosis (SARA) and cause liver damage in ruminants. It has been reported that forkhead box protein A2 (FOXA2) can enhance mitochondrial membrane potential but its function in mitochondrial dysfunction induced by high concentrate diets is still unknown. Therefore, the aim of this study was to elucidate the effect of high-concentrate (HC) diet on hepatic FOXA2 expression, mitochondrial unfolded protein response (UPRmt), mitochondrial dysfunction and oxidative stress. A total of 12 healthy mid-lactation Holstein cows were selected and randomized into 2 groups: the low concentrate (LC) diet group (concentrate:forage = 4:6) and HC diet group (concentrate:forage = 6:4). The trial lasted 21 d. The rumen fluid, blood and liver tissue were collected at the end of the experiment. The results showed that the rumen fluid pH level was reduced in the HC group and the pH was lower than 5.6 for more than 4 h/d, indicating that feeding HC diets successfully induced SARA in dairy cows. Both FOXA2 mRNA and protein abundance were significantly reduced in the liver of the HC group compared with the LC group. The activity of antioxidant enzymes (CAT, G6PDH, T-SOD, Cu/Zn SOD, Mn SOD) and mtDNA copy number in the liver tissue of the HC group decreased, while the level of H2O2 significantly increased, this increase was accompanied by a decrease in oxidative phosphorylation (OXPHOS). The balance of mitochondrial division and fusion was disrupted in the HC group, as evidenced by the decreased mRNA level of OPA1, MFN1, and MFN2 and increased mRNA level of Drp1, Fis1, and MFF. At the same time, HC diet downregulated the expression level of SIRT1, SIRT3, PGC-1α, TFAM, and Nrf 1 to inhibit mitochondrial biogenesis. The HC group induced UPRmt in liver tissue by upregulating the mRNA and protein levels of CLPP, LONP1, CHOP, Hsp10, and Hsp60. In addition, HC diet could increase the protein abundance of Bax, CytoC, Caspase 3 and Cleaved-Caspase 3, while decrease the protein abundance of Bcl-2 and the Bcl-2/Bax ratio. Overall, our study suggests that the decreased expression of FOXA2 may be related to UPRmt, mitochondrial dysfunction, oxidative stress, and apoptosis in the liver of dairy cows fed a high concentrate diet.
Collapse
Affiliation(s)
- Ran Kun Zuo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Can Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Zhi Yuan Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Hui Min Shi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Xiao Kun Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Shen Dong Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Na Na Ma
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Guang Jun Chang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China
| | - Xiang Zhen Shen
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, Jiangsu, PR China.
| |
Collapse
|
46
|
Cui Y, Li Y, Meng S, Song Y, Xie K. Molecular hydrogen attenuates sepsis-induced cardiomyopathy in mice by promoting autophagy. BMC Anesthesiol 2024; 24:72. [PMID: 38395800 PMCID: PMC10885652 DOI: 10.1186/s12871-024-02462-4] [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: 09/30/2023] [Accepted: 02/18/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Approximately 40 to 60% of patients with sepsis develop sepsis-induced cardiomyopathy (SIC), which is associated with a substantial increase in mortality. We have found that molecular hydrogen (H2) inhalation improved the survival rate and cardiac injury in septic mice. However, the mechanism remains unclear. This study aimed to explore the regulatory mechanism by which hydrogen modulates autophagy and its role in hydrogen protection of SIC. METHODS Cecal ligation and puncture (CLP) was used to induce sepsis in adult C57BL/6J male mice. The mice were randomly divided into 4 groups: Sham, Sham + 2% hydrogen inhalation (H2), CLP, and CLP + H2 group. The 7-day survival rate was recorded. Myocardial pathological scores were calculated. Myocardial troponin I (cTnI) levels in serum were detected, and the levels of autophagy- and mitophagy-related proteins in myocardial tissue were measured. Another four groups of mice were also studied: CLP, CLP + Bafilomycin A1 (BafA1), CLP + H2, and CLP + H2 + BafA1 group. Mice in the BafA1 group received an intraperitoneal injection of the autophagy inhibitor BafA1 1 mg/kg 1 h after operation. The detection indicators remained the same as before. RESULTS The survival rate of septic mice treated with H2 was significantly improved, myocardial tissue inflammation was improved, serum cTnI level was decreased, autophagy flux was increased, and mitophagy protein content was decreased (P < 0.05). Compared to the CLP + H2 group, the CLP + H2 + BafA1 group showed a decrease in autophagy level and 7-day survival rate, an increase in myocardial tissue injury and cTnI level, which reversed the protective effect of hydrogen (P < 0.05). CONCLUSION Hydrogen exerts protective effect against SIC, which may be achieved through the promotion of autophagy and mitophagy.
Collapse
Affiliation(s)
- Yan Cui
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Yingning Li
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
| | - Shuqi Meng
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Yu Song
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Keliang Xie
- Department of Anesthesiology, Tianjin Institute of Anesthesiology, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin, 300052, China.
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
47
|
Ma L, Han T, Zhan YA. Mechanism and role of mitophagy in the development of severe infection. Cell Death Discov 2024; 10:88. [PMID: 38374038 PMCID: PMC10876966 DOI: 10.1038/s41420-024-01844-4] [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: 10/23/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 02/21/2024] Open
Abstract
Mitochondria produce adenosine triphosphate and potentially contribute to proinflammatory responses and cell death. Mitophagy, as a conservative phenomenon, scavenges waste mitochondria and their components in the cell. Recent studies suggest that severe infections develop alongside mitochondrial dysfunction and mitophagy abnormalities. Restoring mitophagy protects against excessive inflammation and multiple organ failure in sepsis. Here, we review the normal mitophagy process, its interaction with invading microorganisms and the immune system, and summarize the mechanism of mitophagy dysfunction during severe infection. We highlight critical role of normal mitophagy in preventing severe infection.
Collapse
Affiliation(s)
- Lixiu Ma
- Department of Respiratory and Critical Care Medicine, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Tianyu Han
- Jiangxi Institute of Respiratory Disease, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yi-An Zhan
- Department of Respiratory and Critical Care Medicine, the 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
48
|
Ma M, Zhou H, Zhang Y, Yuan W, Chen C. The DNA-dependent protein kinase catalytic subunit promotes sepsis-induced cardiac dysfunction through disrupting INF-2-dependent mitochondrial dynamics. Int J Med Sci 2024; 21:714-724. [PMID: 38464839 PMCID: PMC10920849 DOI: 10.7150/ijms.91894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 02/03/2024] [Indexed: 03/12/2024] Open
Abstract
Sepsis-induced cardiomyopathy (SIC) represents a severe complication of systemic infection, characterized by significant cardiac dysfunction. This study examines the role of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) and Inverted Formin 2 (INF2) in the pathogenesis of SIC, focusing on their impact on mitochondrial homeostasis and dynamics. Our research demonstrates that silencing DNA-PKcs alleviates lipopolysaccharide (LPS)-induced cardiomyocyte death and dysfunction. Using HL-1 cardiomyocytes treated with LPS, we observed that DNA-PKcs knockdown notably reverses LPS-induced cytotoxicity, indicating a protective role against cellular damage. This effect is further substantiated by the reduction in caspase-3 and caspase-9 activation, key markers of apoptosis, upon DNA-PKcs knockdown. Besides, our data further reveal that DNA-PKcs knockdown attenuates LPS-induced mitochondrial dysfunction, evidenced by improved ATP production, enhanced activities of mitochondrial respiratory complexes, and preserved mitochondrial membrane potential. Moreover, DNA-PKcs deletion counteracts LPS-induced shifts towards mitochondrial fission, indicating its regulatory influence on mitochondrial dynamics. Conclusively, our research elucidates the intricate interplay between DNA-PKcs and INF2 in the modulation of mitochondrial function and dynamics during sepsis-induced cardiomyopathy. These findings offer new insights into the molecular mechanisms underpinning SIC and suggest potential therapeutic targets for mitigating mitochondrial dysfunction in this critical condition.
Collapse
Affiliation(s)
- Mudi Ma
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, China
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Zhou
- Senior Department of Cardiology, The Sixth Medical Center of People's Liberation Army General Hospital, Beijing, China
| | - Ying Zhang
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, China
| | - Woliang Yuan
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, China
| | - Chaoxiong Chen
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, China
| |
Collapse
|
49
|
Chang X, Zhou S, Liu J, Wang Y, Guan X, Wu Q, Zhang Q, Liu Z, Liu R. Zishen Tongyang Huoxue decoction (TYHX) alleviates sinoatrial node cell ischemia/reperfusion injury by directing mitochondrial quality control via the VDAC1-β-tubulin signaling axis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 320:117371. [PMID: 37981118 DOI: 10.1016/j.jep.2023.117371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/22/2023] [Accepted: 10/28/2023] [Indexed: 11/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zishen Tongyang Huoxue decoction (TYHX) has been used clinically for nearly 40 years to treat sick sinus syndrome. Previous reports showed that TYHX can inhibit calcium flux by regulating mitochondrial homeostasis via β-tubulin and increase sinoatrial node cell (SNC) activity. However, the underlying mechanisms remain unclear. AIM OF THE STUDY We aimed to verify the protective effect of TYHX against SNC ischemia by regulating mitochondrial quality control (MQC) through β-tubulin and voltage-dependent anion-selective channel 1 (VDAC1) silencing. MATERIALS AND METHODS We established an in vitro model of SNC ischemia/reperfusion (I/R) injury and performed rescue experiments by silencing β-tubulin and VDAC1 expression. Cell-Counting Kit 8 assays were performed to detect cell viabilities, and terminal deoxynucleotidyl transferase dUTP nick-end labeling assays (paired with confocal microscopy) were performed to detect fragmentation. Mitochondrial-energy metabolism was detected using the Seahorse assay system. Reverse transcription-quantitative polymerase chain reaction analysis was performed to detect the mRNA-expression levels of MQC-related genes. RESULTS TYHX inhibited SNC mitochondrial injury. During I/R simulation, TYHX maintained β-tubulin stability, regulated synergy between mitophagy and the mitochondrial unfolded-protein response (UPRmt), and inhibited mitochondrial oxidative stress and overactive SNC fission. Next-generation sequencing suggested that mitochondrial-membrane injury caused SNC apoptosis. We also found that TYHX regulated β-tubulin expression through VDAC1 and inhibited dynamin-related protein 1 migration to mitochondria from the nucleus. After preventing excessive mitochondrial fission, the mitophagy-UPRmt pathway, mitochondrial-membrane potential, and mitochondrial energy were restored. VDAC1 silencing affected the regulatory mechanism of MQC in a β-tubulin-dependent manner via TYHX. CONCLUSION TYHX regulated mitochondrial membrane-permeability through VDAC1, which affected MQC through β-tubulin and inhibited mitochondrial apoptosis. Our findings may help in developing drugs to protect the sinoatrial node.
Collapse
Affiliation(s)
- Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Siyuan Zhou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Jinfeng Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Yanli Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Xuanke Guan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Qiaomin Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Qin Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Zhiming Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| | - Ruxiu Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
50
|
Li W, Lin M, Li J, Ding Q, Chen X, Chen H, Shen Z, Zhu X. Xijiao Dihuang Decoction Protects Against Murine Sepsis-Induced Cardiac Inflammation and Apoptosis via Suppressing TLR4/NF-κB and Activating PI3K/AKT Pathway. J Inflamm Res 2024; 17:853-863. [PMID: 38348278 PMCID: PMC10860816 DOI: 10.2147/jir.s428305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/01/2024] [Indexed: 02/15/2024] Open
Abstract
Background Xijiao Dihuang decoction (XJDHT), a traditional Chinese medicine, is widely used to treat patients with sepsis. However, the mechanisms underlying the effects of XJDHT on cardiac dysfunction have yet to be fully elucidated. The present study evaluated the potential utility of XJDHT in protecting against sepsis-induced cardiac dysfunction and myocardial injury. Methods The mice were randomly divided into 3 groups and administered Lipopolysaccharide (LPS,10 mg/kg) or equivalent saline solution (control) and treated with XJDHT (10 g/kg/day) or saline by gavage for 72 hours. XJDHT was dissolved in 0.9% sodium chloride and administered at 200 μL per mouse. Transthoracic echocardiography, RNA-seq, TUNEL assays and hematoxylin and eosin (H&E) staining of cardiac tissues were performed. Results Treatment with XJDHT significantly enhanced myocardial function and attenuated pathological change, infiltration of inflammatory cells, levels of TNF-α, IL-1β and expression of TLR4 and NF-κB in mice with sepsis. RNA sequencing and Kyoto Encyclopedia of Genes and Genomes pathway analyses identified 531 differentially expressed genes and multiple enriched signaling pathways including the PI3K/AKT pathway. Further, XJDHT attenuated cardiac apoptosis and decreased Bax protein expression while increasing protein levels of Bcl-2, PI3K, and p-AKT in cardiac tissues of mice with sepsis. Conclusion In summary, XJDHT improves cardiac function in a murine model of sepsis by attenuating cardiac inflammation and apoptosis via suppressing the TLR4/NF-κB pathway and activating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Wei Li
- The People’s Hospital of Fujian Traditional Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Mingrui Lin
- The People’s Hospital of Fujian Traditional Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Jiapeng Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, People’s Republic of China
| | - Qihang Ding
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, People’s Republic of China
| | - Xiaoling Chen
- Department of Infectious Disease, Fujian Medical University Union Hospital, Fuzhou, Fujian, People’s Republic of China
| | - Huaiyu Chen
- The People’s Hospital of Fujian Traditional Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Zhiqing Shen
- The People’s Hospital of Fujian Traditional Medical University, Fuzhou, Fujian, People’s Republic of China
| | - Xueli Zhu
- The People’s Hospital of Fujian Traditional Medical University, Fuzhou, Fujian, People’s Republic of China
| |
Collapse
|