1
|
Li SJ, Tetri LH, Vijayan V, Elezaby A, Chiang CH, Lopez I, Ostberg NP, Cornell TT, Chen CY, Haileselassie B. Excessive mitochondrial fission and associated extracellular mitochondria mediate cardiac dysfunction in obesity cardiomyopathy. Life Sci 2025; 373:123658. [PMID: 40287058 DOI: 10.1016/j.lfs.2025.123658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
AIMS Obesity cardiomyopathy (OCM) is associated with mitochondrial dysfunction caused by altered mitochondrial dynamics. Extracellular mitochondria (exMito) are released following tissue injury under various conditions. While the excessive mitochondrial fission-mediated release of exMito as a mechanism for mitochondrial quality control in several inflammatory disorders, its role in OCM remains unclear. The present work aimed to determine if excessive mitochondrial fission and associated exMito mediate the chronic inflammatory response and cardiac remodeling in OCM. MATERIALS AND METHODS H9c2 cardiomyoblasts were treated with 200 μM palmitate (PA) to induce lipotoxicity. C57BL/6J mice were fed a high-fat diet (HFD) for 12 weeks to induce OCM. P110, a peptide inhibitor of Drp1/Fis1 interaction, was used to evaluate the impact of excessive mitochondrial fission on cardiac mitochondrial function, quality, and quantity of exMito, systemic inflammatory response, and cardiac contractile function in both models of OCM. KEY FINDINGS PA induced excessive mitochondrial fission, increased oxidative stress, decreased ATP level, and damaged exMito release in vitro. Exposure of naïve cardiomyoblasts to exMito isolated from PA treated cells resulted in mitochondrial dysfunction and a pro-inflammatory response. In vivo, HFD induced cardiac mitochondrial and contractile dysfunction, exMito release, and a pro-inflammatory response. Inhibition of Drp1/Fis1 interaction with P110 attenuated the observed effects both in vitro and in vivo. SIGNIFICANCE P110 limited lipid-induced mitochondrial dysfunction and decreased exMito release, subsequently improving the inflammatory state and contractile function in our OCM model. Drp1/Fis1 dependent fission and associated exMito release might serve as a therapeutic target for obesity induced cardiomyopathy.
Collapse
Affiliation(s)
- Sin-Jin Li
- Bachelor Program of Biotechnology and Food and Nutrition, National Taiwan University, Taipei 10617, Taiwan; Institute of Biotechnology, National Taiwan University, Taipei 10617, Taiwan; Department of Animal Science and Technology, National Taiwan University, Taipei 10672, Taiwan.
| | - Laura H Tetri
- Department of Pediatrics, Stanford University of School of Medicine, Stanford, CA 94305, USA; Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vijith Vijayan
- Department of Pediatrics, Stanford University of School of Medicine, Stanford, CA 94305, USA
| | - Aly Elezaby
- Department of Chemical and Systems Biology, Stanford, Stanford University, CA 94305, USA
| | - Chun-Hsien Chiang
- Department of Animal Science and Technology, National Taiwan University, Taipei 10672, Taiwan
| | - Ivan Lopez
- Department of Pediatrics, Stanford University of School of Medicine, Stanford, CA 94305, USA
| | - Nicolai P Ostberg
- Department of Chemical and Systems Biology, Stanford, Stanford University, CA 94305, USA
| | - Timothy T Cornell
- Department of Pediatrics, Stanford University of School of Medicine, Stanford, CA 94305, USA
| | - Ching-Yi Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei 10672, Taiwan
| | | |
Collapse
|
2
|
de Oliveira MR. Pre-clinical evidence for mitochondria as a therapeutic target for luteolin: A mechanistic view. Chem Biol Interact 2025; 413:111492. [PMID: 40154935 DOI: 10.1016/j.cbi.2025.111492] [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: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Pre-clinical evidence indicates that mitochondria may be a therapeutic target for luteolin (3',4',5,7-tetrahydroxyflavone; LUT) in different conditions. LUT modulates mitochondrial physiology in in vitro, ex vivo, and in vivo experimental models. This flavone exerted mitochondria-related antioxidant and anti-apoptotic effects, stimulated mitochondrial fusion and fission, induced mitophagy, and promoted mitochondrial biogenesis in human and animal cells and tissues. Moreover, LUT modulated the activity of components of the oxidative phosphorylation (OXPHOS) system, improving the ability of mitochondria to produce adenosine triphosphate (ATP) in certain circumstances. The mechanism of action by which LUT promoted mitochondrial benefits and protection are not completely clear yet. Nonetheless, LUT is a potential candidate to be utilized in mitochondrial therapy in the future. In this work, it is explored the mechanisms of action by which LUT modulates mitochondrial physiology in different pre-clinical experimental models.
Collapse
Affiliation(s)
- Marcos Roberto de Oliveira
- Grupo de Estudos em Neuroquímica e Neurobiologia de Moléculas Bioativas, Departamento de Química, Universidade Federal de Mato Grosso (UFMT), CEP 78060-900, Cuiaba, Mato Grosso, Brazil; Grupo de Estudos em Terapia Mitocondrial, Instituto de Ciências Básicas da Saúde (ICBS), Universidade Federal do Rio Grande do Sul (UFRGS), CEP 90035-003, Porto Alegre, Rio Grande do Sul, Brazil; Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul (UFRGS), CEP 90035-003, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
3
|
Malczak I, Gajda A, Jedziniak P. Deoxynivalenol and pigs: review of harmful effect of Mycotoxin on swine health. Porcine Health Manag 2025; 11:27. [PMID: 40349078 PMCID: PMC12066055 DOI: 10.1186/s40813-025-00441-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/17/2025] [Indexed: 05/14/2025] Open
Abstract
Mycotoxins are compounds produced by certain types of fungi, and the mycotoxin one of the most most frequently found in the tested cereal samples is deoxynivalenol (DON), naturally-occurring mycotoxin produced by Fusarium spp. An animal sensitive to the effects of this mycotoxin is the pig due to the diet containing primarily cereals and the structure of a digestive system, which causes DON to be quickly absorbed unchanged into the bloodstream and partially metabolized in the liver. An important aspect when considering the toxicity of DON is the occurrence of its modified forms, which can be transformed into DON in the digestive system. The toxic effect of DON can also be caused by other mycotoxins which co-occur in cereals. The toxic effect of DON on the body of pigs was observed mainly in the digestive, immune, and reproductive systems. The noticeable of DON's toxic effects depends on the exposure time, route of administration and mycotoxin concentration. The changes are mainly caused by impaired gene expression, inhibiting protein synthesis or the effect of DON on pathways in pigs' bodies. The negative impact of DON on the health of pigs may lead to reduced weight gain, poor health, and increased susceptibility to infections and reproductive disorders. There have also been many methods of DON elimination from cereals, but their efficiency is insufficient.
Collapse
Affiliation(s)
- Izabela Malczak
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, Partyzantów 57, Pulawy, 24-100, Poland.
| | - Anna Gajda
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, Partyzantów 57, Pulawy, 24-100, Poland
| | - Piotr Jedziniak
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, Partyzantów 57, Pulawy, 24-100, Poland
| |
Collapse
|
4
|
Anash M, Maparu K, Singh S. Unraveling cell death mechanisms in traumatic brain injury: dynamic roles of ferroptosis and necroptosis. Mol Biol Rep 2025; 52:381. [PMID: 40208458 DOI: 10.1007/s11033-025-10489-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Traumatic brain injury (TBI) remains a major cause of mortality and long-term disability worldwide, with ferroptosis and necroptosis emerging as key drivers of secondary neuronal damage. Ferroptosis, characterized by iron-dependent lipid peroxidation and mitochondrial dysfunction, exacerbates oxidative stress and neuronal cell death. In parallel, necroptosis, mediated by receptor-interacting protein kinases (RIPK1 and RIPK3), amplifies inflammation through membrane rupture and the release of cellular components. Mitochondrial dynamics, involving fission and fusion processes, play a dual role in regulating these pathways. While mitochondrial fusion preserves cellular integrity and reduces oxidative stress, excessive mitochondrial fission driven by dynamin-related protein 1 (DRP1) accelerates necroptotic signaling and neuronal injury. This intricate interplay between ferroptosis, necroptosis, and mitochondrial dynamics highlights potential therapeutic targets. Modulating these pathways through tailored interventions could reduce neuronal damage, mitigate neuroinflammation, and improve functional outcomes in TBI patients. Advancing our understanding of these mechanisms is essential for developing precision therapies that address the complex pathology of traumatic brain injury.
Collapse
Affiliation(s)
- Mohd Anash
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Kousik Maparu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Shamsher Singh
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
5
|
Wang Z, Zhu J, Xu M, Ma X, Shen M, Yan J, Gan G, Zhou X. Transplantation of exogenous mitochondria mitigates myocardial dysfunction after cardiac arrest. eLife 2025; 13:RP98554. [PMID: 40207621 PMCID: PMC11984951 DOI: 10.7554/elife.98554] [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] [Indexed: 04/11/2025] Open
Abstract
The incidence of post-cardiac arrest myocardial dysfunction (PAMD) is high, and there is currently no effective treatment available. This study aims to investigate the protective effects of exogenous mitochondrial transplantation in Sprague-Dawley (SD) rats. Exogenous mitochondrial transplantation can enhance myocardial function and improve the survival rate. Mechanistic studies suggest that mitochondrial transplantation can limit impairment in mitochondrial morphology, augment the activity of mitochondrial complexes II and IV, and raise ATP level. As well, mitochondrial therapy ameliorated oxidative stress imbalance, reduced myocardial injury, and thus improved PAMD after cardiopulmonary resuscitation (CPR).
Collapse
Affiliation(s)
- Zhen Wang
- The First School of Clinical Medicine, Southern Medical UniversityGuangzhouChina
| | - Jie Zhu
- The First School of Clinical Medicine, Southern Medical UniversityGuangzhouChina
| | - Mengda Xu
- Department of Anesthesiology, General Hospital of Central Theater Command of PLAWuhanChina
| | - Xuyuan Ma
- Base of Central Theater Command of People's Liberation Army, Hubei University of MedicineWuhanChina
| | - Maozheng Shen
- Base of Central Theater Command of People's Liberation Army, Hubei University of MedicineWuhanChina
| | - Jingyu Yan
- Department of Anesthesiology, General Hospital of Central Theater Command of PLAWuhanChina
| | - Guosheng Gan
- The First School of Clinical Medicine, Southern Medical UniversityGuangzhouChina
- Department of Anesthesiology, General Hospital of Central Theater Command of PLAWuhanChina
| | - Xiang Zhou
- The First School of Clinical Medicine, Southern Medical UniversityGuangzhouChina
- Department of Anesthesiology, General Hospital of Central Theater Command of PLAWuhanChina
| |
Collapse
|
6
|
Yu L, Wang X, Lei Q, Liu Y, Li Z, Dai X, Song Z, He Y, Gao S, Yu C, Li L. Tongmai Yangxin pill alleviates myocardial ischemia/reperfusion injury by regulating mitochondrial fusion and fission through the estrogen receptor alpha/peroxisome proliferator-activated receptor gamma coactivator-1 alpha signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119639. [PMID: 40096900 DOI: 10.1016/j.jep.2025.119639] [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: 12/30/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Tongmai Yangxin Pill (TMYX) is derived from the Zhigancao decoction recorded in Treatise on Cold Damage Disorders (Shang Han Lun) by Zhang Zhongjing during the Han dynasty. The prescription of TMYX reflects a therapeutic rationale and efficacy unique to traditional Chinese medicine. TMYX is clinically effective in alleviating myocardial ischemia-reperfusion injury (MI/RI). However, the precise active ingredients and underlying mechanisms remain unclear. AIM OF THE STUDY The primary objective of this study was to investigate the potential of TMYX in addressing MI/RI by activating the estrogen receptor ERα. We hypothesized that this action upregulates PGC-1α activity, subsequently promoting a balanced regulation of mitochondrial fusion and fission. MATERIALS AND METHODS UPLC-Q-TOF-MS/MS was used to identify the active components of TMYX. Subsequently, a network pharmacology approach was used to uncover the therapeutic targets and underlying pharmacological mechanisms through which TMYX mitigates MI/RI. Lastly, the anticipated outcomes were confirmed through in vivo and in vitro experimental validations. RESULTS Using UPLC-Q-TOF-MS/MS, we successfully identified 53 distinct compounds in TMYX. Network pharmacology analysis revealed 20 key TMYX targets associated with MI/RI. Enrichment studies using GO and KEGG analyses revealed that these targets were mainly associated with mitochondrial processes and estrogen signaling pathways. Both in vivo and in vitro studies confirmed that TMYX markedly improved mitochondrial function through the ERα/PGC-1α signaling cascade, leading to a reduction in the size of myocardial infarctions and the incidence of apoptosis. Notably, combining TMYX with siERα abolished the protective effect of TMYX on the mitochondria. CONCLUSION TMYX therapy can improve cardiac function in MI/RI. This effect is likely mediated by the ERα/PGC-1α signaling pathway. However, given the complex multi-component composition of traditional Chinese medicine formulas, additional studies are necessary to confirm the findings of this research.
Collapse
Affiliation(s)
- Lu Yu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Xu Wang
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Qina Lei
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Yutong Liu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Zhu Li
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, 310053, China
| | - Xiangdong Dai
- Pharmaron Beijing Co., Ltd. (China), BDA, 6 Taihe Road, Beijing, 100176, China
| | - Zhihui Song
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Yuanyuan He
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Shan Gao
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Chunquan Yu
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China
| | - Lin Li
- Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, West Area, Tuanbo New Town, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
7
|
Zuo X. Mitochondrial Imbalance in Down Syndrome: A Driver of Accelerated Brain Aging? Aging Dis 2025:AD.2025.0189. [PMID: 40249934 DOI: 10.14336/ad.2025.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/06/2025] [Indexed: 04/20/2025] Open
Abstract
Down syndrome (DS), caused by trisomy of chromosome 21 (HSA21), is a complex condition associated with neurodevelopmental impairments and accelerated brain aging, often culminating in early-onset Alzheimer's disease (AD). Central to this accelerated aging is mitochondrial imbalance, characterized by disrupted energy metabolism, increased oxidative stress, impaired dynamics, and defective quality control mechanisms like mitophagy. These abnormalities exacerbate neuronal vulnerability, driving cognitive decline and neurodegeneration. This review examines the genetic and biochemical underpinnings of mitochondrial dysfunction in DS, with a focus on the role of HSA21-encoded genes. We also highlight how mitochondrial dysfunction, amplified by oxidative stress and HSA21 gene dosage effects, converges with cellular senescence and neuroinflammation to accelerate Alzheimer-like pathology and brain aging in DS. Finally, we discuss emerging therapeutic strategies targeting mitochondrial pathways, which hold promise for mitigating neurodegenerative phenotypes and improving outcomes in DS.
Collapse
|
8
|
Solta A, Ernhofer B, Boettiger K, Lang C, Megyesfalvi Z, Mendrina T, Kirchhofer D, Timelthaler G, Szeitz B, Rezeli M, Aigner C, Haschemi A, Unger LW, Dome B, Schelch K. Unveiling the powerhouse: ASCL1-driven small cell lung cancer is characterized by higher numbers of mitochondria and enhanced oxidative phosphorylation. Cancer Metab 2025; 13:16. [PMID: 40165271 PMCID: PMC11959836 DOI: 10.1186/s40170-025-00382-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 03/01/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Small cell lung cancer (SCLC) is an aggressive malignancy with distinct molecular subtypes defined by transcription factors and inflammatory characteristics. This follow-up study aimed to validate the unique metabolic phenotype in achaete-scute homologue 1 (ASCL1)-driven SCLC cell lines and human tumor tissue. METHODS Metabolic alterations were analyzed using proteomic data. Structural and functional differences of mitochondria were investigated using qPCR, flow cytometry, confocal imaging, and transmission electron microscopy and seahorse assays. Several metabolic inhibitors were tested using MTT-based and clonogenic assays. Single-cell enzyme activity assays were conducted on cell lines and tumor tissue samples of SCLC patients. RESULTS We found increased mitochondrial numbers correlating with higher oxidative phosphorylation activity in ASCL1-dominant cells compared to other SCLC subtypes. Metabolic inhibitors targeting mitochondrial respiratory complex-I or carnitine palmitoyltransferase 1 revealed higher responsiveness in SCLC-A. Conversely, we demonstrated that non-ASCL1-driven SCLCs with lower oxidative signatures show dependence on glutaminolysis as evidenced by the enhanced susceptibility to glutaminase inhibition. Accordingly, we detected increased glutamate-dehydrogenase activity in non-ASCL1-dominant cell lines as well as in human SCLC tissue samples. CONCLUSIONS Distinct SCLC subtypes exhibit unique metabolic vulnerabilities, suggesting potential for subtype-specific therapies targeting the respiratory chain, fatty acid transport, or glutaminolysis.
Collapse
Grants
- FWF No. T 1062-B33, FWF I3522, FWF I3977 and I4677, Sonderforschungsbereich F83 Austrian Science Fund
- FWF No. T 1062-B33, FWF I3522, FWF I3977 and I4677, Sonderforschungsbereich F83 Austrian Science Fund
- FWF No. T 1062-B33, FWF I3522, FWF I3977 and I4677, Sonderforschungsbereich F83 Austrian Science Fund
- FWF No. T 1062-B33, FWF I3522, FWF I3977 and I4677, Sonderforschungsbereich F83 Austrian Science Fund
- 2020-1.1.6-JÖVŐ, TKP2021-EGA-33, FK-143751 and FK-147045, UNKP-20-3, UNKP-21-3 and UNKP-23-5, ÚNKP-22-3-II Nemzeti Kutatási Fejlesztési és Innovációs Hivatal
- 2020-1.1.6-JÖVŐ, TKP2021-EGA-33, FK-143751 and FK-147045, UNKP-20-3, UNKP-21-3 and UNKP-23-5, ÚNKP-22-3-II Nemzeti Kutatási Fejlesztési és Innovációs Hivatal
- 2020-1.1.6-JÖVŐ, TKP2021-EGA-33, FK-143751 and FK-147045, UNKP-20-3, UNKP-21-3 and UNKP-23-5, ÚNKP-22-3-II Nemzeti Kutatási Fejlesztési és Innovációs Hivatal
- EFOP-3.6.3-VEKOP-16-2017-00009 Semmelweis Egyetem
- FBKS-2020-22-(291) Fru Berta Kamprads Stiftelse
- 101131228-BIOSMALL HORIZON EUROPE Framework Programme
Collapse
Affiliation(s)
- Anna Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Büsra Ernhofer
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Theresa Mendrina
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Dominik Kirchhofer
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Gerald Timelthaler
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Beata Szeitz
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Melinda Rezeli
- Department of Biomedical Engineering, Lund University, Lund, Sweden
| | - Clemens Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Lukas W Unger
- Deptartment of Colorectal Surgery, Oxford University Hospitals, Oxford, UK
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria.
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary.
- National Koranyi Institute of Pulmonology, Budapest, Hungary.
- Department of Translational Medicine, Lund University, Lund, Sweden.
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Waehringer Guertel 18-20, Vienna, A-1090, Austria.
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Li T, Yang B, Liu X, Shi D, Wang Z, Chen Y, Shen C. Silica Nanoparticles Loaded With Selenium Quantum Dots Reduce Myocardial Ischemia-Reperfusion Injury by Alleviating Ferroptosis and Mitochondrial Dysfunction. Int J Nanomedicine 2025; 20:1843-1864. [PMID: 39958324 PMCID: PMC11829639 DOI: 10.2147/ijn.s500810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 01/16/2025] [Indexed: 02/18/2025] Open
Abstract
Purpose Myocardial ischemia-reperfusion (IR) injury, a significant challenge in cardiovascular treatment, is primarily driven by ferroptosis and mitochondrial dysfunction. Despite extensive research, no clinical therapies effectively target ferroptosis in IR injury. This study aims to develop selenium-quantum-dot-loaded porous silica nanospheres (Se@PSN) as a novel therapeutic approach to address IR injury. Patients and Methods Se@PSN were synthesized and tested for their reactive oxygen species (ROS) scavenging capabilities and biocompatibility. Additionally, the effects of Se@PSN on ferroptosis, mitochondrial damage, oxidative stress, and myocardial IR injury severity were evaluated. Results Se@PSN enhanced the stability of selenium quantum dots and exhibited strong ROS scavenging abilities. Additionally, Se@PSN exhibited excellent biocompatibility. The Se@PSN treatment increased GPX4 levels, effectively inhibiting ferroptosis in cardiomyocytes. Furthermore, Se@PSN promoted the expression of mitochondrial respiratory complexes, mitigating oxidative phosphorylation damage and preserving mitochondrial function. These effects collectively resulted in reduced myocardial loss, inflammation, and fibrosis following IR injury. Compared to PSN alone, Se@PSN showed superior therapeutic efficacy against IR injury. Conclusion Se@PSN exhibit great potential in reducing ferroptosis and protecting mitochondrial function, making them a promising therapeutic approach for the treatment of myocardial IR injury.
Collapse
Affiliation(s)
- Taixi Li
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Boshen Yang
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Xijian Liu
- School of Chemistry and Chemical Engineering, Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai University of Engineering Science, Shanghai, 201620, People’s Republic of China
| | - Dongmei Shi
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Zhixiang Wang
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Yizhi Chen
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| | - Chengxing Shen
- Department of Cardiology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, People’s Republic of China
| |
Collapse
|
10
|
Abbasi Moajani F, Soozangar N, Amani M, Jeddi F, Salimnejad R, Aslani MR. The suppressive effects of crocin from saffron on allergic airway inflammation through Drp1/Nfr1/Mfn2/Pgc1-alpha signaling pathway in mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118862. [PMID: 39326816 DOI: 10.1016/j.jep.2024.118862] [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: 06/15/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saffron is derived from the dried stigmas of Crocus sativus L., which was considered by ancient nations for food and medicinal purposes. In traditional medicine, the therapeutic use of Crocus sativus includes antispasmodic, antitussive and expectorant. AIM OF THE STUDY Mitochondrial fusion, fission, biogenesis, and mitophagy are essential processes for maintaining mitochondrial dynamics in response to cellular stress. The primary objective of this research was to examine how crocin affected the levels of important mitochondrial regulators, including Drp1, Pgc1α, Nrf1, and Mfn2, in the lung tissue of ovalbumin-sensitized mice. MATERIALS AND METHODS A total of fifty male BALB/C mice were randomly assigned to five unique groups (n = 10 for each group), including the control group, ovalbumin-sensitized group (OVA), OVA group treated with 30 mg/kg of crocin, OVA group treated with 60 mg/kg of crocin, and OVA group treated with 1 mg/kg of dexamethasone. Post-sensitization and ovalbumin challenge, mice lung tissues were evaluated for the expression of Drp1, Pgc1α, Nrf1, and Mfn2 mRNA levels using real-time PCR as well as histopathological assessments. RESULTS In the OVA group, there was a significant elevated in inflammatory cells such as eosinophils, neutrophils, macrophages, and lymphocytes; however, crocin (both concentrations) and dexamethasone intervention showed significant inhibitory effects (P < 0.01 to P < 0.001). Moreover, an increase in the expression of Drp1, Pgc1α, and Nrf1 levels was seen in the OVA group, while crocin and dexamethasone showed protective benefits (P < 0.05 to P < 0.001). Furthermore, the levels of Mfn2 were reduced in the lung tissue of mice exposed to ovalbumin, but this decrease was reversed by crocin 60 (P < 0.05) and dexamethasone treatment (P < 0.001). CONCLUSION In mice with OVA sensitization, the balance of mitochondrial dynamics in lung tissue was disrupted, but intervention of crocin identified to have a protective effect.
Collapse
Affiliation(s)
- Fatima Abbasi Moajani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Narges Soozangar
- Zoonoses Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mojtaba Amani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ramin Salimnejad
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mohammad Reza Aslani
- Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Physiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
11
|
Fedotova EI, Berezhnov AV, Popov DY, Shitikova EY, Vinokurov AY. The Role of mtDNA Mutations in Atherosclerosis: The Influence of Mitochondrial Dysfunction on Macrophage Polarization. Int J Mol Sci 2025; 26:1019. [PMID: 39940788 PMCID: PMC11817597 DOI: 10.3390/ijms26031019] [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: 11/27/2024] [Revised: 01/06/2025] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Atherosclerosis is a complex inflammatory process associated with high-mortality cardiovascular diseases. Today, there is a growing body of evidence linking atherosclerosis to mutations of mitochondrial DNA (mtDNA). But the mechanism of this link is insufficiently studied. Atherosclerosis progression involves different cell types and macrophages are one of the most important. Due to their high plasticity, macrophages can demonstrate pro-inflammatory and pro-atherogenic (macrophage type M1) or anti-inflammatory and anti-atherogenic (macrophage type M2) effects. These two cell types, formed as a result of external stimuli, differ significantly in their metabolic profile, which suggests the central role of mitochondria in the implementation of the macrophage polarization route. According to this, we assume that mtDNA mutations causing mitochondrial disturbances can play the role of an internal trigger, leading to the formation of macrophage M1 or M2. This review provides a comparative analysis of the characteristics of mitochondrial function in different types of macrophages and their possible associations with mtDNA mutations linked with inflammation-based pathologies including atherosclerosis.
Collapse
Affiliation(s)
- Evgeniya I. Fedotova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (E.I.F.); (A.V.B.)
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Alexey V. Berezhnov
- Institute of Cell Biophysics of the Russian Academy of Sciences, Pushchino 142290, Russia; (E.I.F.); (A.V.B.)
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Daniil Y. Popov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Elena Y. Shitikova
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| | - Andrey Y. Vinokurov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (D.Y.P.); (E.Y.S.)
| |
Collapse
|
12
|
Zi-Chang N, Ran A, Hui-Hui S, Qi J, Jun-Li S, Yan-Xu C, Yu-Hong L, Shu-Fei F, Hao-Ping M. Columbianadin Ameliorates Myocardial Injury by Inhibiting Autophagy Through the PI3K/Akt/mTOR Signaling Pathway in AMI Mice and Hypoxic H9c2 Cells. Phytother Res 2025; 39:521-535. [PMID: 39568432 DOI: 10.1002/ptr.8387] [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: 05/23/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 11/22/2024]
Abstract
Acute myocardial infarction (AMI) is a leading cause of mortality among cardiovascular diseases, yet effective therapies for AMI are limited. Previous studies have suggested cardioprotective effects of columbianadin (CBN), but its specific role in AMI and the underlying mechanisms remain unclear. This study aims to investigate whether CBN influences AMI and to elucidate the underlying mechanisms. We conducted a network pharmacology analysis to investigate the relationship between CBN and AMI. The AMI model was established by ligating the left anterior descending (LAD) artery in C57BL/6J mice, which were subsequently administered CBN. Hypoxic H9c2 cells were utilized to evaluate the effects of CBN in vitro. Our study revealed that CBN treatment significantly reduced myocardial infarction in AMI mice. It enhanced mitochondrial function and suppressed autophagy flux in hypoxic H9c2 cells. Furthermore, CBN downregulated the expression of LC3, Beclin1, and Atg 5 genes and proteins. In response to CBN treatment, the phosphorylation levels of PI3K, Akt, and mTOR increased. Notably, RAPA attenuated the protective effect of CBN in enhancing the survival of hypoxic H9c2 cells and abolished its regulation of autophagy-related proteins via the PI3K/Akt/mTOR signaling pathway. In conclusion, CBN reduces myocardial damage by suppressing autophagy via the PI3K/Akt/mTOR signaling pathway in AMI mice and hypoxic H9c2 cells.
Collapse
Affiliation(s)
- Niu Zi-Chang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - An Ran
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Shi Hui-Hui
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Jin Qi
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Song Jun-Li
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Chang Yan-Xu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Li Yu-Hong
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Fu Shu-Fei
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- Shcool of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Mao Hao-Ping
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulea, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| |
Collapse
|
13
|
Zhao M, Xie J, Zhang J, Zhao B, Zhang Y, Xue J, Zhang R, Zhang R, Wang H, Li Y, Ge W, Zhou X. Disturbance of mitochondrial dynamics led to spermatogenesis disorder in mice exposed to polystyrene micro- and nanoplastics. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124935. [PMID: 39260550 DOI: 10.1016/j.envpol.2024.124935] [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: 05/17/2024] [Revised: 08/11/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
The widespread presence of polystyrene micro- and nanoplastics (PS-MPs/NPs) in the environment poses a threat to the health of the population. Animal studies have shown PS-MPs/NPs had male reproductive toxicity, while its mechanisms are unclear. To investigate that, male Balb/c mice were randomized into 3 groups: the control, 1 μm PS-MPs and 70 nm PS-NPs group, and they were given PS-MPs/NPs by intratracheal instillation for 28 days. Results revealed that PS-MPs/NPs up-regulated the expression of mitochondrial fission related factors (p-DRP1/DRP1, FIS1) and down-regulated the level of mitochondrial fusion related factors (MFN1/2, OPA1), causing over mitochondrial fission, which activating mitochondrial apoptotic pathway (BAX, Cleaved-Caspase9, Cleaved-Caspase3), resulting in cell apoptosis. Moreover, the damaged structure of mitochondria and over mitochondrial fission caused mitochondrial DNA (mtDNA) to translocate from mitochondria to cytoplasm, which activated DNA sensing pathway (cGAS-STING) and induced cell pyroptosis in testis by raising the expression of inflammation factors (NLRP3, ASC, Caspase1 p20, IL-1β). In vitro, by using the mitochondrial fission inhibitor Mdivi-1, it is found that PS-NPs-induced cell apoptosis and pyroptosis were associated with over mitochondrial fission. Taken together, we conclude that PS-MPs/NPs cause spermatogenesis disorder possibly through damaging mitochondrial structure and dynamic homeostasis, which on the one hand results in mitochondria-mediated apoptosis, and on the other hand leads to mtDNA mislocalization, activating cGAS-STING pathway and inflammation, ultimately resulting in pyroptosis. This study may provide a new reference to the potential mechanisms of male reproductive toxicity caused by PS-MPs/NPs.
Collapse
Affiliation(s)
- Moxuan Zhao
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Junhong Xie
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Jiaxiang Zhang
- Class of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Bosen Zhao
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yue Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Jinglong Xue
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Ruxuan Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Ruiyang Zhang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Hongou Wang
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yanbo Li
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Wei Ge
- Department of Biomedical Sciences and Centre of Reproduction, Development and Aging (CRDA), Faculty of Health Sciences, University of Macau, Taipa, Macau, 519000, China
| | - Xianqing Zhou
- Department of Toxicology and Hygienic Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
14
|
Paraskevaidis I, Kourek C, Farmakis D, Tsougos E. Mitochondrial Dysfunction in Cardiac Disease: The Fort Fell. Biomolecules 2024; 14:1534. [PMID: 39766241 PMCID: PMC11673776 DOI: 10.3390/biom14121534] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/10/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Myocardial cells and the extracellular matrix achieve their functions through the availability of energy. In fact, the mechanical and electrical properties of the heart are heavily dependent on the balance between energy production and consumption. The energy produced is utilized in various forms, including kinetic, dynamic, and thermal energy. Although total energy remains nearly constant, the contribution of each form changes over time. Thermal energy increases, while dynamic and kinetic energy decrease, ultimately becoming insufficient to adequately support cardiac function. As a result, toxic byproducts, unfolded or misfolded proteins, free radicals, and other harmful substances accumulate within the myocardium. This leads to the failure of crucial processes such as myocardial contraction-relaxation coupling, ion exchange, cell growth, and regulation of apoptosis and necrosis. Consequently, both the micro- and macro-architecture of the heart are altered. Energy production and consumption depend on the heart's metabolic resources and the functional state of the cardiac structure, including cardiomyocytes, non-cardiomyocyte cells, and their metabolic and energetic behavior. Mitochondria, which are intracellular organelles that produce more than 95% of ATP, play a critical role in fulfilling all these requirements. Therefore, it is essential to gain a deeper understanding of their anatomy, function, and homeostatic properties.
Collapse
Affiliation(s)
- Ioannis Paraskevaidis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (I.P.); (D.F.)
- Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece;
| | - Christos Kourek
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (I.P.); (D.F.)
| | - Dimitrios Farmakis
- Medical School of Athens, National and Kapodistrian University of Athens, 15772 Athens, Greece; (I.P.); (D.F.)
| | - Elias Tsougos
- Department of Cardiology, Hygeia Hospital, 15123 Athens, Greece;
| |
Collapse
|
15
|
Bruszt K, Horvath O, Ordog K, Toth S, Juhasz K, Vamos E, Fekete K, Gallyas F, Toth K, Halmosi R, Deres L. Cardiac effects of OPA1 protein promotion in a transgenic animal model. PLoS One 2024; 19:e0310394. [PMID: 39570915 PMCID: PMC11581344 DOI: 10.1371/journal.pone.0310394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/30/2024] [Indexed: 11/24/2024] Open
Abstract
Mitochondria form a dynamic network in cells, regulated by the balance between mitochondrial fusion and fission. The inhibition of mitochondrial fission can have positive effects in acute ischemic/reperfusion injury models by preventing the fall in mitochondrial membrane potential associated with fission processes. However, inhibition of fission in chronic models is disadvantageous because it obstructs the elimination of damaged mitochondrial fragments. OPA1, in view of previous results, is a possible therapeutic target as a fusion promoter and structure stabilizer protein. We used transgenic mice in which the OMA1 cleavage sites of OPA1 were deleted. This resulted in a higher representation of L-OPA1 compared to S-OPA1. After genotyping and model validation, all animals were examined by echocardiograph on two occasions, at weeks 11 and 36. Histological samples were taken from hearts to examine mitochondrial morphology and structure remodeling. The signaling pathways related to mitochondrial dynamic processes were evaluated. Cardiomyocytes were isolated from neonatal mice to determine the efficiency of mitochondrial respiration using the SeaHorse assay method. OPA1 protein promotion has a negative effect on systolic function during aging. We confirmed that volume overload and ventricular remodeling did not manifest. The reason behind the loss of pump function might be, at least partly, due to the energy deficit caused by mitochondrial respiratory failure and damage in mitochondrial quality control pathways.
Collapse
Affiliation(s)
- Kitti Bruszt
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Orsolya Horvath
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Katalin Ordog
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Szilard Toth
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Kata Juhasz
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Eszter Vamos
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Katalin Fekete
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Ferenc Gallyas
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, Pecs, Hungary
| | - Kalman Toth
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Robert Halmosi
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| | - Laszlo Deres
- 1st Department of Medicine, University of Pecs Medical School, Pecs, Hungary
- Szentagothai Research Centre, University of Pecs, Pecs, Hungary
| |
Collapse
|
16
|
Park JS, Jeon H, Lee Y, Cheon SY, Lee D, Lim SG, Koo H. Rapid DNA Repair in Mesenchymal Stem Cells and Bone Regeneration by Nanoparticle-Based Codelivery of Nrf2-mRNA and Dexamethasone. ACS NANO 2024; 18:31877-31890. [PMID: 39520360 DOI: 10.1021/acsnano.4c08939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Directional differentiation is a key factor determining the result of stem cell therapy. Herein, we developed a polyethylenimine (PEI)-coated poly(lactic-co-glycolic) acid (PLGA) nanoparticle (mPDN) carrying both nuclear factor erythroid 2-related factor 2 (Nrf2) mRNA and dexamethasone (Dex) to human mesenchymal stem cells (hMSCs). The combination of Dex and Nrf2-mRNA delivered by mPDN promoted the osteogenic differentiation of hMSCs. In particular, Nrf2-mRNA rapidly reduced the DNA damage caused by ROS due to early and efficient gene expression at 3 h after treatment, which was not achieved in traditional pDNA systems. High and rapid transfection, effective ROS-scavenging effect, and protection of mitochondrial dynamics were observed in hMSCs after treatment with the resulting Nrf2-mPDN. Osteogenic differentiation was also observed in 3D pellets for up to 5 weeks. Finally, the effects of rapid DNA repair in hMSCs by Nrf2-mPDN and on in vivo bone regeneration were evaluated in a rat femoral bone defect model using CT. This study demonstrated the potential of an NP-based codelivery system and efficient transfection of mRNA at early stages in hMSCs for bone regeneration and stem cell therapy.
Collapse
Affiliation(s)
- Ji Sun Park
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Hayoung Jeon
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yeeun Lee
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seo Young Cheon
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Seong Gi Lim
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Medical Sciences (Graduate School), and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
17
|
Paing YMM, Eom Y, Lee SH. Benzopyrene represses mitochondrial fission factors and PINK1/Parkin-mediated mitophagy in primary astrocytes. Toxicology 2024; 508:153926. [PMID: 39147092 DOI: 10.1016/j.tox.2024.153926] [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: 04/30/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Mitochondria are essential for various physiological functions in astrocytes in the brain, such as maintaining ion and pH homeostasis, regulating neurotransmission, and modulating neuroinflammation. Mitophagy, a form of autophagy specific to mitochondria, is essential for ensuring mitochondrial quality and function. Benzo[a]pyrene (BaP) accumulates in the brain, and exposure to it is recognized as an environmental risk factor for neurodegenerative diseases. However, while the toxic mechanisms of BaP have been investigated in neurons, their effects on astrocytes-the most prevalent glial cells in the brain-are not clearly understood. Therefore, this study aims to investigate the toxic effects of exposure to BaP on mitochondria in primary astrocytes. Fluorescent probes and genetically encoded indicators were utilized to visualize mitochondrial morphology and physiology, and regulatory factors involved in mitochondrial morphology and mitophagy were assessed. Additionally, the mitochondrial respiration rate was measured in BaP-exposed astrocytes. BaP exposure resulted in mitochondrial enlargement owing to the suppression of mitochondrial fission factors. Furthermore, BaP-exposed astrocytes demonstrated reduced mitophagy and exhibited aberrant mitochondrial function and physiology, such as altered mitochondrial respiration rates, increased mitochondrial superoxide, disrupted mitochondrial membrane potential, and dysregulated mitochondrial Ca2+. These findings offer insights into the underlying toxic mechanisms of BaP exposure in neurodegenerative diseases by inducing aberrant mitophagy and mitochondrial dysfunction in astrocytes.
Collapse
Affiliation(s)
- Yunn Me Me Paing
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Yunkyung Eom
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| |
Collapse
|
18
|
Zhao YC, Gao BH. Integrative effects of resistance training and endurance training on mitochondrial remodeling in skeletal muscle. Eur J Appl Physiol 2024; 124:2851-2865. [PMID: 38981937 DOI: 10.1007/s00421-024-05549-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 07/03/2024] [Indexed: 07/11/2024]
Abstract
Resistance training activates mammalian target of rapamycin (mTOR) pathway of hypertrophy for strength gain, while endurance training increases peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) pathway of mitochondrial biogenesis benefiting oxidative phosphorylation. The conventional view suggests that resistance training-induced hypertrophy signaling interferes with endurance training-induced mitochondrial remodeling. However, this idea has been challenged because acute leg press and knee extension in humans enhance both muscle hypertrophy and mitochondrial remodeling signals. Thus, we first examined the muscle mitochondrial remodeling and hypertrophy signals with endurance training and resistance training, respectively. In addition, we discussed the influence of resistance training on muscle mitochondria, demonstrating that the PGC-1α-mediated muscle mitochondrial adaptation and hypertrophy occur simultaneously. The second aim was to discuss the integrative effects of concurrent training, which consists of endurance and resistance training sessions on mitochondrial remodeling. The study found that the resistance training component does not reduce muscle mitochondrial remodeling signals in concurrent training. On the contrary, concurrent training has the potential to amplify skeletal muscle mitochondrial biogenesis compared to a single exercise model. Concurrent training involving differential sequences of resistance and endurance training may result in varied mitochondrial biogenesis signals, which should be linked to the pre-activation of mTOR or PGC-1α signaling. Our review proposed a mechanism for mTOR signaling that promotes PGC-1α signaling through unidentified pathways. This mechanism may be account for the superior muscle mitochondrial remodeling change following the concurrent training. Our review suggested an interaction between resistance training and endurance training in skeletal muscle mitochondrial adaptation.
Collapse
Affiliation(s)
- Yong-Cai Zhao
- College of Exercise and Health, Tianjin University of Sport, No. 16 Donghai Road, Jinghai District, Tianjin, 301617, China.
| | - Bing-Hong Gao
- School of Athletic Performance, Shanghai University of Sport, No. 399 Changhai Road, Yangpu District, Shanghai, 200438, China
| |
Collapse
|
19
|
Choi DH, Lee SM, Park BN, Lee MH, Yang DE, Son YK, Kim SE, An WS. Omega-3 Fatty Acids Modify Drp1 Expression and Activate the PINK1-Dependent Mitophagy Pathway in the Kidney and Heart of Adenine-Induced Uremic Rats. Biomedicines 2024; 12:2107. [PMID: 39335620 PMCID: PMC11429207 DOI: 10.3390/biomedicines12092107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Mitochondrial homeostasis is controlled by biogenesis, dynamics, and mitophagy. Mitochondrial dysfunction plays a central role in cardiovascular and renal disease and omega-3 fatty acids (FAs) are beneficial for cardiovascular disease. We investigated whether omega-3 fatty acids (FAs) regulate mitochondrial biogenesis, dynamics, and mitophagy in the kidney and heart of adenine-induced uremic rats. Eighteen male Sprague Dawley rats were divided into normal control, adenine control, and adenine with omega-3 FA groups. Using Western blot analysis, the kidney and heart expression of mitochondrial homeostasis-related molecules, including peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), dynamin-related protein 1 (Drp1), and phosphatase and tensin homolog-induced putative kinase 1 (PINK1) were investigated. Compared to normal, serum creatinine and heart weight/body weight in adenine control were increased and slightly improved in the omega-3 FA group. Compared to the normal controls, the expression of PGC-1α and PINK1 in the kidney and heart of the adenine group was downregulated, which was reversed after omega-3 FA supplementation. Drp1 was upregulated in the kidney but downregulated in the heart in the adenine group. Drp1 expression in the heart recovered in the omega-3 FA group. Mitochondrial DNA (mtDNA) was decreased in the kidney and heart of the adenine control group but the mtDNA of the heart was recovered in the omega-3 FA group. Drp1, which is related to mitochondrial fission, may function oppositely in the uremic kidney and heart. Omega-3 FAs may be beneficial for mitochondrial homeostasis by activating mitochondrial biogenesis and PINK1-dependent mitophagy in the kidney and heart of uremic rats.
Collapse
Affiliation(s)
- Dong Ho Choi
- Department of Internal Medicine, Good Moon Hwa Hospital, Busan 48735, Republic of Korea
| | - Su Mi Lee
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Bin Na Park
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Mi Hwa Lee
- Department of Anatomy and Cell Biology, Dong-A University, Busan 49201, Republic of Korea;
| | - Dong Eun Yang
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Young Ki Son
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Seong Eun Kim
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
| | - Won Suk An
- Department of Internal Medicine, Dong-A University, Busan 49201, Republic of Korea; (S.M.L.); (B.N.P.); (D.E.Y.); (Y.K.S.); (S.E.K.)
- Medical Science Research Center, Dong-A University, Busan 49201, Republic of Korea
| |
Collapse
|
20
|
Guo M, Liu R, Zhang F, Qu J, Yang Y, Li X. A new perspective on liver diseases: Focusing on the mitochondria-associated endoplasmic reticulum membranes. Pharmacol Res 2024; 208:107409. [PMID: 39284429 DOI: 10.1016/j.phrs.2024.107409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/29/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
The pathogenesis of liver diseases is multifaceted and intricate, posing a persistent global public health challenge with limited therapeutic options. Therefore, further research into liver diseases is imperative for better comprehension and advancement in treatment strategies. Numerous studies have confirmed the endoplasmic reticulum (ER) and mitochondria as key organelles driving liver diseases. Notably, the mitochondrial-associated ER membranes (MAMs) establish a physical and functional connection between the ER and mitochondria, highlighting the importance of inter-organelle communication in maintaining their functional homeostasis. This review delves into the intricate architecture and regulative mechanism of the integrated MAM that facilitate the physiological transfer of signals and substances between organelles. Additionally, we also provide a detailed overview regarding the varied pathogenic roles of malfunctioning MAM in liver diseases, focusing on its involvement in the progression of ER stress and mitochondrial dysfunction, the regulation of mitochondrial dynamics and Ca2+ transfer, as well as the disruption of lipid and glucose homeostasis. Furthermore, the current challenges and prospects associated with MAM in liver disease research are thoroughly discussed. In conclusion, elucidating the specific structure and function of MAM in different liver diseases may pave the way for novel therapeutic strategies.
Collapse
Affiliation(s)
- Mengyu Guo
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Runping Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, 100029, China
| | - Fukun Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Jiaorong Qu
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Yun Yang
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, 100029, China.
| |
Collapse
|
21
|
Kaur S, Khullar N, Navik U, Bali A, Bhatti GK, Bhatti JS. Multifaceted role of dynamin-related protein 1 in cardiovascular disease: From mitochondrial fission to therapeutic interventions. Mitochondrion 2024; 78:101904. [PMID: 38763184 DOI: 10.1016/j.mito.2024.101904] [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: 12/06/2023] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Mitochondria are central to cellular energy production and metabolic regulation, particularly in cardiomyocytes. These organelles constantly undergo cycles of fusion and fission, orchestrated by key proteins like Dynamin-related Protein 1 (Drp-1). This review focuses on the intricate roles of Drp-1 in regulating mitochondrial dynamics, its implications in cardiovascular health, and particularly in myocardial infarction. Drp-1 is not merely a mediator of mitochondrial fission; it also plays pivotal roles in autophagy, mitophagy, apoptosis, and necrosis in cardiac cells. This multifaceted functionality is often modulated through various post-translational alterations, and Drp-1's interaction with intracellular calcium (Ca2 + ) adds another layer of complexity. We also explore the pathological consequences of Drp-1 dysregulation, including increased reactive oxygen species (ROS) production and endothelial dysfunction. Furthermore, this review delves into the potential therapeutic interventions targeting Drp-1 to modulate mitochondrial dynamics and improve cardiovascular outcomes. We highlight recent findings on the interaction between Drp-1 and sirtuin-3 and suggest that understanding this interaction may open new avenues for therapeutically modulating endothelial cells, fibroblasts, and cardiomyocytes. As the cardiovascular system increasingly becomes the focal point of aging and chronic disease research, understanding the nuances of Drp-1's functionality can lead to innovative therapeutic approaches.
Collapse
Affiliation(s)
- Satinder Kaur
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda India
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda India.
| |
Collapse
|
22
|
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
|
23
|
Sun Y, Zhang R, Tian L, Pan Y, Sun X, Huang Z, Fan J, Chen J, Zhang K, Li S, Chen W, Bazzano LA, Kelly TN, He J, Bundy JD, Li C. Novel Metabolites Associated With Blood Pressure After Dietary Interventions. Hypertension 2024; 81:1966-1975. [PMID: 39005213 PMCID: PMC11324412 DOI: 10.1161/hypertensionaha.124.22999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 06/21/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND The blood pressure (BP) etiologic study is complex due to multifactorial influences, including genetic, environmental, lifestyle, and their intricate interplays. We used a metabolomics approach to capture internal pathways and external exposures and to study BP regulation mechanisms after well-controlled dietary interventions. METHODS In the ProBP trail (Protein and Blood Pressure), a double-blinded crossover randomized controlled trial, participants underwent dietary interventions of carbohydrate, soy protein, and milk protein, receiving 40 g daily for 8 weeks, with 3-week washout periods. We measured plasma samples collected at baseline and at the end of each dietary intervention. Multivariate linear models were used to evaluate the association between metabolites and systolic/diastolic BP. Nominally significant metabolites were examined for enriching biological pathways. Significant ProBP findings were evaluated for replication among 1311 participants of the BHS (Bogalusa Heart Study), a population-based study conducted in the same area as ProBP. RESULTS After Bonferroni correction for 77 independent metabolite clusters (α=6.49×10-4), 18 metabolites were significantly associated with BP at baseline or the end of a dietary intervention, of which 11 were replicated in BHS. Seven emerged as novel discoveries, which are as follows: 1-linoleoyl-GPE (18:2), 1-oleoyl-GPE (18:1), 1-stearoyl-2-linoleoyl-GPC (18:0/18:2), 1-palmitoyl-2-oleoyl-GPE (16:0/18:1), maltose, N-stearoyl-sphinganine (d18:0/18:0), and N6-carbamoylthreonyladenosine. Pathway enrichment analyses suggested dietary protein intervention might reduce BP through pathways related to G protein-coupled receptors, incretin function, selenium micronutrient network, and mitochondrial biogenesis. CONCLUSIONS Seven novel metabolites were identified to be associated with BP at the end of different dietary interventions. The beneficial effects of protein interventions might be mediated through specific metabolic pathways.
Collapse
Affiliation(s)
- Yixi Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Ruiyuan Zhang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Ling Tian
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Yang Pan
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago (Y.P., X.S., T.N.K.)
| | - Xiao Sun
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago (Y.P., X.S., T.N.K.)
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Jia Fan
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Kai Zhang
- Department of Environmental Health Sciences, University of Albany, State University of New York, Rensselaer (K.Z.)
| | - Shengxu Li
- Children's Minnesota Research Institute, Children's Minnesota, Minneapolis (S.L.)
| | - Wei Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Lydia A Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Tanika N Kelly
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago (Y.P., X.S., T.N.K.)
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Joshua D Bundy
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (Y.S., R.Z., L.T., Z.H., J.F., J.C., W.C., L.B., J.H., J.D.B., C.L.)
| |
Collapse
|
24
|
Huang J, Hao J, Wang P, Xu Y. The Role of Mitochondrial Dysfunction in CKD-Related Vascular Calcification: From Mechanisms to Therapeutics. Kidney Int Rep 2024; 9:2596-2607. [PMID: 39291213 PMCID: PMC11403042 DOI: 10.1016/j.ekir.2024.05.005] [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: 02/27/2024] [Revised: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 09/19/2024] Open
Abstract
Vascular calcification (VC) is a common complication of chronic kidney disease (CKD) and is closely associated with cardiovascular events. The transdifferentiation of vascular smooth muscles (VSMCs) into an osteogenic phenotype is hypothesized to be the primary cause underlying VC. However, there is currently no effective clinical treatment for VC. Growing evidence suggests that mitochondrial dysfunction accelerates the osteogenic differentiation of VSMCs and VC via multiple mechanisms. Therefore, elucidating the relationship between the osteogenic differentiation of VSMCs and mitochondrial dysfunction may assist in improving VC-related adverse clinical outcomes in patients with CKD. This review aimed to summarize the role of mitochondrial biogenesis, mitochondrial dynamics, mitophagy, and metabolic reprogramming, as well as mitochondria-associated oxidative stress (OS) and senescence in VC in patients with CKD to offer valuable insights into the clinical treatment of VC.
Collapse
Affiliation(s)
- Junmin Huang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Junfeng Hao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Peng Wang
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Yongzhi Xu
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
25
|
Lee HT, Lin CS, Liu CY, Chen P, Tsai CY, Wei YH. Mitochondrial Plasticity and Glucose Metabolic Alterations in Human Cancer under Oxidative Stress-From Viewpoints of Chronic Inflammation and Neutrophil Extracellular Traps (NETs). Int J Mol Sci 2024; 25:9458. [PMID: 39273403 PMCID: PMC11395599 DOI: 10.3390/ijms25179458] [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: 07/24/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Oxidative stress elicited by reactive oxygen species (ROS) and chronic inflammation are involved both in deterring and the generation/progression of human cancers. Exogenous ROS can injure mitochondria and induce them to generate more endogenous mitochondrial ROS to further perpetuate the deteriorating condition in the affected cells. Dysfunction of these cancer mitochondria may possibly be offset by the Warburg effect, which is characterized by amplified glycolysis and metabolic reprogramming. ROS from neutrophil extracellular traps (NETs) are an essential element for neutrophils to defend against invading pathogens or to kill cancer cells. A chronic inflammation typically includes consecutive NET activation and tissue damage, as well as tissue repair, and together with NETs, ROS would participate in both the destruction and progression of cancers. This review discusses human mitochondrial plasticity and the glucose metabolic reprogramming of cancer cells confronting oxidative stress by the means of chronic inflammation and neutrophil extracellular traps (NETs).
Collapse
Affiliation(s)
- Hui-Ting Lee
- Division of Allergy, Immunology & Rheumatology, Department of Internal Medicine, Mackay Memorial Hospital, Taipei 104, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
| | - Chen-Sung Lin
- Division of Thoracic Surgery, Department of Surgery, Taipei Hospital, Ministry of Health and Welfare, New Taipei City 242, Taiwan
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for General Education, Kainan University, Taoyuan City 338, Taiwan
| | - Chao-Yu Liu
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Division of Thoracic Surgery, Department of Surgery, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Po Chen
- Cancer Free Biotech, Taipei 114, Taiwan
| | - Chang-Youh Tsai
- School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Clinical Trial Center, Division of Immunology & Rheumatology, Fu Jen Catholic University Hospital, New Taipei City 243, Taiwan
- Faculty of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yau-Huei Wei
- Department of Medicine, Mackay Medical College, New Taipei City 252, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for Mitochondrial Medicine and Free Radical Research, Changhua Christian Hospital, Changhua City 500, Taiwan
| |
Collapse
|
26
|
Song D, Zhou X, Yu Q, Li R, Dai Q, Zeng M. ML335 inhibits TWIK2 channel-mediated potassium efflux and attenuates mitochondrial damage in MSU crystal-induced inflammation. J Transl Med 2024; 22:785. [PMID: 39175013 PMCID: PMC11342740 DOI: 10.1186/s12967-024-05303-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/22/2023] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Activation of the NLRP3 inflammasome is critical in the inflammatory response to gout. Potassium ion (K+) efflux mediated by the TWIK2 channel is an important upstream mechanism for NLRP3 inflammasome activation. Therefore, the TWIK2 channel may be a promising therapeutic target for MSU crystal-induced inflammation. In the present study, we investigated the effect of ML335, a known K2P channel modulator, on MSU crystal-induced inflammatory responses and its underlying molecular mechanisms. METHODS By molecular docking, we calculated the binding energies and inhibition constants of five K2P channel modulators (Hydroxychloroquine, Fluoxetine, DCPIB, ML365 and ML335) with TWIK2. Intracellular potassium ion concentration and mitochondrial function were assessed by flow cytometry. The interaction between MARCH5 and SIRT3 was demonstrated by immunoprecipitation and Western blotting assay. MSU suspensions were injected into mouse paw and peritoneal cavity to induce acute gout model. RESULTS ML335 has the highest binding energy and the lowest inhibition constant with TWIK2 in the five calculated K2P channel modulators. In comparison, among these five compounds, ML335 efficiently inhibited the release of IL-1β from MSU crystal-treated BMDMs. ML335 decreased MSU crystal-induced K+ efflux mainly dependent on TWIK2 channel. More importantly, ML335 can effectively inhibit the expression of the mitochondrial E3 ubiquitin ligase MARCH5 induced by MSU crystals, and MARCH5 can interact with the SIRT3 protein. ML335 blocked MSU crystal-induced ubiquitination of SIRT3 protein by MARCH5. In addition, ML335 improved mitochondrial dynamics homeostasis and mitochondrial function by inhibiting MARCH5 protein expression. ML335 attenuated the inflammatory response induced by MSU crystals in vivo and in vitro. CONCLUSION Inhibition of TWIK2-mediated K+ efflux by ML335 alleviated mitochondrial injury via suppressing March5 expression, suggesting that ML335 may be an effective candidate for the future treatment of gout.
Collapse
Affiliation(s)
- Dianze Song
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College and Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637001, Sichuan, China
| | - Xiaoqin Zhou
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College and Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637001, Sichuan, China
| | - Qingqing Yu
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College and Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637001, Sichuan, China
| | - Renjie Li
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College and Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637001, Sichuan, China
| | - Qian Dai
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College and Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637001, Sichuan, China.
- Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College, Nanchong, 637001, Sichuan, China.
| | - Mei Zeng
- Institute of Rheumatology and Immunology, The Affiliated Hospital of North Sichuan Medical College and Institute of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637001, Sichuan, China.
- Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College, Nanchong, 637001, Sichuan, China.
- North Sichuan Medical College Innovation Centre for Science and Technology, North Sichuan Medical College, Nanchong, 637001, Sichuan, China.
| |
Collapse
|
27
|
Pokharel MD, Fu P, Garcia-Flores A, Yegambaram M, Lu Q, Sun X, Unwalla H, Aggarwal S, Fineman JR, Wang T, Black SM. Inflammatory lung injury is associated with endothelial cell mitochondrial fission and requires the nitration of RhoA and cytoskeletal remodeling. Free Radic Biol Med 2024; 221:125-135. [PMID: 38734269 PMCID: PMC11179967 DOI: 10.1016/j.freeradbiomed.2024.05.019] [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: 02/20/2024] [Revised: 04/12/2024] [Accepted: 05/09/2024] [Indexed: 05/13/2024]
Abstract
Higher levels of extracellular nicotinamide phosphoribosyltransferase (eNAMPT), a TLR4 agonist, are associated with poor clinical outcomes in sepsis-induced acute lung injury (ALI). Little is known regarding the mechanisms by which eNAMPT is involved in ALI. Our recent work has identified a crucial role for mitochondrial dysfunction in ALI. Thus, this study aimed to determine if eNAMPT-mediated inflammatory injury is associated with the loss of mitochondrial function. Our data show that eNAMPT disrupted mitochondrial bioenergetics. This was associated with cytoskeleton remodeling and the loss of endothelial barrier integrity. These changes were associated with enhanced mitochondrial fission and blocked when Rho-kinase (ROCK) was inhibited. The increases in mitochondrial fission were also associated with the nitration-mediated activation of the small GTPase activator of ROCK, RhoA. Blocking RhoA nitration decreased eNAMPT-mediated mitochondrial fission and endothelial barrier dysfunction. The increase in fission was linked to a RhoA-ROCK mediated increase in Drp1 (dynamin-related protein 1) at serine(S)616. Another TLR4 agonist, lipopolysaccharide (LPS), also increased mitochondrial fission in a Drp1 and RhoA-ROCK-dependent manner. To validate our findings in vivo, we challenged C57BL/6 mice with eNAMPT in the presence and absence of the Drp1 inhibitor, Mdivi-1. Mdivi-1 treatment protected against eNAMPT-induced lung inflammation, edema, and lung injury. These studies demonstrate that mitochondrial fission-dependent disruption of mitochondrial function is essential in TLR4-mediated inflammatory lung injury and identify a key role for RhoA-ROCK signaling. Reducing mitochondrial fission could be a potential therapeutic strategy to improve ARDS outcomes.
Collapse
Affiliation(s)
- Marissa D Pokharel
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA; Center for Translational Science, Florida International University, Port St. Lucie, FL, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | | | - Manivannan Yegambaram
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Xutong Sun
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Hoshang Unwalla
- Department of Immunology and Nano-Medicine, Howard Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Saurabh Aggarwal
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94143, USA; Department of Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA
| | - Stephen M Black
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Miami, FL, USA; Center for Translational Science, Florida International University, Port St. Lucie, FL, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, University Park, FL, USA.
| |
Collapse
|
28
|
Kumar H, Kim P. Artificial intelligence in fusion protein three-dimensional structure prediction: Review and perspective. Clin Transl Med 2024; 14:e1789. [PMID: 39090739 PMCID: PMC11294035 DOI: 10.1002/ctm2.1789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 07/16/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
Recent advancements in artificial intelligence (AI) have accelerated the prediction of unknown protein structures. However, accurately predicting the three-dimensional (3D) structures of fusion proteins remains a difficult task because the current AI-based protein structure predictions are focused on the WT proteins rather than on the newly fused proteins in nature. Following the central dogma of biology, fusion proteins are translated from fusion transcripts, which are made by transcribing the fusion genes between two different loci through the chromosomal rearrangements in cancer. Accurately predicting the 3D structures of fusion proteins is important for understanding the functional roles and mechanisms of action of new chimeric proteins. However, predicting their 3D structure using a template-based model is challenging because known template structures are often unavailable in databases. Deep learning (DL) models that utilize multi-level protein information have revolutionized the prediction of protein 3D structures. In this review paper, we highlighted the latest advancements and ongoing challenges in predicting the 3D structure of fusion proteins using DL models. We aim to explore both the advantages and challenges of employing AlphaFold2, RoseTTAFold, tr-Rosetta and D-I-TASSER for modelling the 3D structures. HIGHLIGHTS: This review provides the overall pipeline and landscape of the prediction of the 3D structure of fusion protein. This review provides the factors that should be considered in predicting the 3D structures of fusion proteins using AI approaches in each step. This review highlights the latest advancements and ongoing challenges in predicting the 3D structure of fusion proteins using deep learning models. This review explores the advantages and challenges of employing AlphaFold2, RoseTTAFold, tr-Rosetta, and D-I-TASSER to model 3D structures.
Collapse
Affiliation(s)
- Himansu Kumar
- Department of Bioinformatics and Systems MedicineMcWilliams School of Biomedical InformaticsThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| | - Pora Kim
- Department of Bioinformatics and Systems MedicineMcWilliams School of Biomedical InformaticsThe University of Texas Health Science Center at HoustonHoustonTexasUSA
| |
Collapse
|
29
|
Hong GL, Kim KH, Cho SP, Lee HJ, Kim YJ, Jung JY. Korean red ginseng alleviates benign prostatic hyperplasia by dysregulating androgen receptor signaling and inhibiting DRP1-mediated mitochondrial fission. Chin J Nat Med 2024; 22:599-607. [PMID: 39059829 DOI: 10.1016/s1875-5364(24)60671-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Indexed: 07/28/2024]
Abstract
Panax ginseng (C.A. Mey.) has been traditionally employed in Korea and China to alleviate fatigue and digestive disorders. In particular, Korean red ginseng (KRG), derived from streamed and dried P. ginseng, is known for its anti-aging and anti-inflammatory properties. However, its effects on benign prostatic hyperplasia (BPH), a representative aging-related disease, and the underlying mechanisms remain unclear. This study aims to elucidate the therapeutic effects of KRG on BPH, with a particular focus on mitochondrial dynamics, including fission and fusion processes. The effects of KRG on cell proliferation, apoptosis, and mitochondrial dynamics and morphology were evaluated in a rat model of testosterone propionate (TP)-induced BPH and TP-treated LNCaP cells, with mdivi-1 as a control. The results revealed that KRG treatment reduced the levels of androgen receptors (AR) and prostate-specific antigens in the BPH group. KRG inhibited cell proliferation by downregulating cyclin D and proliferating cell nuclear antigen (PCNA) levels, and it promoted apoptosis by increasing the ratio of B-cell lymphoma protein 2 (Bcl-2)-associated X protein (Bax) to Bcl-2 expression. Notably, KRG treatment enhanced the phosphorylation of dynamin-related protein 1 (DRP-1, serine 637) compared with that in the BPH group, which inhibited mitochondrial fission and led to mitochondrial elongation. This modulation of mitochondrial dynamics was associated with decreased cell proliferation and increased apoptosis. By dysregulating AR signaling and inhibiting mitochondrial fission through enhanced DRP-1 (ser637) phosphorylation, KRG effectively reduced cell proliferation and induced apoptosis. These findings suggest that KRG's regulation of mitochondrial dynamics offers a promising clinical approach for the treatment of BPH.
Collapse
Affiliation(s)
- Geum-Lan Hong
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea; Department of Anatomy, College of Medicine, Konyang University, Daejeon 35365, Korea
| | - Kyung-Hyun Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea
| | - Sung-Pil Cho
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea
| | - Hui-Ju Lee
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea
| | - Yae-Ji Kim
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea
| | - Ju-Young Jung
- Department of Veterinary Medicine & Institute of Veterinary Science, Chungnam National University, Daejeon 34134, Korea.
| |
Collapse
|
30
|
Huang SQ, Cao KX, Wang CL, Chen PL, Chen YX, Zhang YT, Yu SH, Bai ZX, Guo S, Liao MX, Li QW, Zhang GQ, He J, Xu YM. Decreasing mitochondrial fission ameliorates HIF-1α-dependent pathological retinal angiogenesis. Acta Pharmacol Sin 2024; 45:1438-1450. [PMID: 38565961 PMCID: PMC11192750 DOI: 10.1038/s41401-024-01262-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
Angiogenesis plays a critical role in many pathological processes, including irreversible blindness in eye diseases such as retinopathy of prematurity. Endothelial mitochondria are dynamic organelles that undergo constant fusion and fission and are critical signalling hubs that modulate angiogenesis by coordinating reactive oxygen species (ROS) production and calcium signalling and metabolism. In this study, we investigated the role of mitochondrial dynamics in pathological retinal angiogenesis. We showed that treatment with vascular endothelial growth factor (VEGF; 20 ng/ml) induced mitochondrial fission in HUVECs by promoting the phosphorylation of dynamin-related protein 1 (DRP1). DRP1 knockdown or pretreatment with the DRP1 inhibitor Mdivi-1 (5 μM) blocked VEGF-induced cell migration, proliferation, and tube formation in HUVECs. We demonstrated that VEGF treatment increased mitochondrial ROS production in HUVECs, which was necessary for HIF-1α-dependent glycolysis, as well as proliferation, migration, and tube formation, and the inhibition of mitochondrial fission prevented VEGF-induced mitochondrial ROS production. In an oxygen-induced retinopathy (OIR) mouse model, we found that active DRP1 was highly expressed in endothelial cells in neovascular tufts. The administration of Mdivi-1 (10 mg·kg-1·d-1, i.p.) for three days from postnatal day (P) 13 until P15 significantly alleviated pathological angiogenesis in the retina. Our results suggest that targeting mitochondrial fission may be a therapeutic strategy for proliferative retinopathies and other diseases that are dependent on pathological angiogenesis.
Collapse
Affiliation(s)
- Shu-Qi Huang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kai-Xiang Cao
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Cai-Ling Wang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Pei-Ling Chen
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yi-Xin Chen
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yu-Ting Zhang
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shi-Hui Yu
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zai-Xia Bai
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Shuai Guo
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Mu-Xi Liao
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, China
| | - Qiao-Wen Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511520, China
| | - Guo-Qi Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511520, China.
| | - Jun He
- Department of Rehabilitation Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510080, China.
| | - Yi-Ming Xu
- School of Basic Medical Sciences; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
31
|
Liu X, Meng Q, Shi S, Geng X, Wang E, Li Y, Lin F, Liang X, Xi X, Han W, Fan H, Zhou X. Cardiac-derived extracellular vesicles improve mitochondrial function to protect the heart against ischemia/reperfusion injury by delivering ATP5a1. J Nanobiotechnology 2024; 22:385. [PMID: 38951822 PMCID: PMC11218245 DOI: 10.1186/s12951-024-02618-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: 07/27/2023] [Accepted: 02/28/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Numerous studies have confirmed the involvement of extracellular vesicles (EVs) in various physiological processes, including cellular death and tissue damage. Recently, we reported that EVs derived from ischemia-reperfusion heart exacerbate cardiac injury. However, the role of EVs from healthy heart tissue (heart-derived EVs, or cEVs) on myocardial ischemia-reperfusion (MI/R) injury remains unclear. RESULTS Here, we demonstrated that intramyocardial administration of cEVs significantly enhanced cardiac function and reduced cardiac damage in murine MI/R injury models. cEVs treatment effectively inhibited ferroptosis and maintained mitochondrial homeostasis in cardiomyocytes subjected to ischemia-reperfusion injury. Further results revealed that cEVs can transfer ATP5a1 into cardiomyocytes, thereby suppressing mitochondrial ROS production, alleviating mitochondrial damage, and inhibiting cardiomyocyte ferroptosis. Knockdown of ATP5a1 abolished the protective effects of cEVs. Furthermore, we found that the majority of cEVs are derived from cardiomyocytes, and ATP5a1 in cEVs primarily originates from cardiomyocytes of the healthy murine heart. Moreover, we demonstrated that adipose-derived stem cells (ADSC)-derived EVs with ATP5a1 overexpression showed much better efficacy on the therapy of MI/R injury compared to control ADSC-derived EVs. CONCLUSIONS These findings emphasized the protective role of cEVs in cardiac injury and highlighted the therapeutic potential of targeting ATP5a1 as an important approach for managing myocardial damage induced by MI/R injury.
Collapse
Affiliation(s)
- Xuan Liu
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qingshu Meng
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Shanshan Shi
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xuedi Geng
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Enhao Wang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yinzhen Li
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Fang Lin
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoting Liang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Xiaoling Xi
- Department of Heart Failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wei Han
- Department of Heart Failure, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Huimin Fan
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China.
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
- Department of Cardiothoracic Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, School of Medicine, Tongji University, 150 Jimo Rd, Pudong, Shanghai, 200092, China.
- Shanghai Heart Failure Research Center, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
32
|
Blocher R, Liu Y, Patrick T, Polejaeva IA. Cytokine-Supplemented Maturation Medium Enhances Cytoplasmic and Nuclear Maturation in Bovine Oocytes. Animals (Basel) 2024; 14:1837. [PMID: 38929455 PMCID: PMC11200980 DOI: 10.3390/ani14121837] [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: 05/15/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 06/28/2024] Open
Abstract
Bovine in vitro oocyte maturation (IVM) is an easy way to obtain oocytes for subsequent assisted reproductive techniques but is inefficient compared to in vivo maturation. Supplementation of three cytokines, fibroblast growth factor 2 (FGF2), leukemia inhibitory factor (LIF), and insulin-like growth factor 1 (IGF1), or FLI, has increased oocyte maturation and embryo development in multiple species, but studies have not explored the oocyte differences caused by FLI IVM supplementation. This study aimed to assess important nuclear and cytoplasmic maturation events in high-quality oocytes. FLI-supplemented oocytes had a decreased GV (3.0% vs. 13.7%, p < 0.01) and increased telophase I incidence (34.6% vs. 17.6%, p < 0.05) after IVM, increased normal meiotic spindles (68.8% vs. 50.0%, p < 0.001), and an increased nuclear maturation rate (75.1% vs. 66.8%, p < 0.001). Moreover, in metaphase II oocytes, the percentage of FLI-treated oocytes with a diffuse mitochondrial distribution was higher (87.7% vs. 77.5%, p < 0.05) and with a cortical mitochondrial distribution was lower (11.6% vs. 17.4%, p < 0.05). Additionally, FLI-supplemented oocytes had more pattern I cortical granules (21.3% vs. 14.4%, p < 0.05). These data suggest that FLI supplementation in bovine in vitro maturation medium coordinates nuclear and cytoplasmic maturation to produce higher-quality oocytes.
Collapse
Affiliation(s)
| | | | | | - Irina A. Polejaeva
- Animal, Dairy, and Veterinary Sciences Department, Utah State University, 4815 Old Main Hill, Logan, UT 84322, USA; (R.B.); (Y.L.); (T.P.)
| |
Collapse
|
33
|
Divya KP, Kanwar N, Anuranjana PV, Kumar G, Beegum F, George KT, Kumar N, Nandakumar K, Kanwal A. SIRT6 in Regulation of Mitochondrial Damage and Associated Cardiac Dysfunctions: A Possible Therapeutic Target for CVDs. Cardiovasc Toxicol 2024; 24:598-621. [PMID: 38689163 DOI: 10.1007/s12012-024-09858-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 04/05/2024] [Indexed: 05/02/2024]
Abstract
Cardiovascular diseases (CVDs) can be described as a global health emergency imploring possible prevention strategies. Although the pathogenesis of CVDs has been extensively studied, the role of mitochondrial dysfunction in CVD development has yet to be investigated. Diabetic cardiomyopathy, ischemic-reperfusion injury, and heart failure are some of the CVDs resulting from mitochondrial dysfunction Recent evidence from the research states that any dysfunction of mitochondria has an impact on metabolic alteration, eventually causes the death of a healthy cell and therefore, progressively directing to the predisposition of disease. Cardiovascular research investigating the targets that both protect and treat mitochondrial damage will help reduce the risk and increase the quality of life of patients suffering from various CVDs. One such target, i.e., nuclear sirtuin SIRT6 is strongly associated with cardiac function. However, the link between mitochondrial dysfunction and SIRT6 concerning cardiovascular pathologies remains poorly understood. Although the Role of SIRT6 in skeletal muscles and cardiomyocytes through mitochondrial regulation has been well understood, its specific role in mitochondrial maintenance in cardiomyocytes is poorly determined. The review aims to explore the domain-specific function of SIRT6 in cardiomyocytes and is an effort to know how SIRT6, mitochondria, and CVDs are related.
Collapse
Affiliation(s)
- K P Divya
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Navjot Kanwar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab, Technical University, Bathinda, Punjab, 151005, India
| | - P V Anuranjana
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Gautam Kumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
- School of Pharmacy, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Fathima Beegum
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Krupa Thankam George
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Nitesh Kumar
- Department of Pharmacology, National Institute of Pharmaceutical Educations and Research, Hajipur, Bihar, 844102, India
| | - K Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India.
| | - Abhinav Kanwal
- Department of Pharmacology, All India Institute of Medical Sciences, Bathinda, Punjab, 151005, India.
| |
Collapse
|
34
|
Hinton AO, N'jai AU, Vue Z, Wanjalla C. Connection Between HIV and Mitochondria in Cardiovascular Disease and Implications for Treatments. Circ Res 2024; 134:1581-1606. [PMID: 38781302 PMCID: PMC11122810 DOI: 10.1161/circresaha.124.324296] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
HIV infection and antiretroviral therapy alter mitochondrial function, which can progressively lead to mitochondrial damage and accelerated aging. The interaction between persistent HIV reservoirs and mitochondria may provide insight into the relatively high rates of cardiovascular disease and mortality in persons living with HIV. In this review, we explore the intricate relationship between HIV and mitochondrial function, highlighting the potential for novel therapeutic strategies in the context of cardiovascular diseases. We reflect on mitochondrial dynamics, mitochondrial DNA, and mitochondrial antiviral signaling protein in the context of HIV. Furthermore, we summarize how toxicities related to early antiretroviral therapy and current highly active antiretroviral therapy can contribute to mitochondrial dysregulation, chronic inflammation, and poor clinical outcomes. There is a need to understand the mechanisms and develop new targeted therapies. We further consider current and potential future therapies for HIV and their interplay with mitochondria. We reflect on the next-generation antiretroviral therapies and HIV cure due to the direct and indirect effects of HIV persistence, associated comorbidities, coinfections, and the advancement of interdisciplinary research fields. This includes exploring novel and creative approaches to target mitochondria for therapeutic intervention.
Collapse
Affiliation(s)
- Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.O.H., Z.V.)
| | - Alhaji U N'jai
- Biological Sciences, Fourah Bay College and College of Medicine and Allied Health Sciences (COMAHS), University of Sierra Leone, Freetown, Sierra Leone and Koinadugu College, Kabala (A.U.N.)
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN (A.O.H., Z.V.)
| | - Celestine Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN (C.W.)
| |
Collapse
|
35
|
Lee J, Han Y, Kim S, Jo H, Wang W, Cho U, Kim SI, Kim B, Song YS. Mitochondrial fission enhances IL-6-induced metastatic potential in ovarian cancer via ERK1/2 activation. Cancer Sci 2024; 115:1536-1550. [PMID: 38433313 PMCID: PMC11093201 DOI: 10.1111/cas.16064] [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: 05/02/2023] [Revised: 11/22/2023] [Accepted: 12/18/2023] [Indexed: 03/05/2024] Open
Abstract
Ovarian cancer is a lethal gynecologic cancer mostly diagnosed in an advanced stage with an accumulation of ascites. Interleukin-6 (IL-6), a pro-inflammatory cytokine is highly elevated in malignant ascites and plays a pleiotropic role in cancer progression. Mitochondria are dynamic organelles that undergo fission and fusion in response to external stimuli and dysregulation in their dynamics has been implicated in cancer progression and metastasis. Here, we investigate the effect of IL-6 on mitochondrial dynamics in ovarian cancer cells (OVCs) and its impact on metastatic potential. Treatment with IL-6 on ovarian cancer cell lines (SKOV3 and PA-1) led to an elevation in the metastatic potential of OVCs. Interestingly, a positive association was observed between dynamin-related protein 1 (Drp1), a regulator of mitochondrial fission, and IL-6R in metastatic ovarian cancer tissues. Additionally, IL-6 treatment on OVCs was linked to the activation of Drp1, with a notable increase in the ratio of the inhibitory form p-Drp1(S637) to the active form p-Drp1(S616), indicating enhanced mitochondrial fission. Moreover, IL-6 treatment triggered the activation of ERK1/2, and inhibiting ERK1/2 mitigated IL-6-induced mitochondrial fission. Suppressing mitochondrial fission through siRNA transfection and a pharmacological inhibitor reduced the IL-6-induced migration and invasion of OVCs. This was further supported by 3D invasion assays using patient-derived spheroids. Altogether, our study suggests the role of mitochondrial fission in the metastatic potential of OVCs induced by IL-6. The inhibition of mitochondrial fission could be a potential therapeutic approach to suppress the metastasis of ovarian cancer.
Collapse
Affiliation(s)
- Juwon Lee
- WCU Biomodulation, Department of Agricultural BiotechnologySeoul National UniversitySeoulKorea
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
| | - Youngjin Han
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
| | - Soochi Kim
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCaliforniaUSA
- Paul F. Glenn Laboratories for the Biology of AgingStanford University School of MedicineStanfordCaliforniaUSA
| | - HyunA Jo
- WCU Biomodulation, Department of Agricultural BiotechnologySeoul National UniversitySeoulKorea
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
| | - Wenyu Wang
- Department of Medical Oncology, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Untack Cho
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, College of MedicineSeoul National UniversitySeoulKorea
| | - Boyun Kim
- Department of SmartBio, College of Life and Health ScienceKyungsung UniversityBusanKorea
| | - Yong Sang Song
- WCU Biomodulation, Department of Agricultural BiotechnologySeoul National UniversitySeoulKorea
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
- Department of Obstetrics and Gynecology, College of MedicineSeoul National UniversitySeoulKorea
| |
Collapse
|
36
|
Pokharel MD, Garcia-Flores A, Marciano D, Franco MC, Fineman JR, Aggarwal S, Wang T, Black SM. Mitochondrial network dynamics in pulmonary disease: Bridging the gap between inflammation, oxidative stress, and bioenergetics. Redox Biol 2024; 70:103049. [PMID: 38295575 PMCID: PMC10844980 DOI: 10.1016/j.redox.2024.103049] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/16/2024] [Indexed: 02/02/2024] Open
Abstract
Once thought of in terms of bioenergetics, mitochondria are now widely accepted as both the orchestrator of cellular health and the gatekeeper of cell death. The pulmonary disease field has performed extensive efforts to explore the role of mitochondria in regulating inflammation, cellular metabolism, apoptosis, and oxidative stress. However, a critical component of these processes needs to be more studied: mitochondrial network dynamics. Mitochondria morphologically change in response to their environment to regulate these processes through fusion, fission, and mitophagy. This allows mitochondria to adapt their function to respond to cellular requirements, a critical component in maintaining cellular homeostasis. For that reason, mitochondrial network dynamics can be considered a bridge that brings multiple cellular processes together, revealing a potential pathway for therapeutic intervention. In this review, we discuss the critical modulators of mitochondrial dynamics and how they are affected in pulmonary diseases, including chronic obstructive pulmonary disease (COPD), idiopathic pulmonary fibrosis (IPF), acute lung injury (ALI), and pulmonary arterial hypertension (PAH). A dysregulated mitochondrial network plays a crucial role in lung disease pathobiology, and aberrant fission/fusion/mitophagy pathways are druggable processes that warrant further exploration. Thus, we also discuss the candidates for lung disease therapeutics that regulate mitochondrial network dynamics.
Collapse
Affiliation(s)
- Marissa D Pokharel
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Alejandro Garcia-Flores
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA
| | - David Marciano
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Maria C Franco
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Jeffrey R Fineman
- Department of Pediatrics, UC San Francisco, San Francisco, CA, 94143, USA
| | - Saurabh Aggarwal
- Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA
| | - Stephen M Black
- Center for Translational Science, Florida International University, 11350 SW Village Parkway, Port St. Lucie, FL, 34987-2352, USA; Department of Molecular & Cellular Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA; Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
37
|
Mondal NK, Li S, Elsenousi AE, Mattar A, Nordick KV, Lamba HK, Hochman-Mendez C, Rosengart TK, Liao KK. NADPH oxidase overexpression and mitochondrial OxPhos impairment are more profound in human hearts donated after circulatory death than brain death. Am J Physiol Heart Circ Physiol 2024; 326:H548-H562. [PMID: 38180451 DOI: 10.1152/ajpheart.00616.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/06/2024]
Abstract
This study investigated cardiac stress and mitochondrial oxidative phosphorylation (OxPhos) in human donation after circulatory death (DCD) hearts regarding warm ischemic time (WIT) and subsequent cold storage and compared them with that of human brain death donor (DBD) hearts. A total of 24 human hearts were procured for the research study-6 in the DBD group and 18 in the DCD group. DCD group was divided into three groups (n = 6) based on different WITs (20, 40, and 60 min). All hearts received del Nido cardioplegia before being placed in normal saline cold storage for 6 h. Left ventricular biopsies were performed at hours 0, 2, 4, and 6. Cardiac stress [nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits: 47-kDa protein of phagocyte oxidase (p47phox), 91-kDa glycoprotein of phagocyte oxidase (gp91phox)] and mitochondrial oxidative phosphorylation [OxPhos, complex I (NADH dehydrogenase) subunit of ETC (CI)-complex V (ATP synthase) subunit of ETC (CV)] proteins were measured in cardiac tissue and mitochondria respectively. Modulation of cardiac stress and mitochondrial dysfunction were observed in both DCD and DBD hearts. However, DCD hearts suffered more cardiac stress (overexpressed NADPH oxidase subunits) and diminished mitochondrial OxPhos than DBD hearts. The severity of cardiac stress and impaired oxidative phosphorylation in DCD hearts correlated with the longer WIT and subsequent cold storage time. More drastic changes were evident in DCD hearts with a WIT of 60 min or more. Activation of NADPH oxidase via overproduction of p47phox and gp91phox proteins in cardiac tissue may be responsible for cardiac stress leading to diminished mitochondrial oxidative phosphorylation. These protein changes can be used as biomarkers for myocardium damage and might help assess DCD and DBD heart transplant suitability.NEW & NOTEWORTHY First human DCD heart research studied cardiac stress and mitochondrial dysfunction concerning WIT and the efficacy of del Nido cardioplegia as an organ procurement solution and subsequent cold storage. Mild to moderate cardiac stress and mitochondrial dysfunction were noticed in DCD hearts with WIT 20 and 40 min and cold storage for 4 and 2 h, respectively. These changes can serve as biomarkers, allowing interventions to preserve mitochondria and extend WIT in DCD hearts.
Collapse
Affiliation(s)
- Nandan K Mondal
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, Texas, United States
| | - Shiyi Li
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Abdussalam E Elsenousi
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Aladdein Mattar
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Katherine V Nordick
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Harveen K Lamba
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Camila Hochman-Mendez
- Department of Regenerative Medicine Research, Texas Heart Institute, Houston, Texas, United States
| | - Todd K Rosengart
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| | - Kenneth K Liao
- Division of Cardiothoracic Transplantation and Circulatory Support, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
38
|
Yao BF, Luo XJ, Peng J. A review for the correlation between optic atrophy 1-dependent mitochondrial fusion and cardiovascular disorders. Int J Biol Macromol 2024; 254:127910. [PMID: 37939779 DOI: 10.1016/j.ijbiomac.2023.127910] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/19/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Mitochondrial dynamics homeostasis is sustained by continuous and balanced fission and fusion, which are determinants of morphology, abundance, biogenesis and mitophagy of mitochondria. Optic atrophy 1 (OPA1), as the only inner mitochondrial membrane fusion protein, plays a key role in stabilizing mitochondrial dynamics. The disturbance of mitochondrial dynamics contributes to the pathophysiological progress of cardiovascular disorders, which are the main cause of death worldwide in recent decades and result in tremendous social burden. In this review, we describe the latest findings regarding OPA1 and its role in mitochondrial fusion. We summarize the post-translational modifications (PTMs) for OPA1 and its regulatory role in mitochondrial dynamics. Then the diverse cell fates caused by OPA1 expression during cardiovascular disorders are discussed. Moreover, cardiovascular disorders (such as heart failure, myocardial ischemia/reperfusion injury, cardiomyopathy and cardiac hypertrophy) relevant to OPA1-dependent mitochondrial dynamics imbalance have been detailed. Finally, we highlight the potential that targeting OPA1 to impact mitochondrial fusion may be used as a novel strategy against cardiovascular disorders.
Collapse
Affiliation(s)
- Bi-Feng Yao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| |
Collapse
|
39
|
Alalawy AI, Sakran M, Alzuaibr FM, Alotaibi MA, El-Hefnawy ME, Hazazi AY, El-Gendy SM, Aidy EA, Effat H, Ismail DF, Hessien M. Inhibition of Drp1 orchestrates the responsiveness of breast cancer cells to paclitaxel but insignificantly relieves paclitaxel-related ovarian damage in mice. Sci Rep 2023; 13:22782. [PMID: 38129495 PMCID: PMC10739747 DOI: 10.1038/s41598-023-49578-0] [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: 07/08/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
Chemoresistance and chemotherapy-related ovarian damage are well-reported in breast cancer (BC) young patients. Herein, the inhibition of the mitochondrial fission was invested to explore its chemosensitizing role in Paclitaxel (PTX)-resistant cells, and its ability to restore the ovarian integrity in mice receiving PTX or cisplatin chemotherapy. To establish these aims, PTX-resistance was generated in BC cells, which were treated with PTX in combination with Drp1 deficiency, via mdivi-1, or Drp1-specific siRNA transfection. Furthermore, the alterations in the ovarian structure and the endocrine-related hormones were explored in mice receiving repetitive doses of PTX or cisplatin. We found that combining PTX with mdivi-1 improved cell responsiveness to PTX, induced apoptosis- and autophagy-mediated cell death, and relieved cellular oxidative stress. Additionally, the expression of PCNA1 and cyclin B1 genes were downregulated, meanwhile, p53, p21, and mitochondrial fusion proteins (Mfu1&Mfu2) were increased. The in vivo investigations in mice demonstrated that PTX induced gonadotoxic damage similar to cisplatin, whereas dual treatment of mice with PTX+ mdivi-1 failed to restore their normal follicular count and the circulating levels of E2 and AMH hormones. These results suggested that combining Drp1 inhibition with PTX resensitized breast cancer cells to PTX but failed to offer enough protection against chemotherapy-related gonadotoxicity.
Collapse
Affiliation(s)
- Adel I Alalawy
- Department of Biochemistry, Faculty of Science, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Mohamed Sakran
- Department of Biochemistry, Faculty of Science, University of Tabuk, 71491, Tabuk, Saudi Arabia
- Division of Biochemistry, Faculty of Science, Tanta University, Tanta City, 31512, Egypt
| | - Fahad M Alzuaibr
- Biology Department, Faculty of Science, Tabuk University, Tabuk, Saudi Arabia
| | - Maeidh A Alotaibi
- King Faisal Medical Complex Laboratory, Ministry of Health, Taif, Saudi Arabia
| | - Mohamed E El-Hefnawy
- Division of Biochemistry, Faculty of Science, Tanta University, Tanta City, 31512, Egypt
- Department of Chemistry, Rabigh College of Sciences and Arts, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulelah Y Hazazi
- Department of Biochemistry, Faculty of Science, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Saad M El-Gendy
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Esraa A Aidy
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Heba Effat
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Doha F Ismail
- Department of Cancer Biology, National Cancer Institute, Cairo University, Giza, Egypt
| | - Mohamed Hessien
- Division of Biochemistry, Faculty of Science, Tanta University, Tanta City, 31512, Egypt.
- Molecular Cell Biology Unit, Division of Biochemistry, Department of Chemistry, Faculty of Science, Tanta University, Tanta, 31512, Egypt.
| |
Collapse
|
40
|
Jadli A, Gomes K, Ballasy N, Wijesuriya T, Belke D, Fedak P, Patel V. Inhibition of smooth muscle cell death by Angiotensin 1-7 protects against abdominal aortic aneurysm. Biosci Rep 2023; 43:BSR20230718. [PMID: 37947205 PMCID: PMC10695742 DOI: 10.1042/bsr20230718] [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: 04/29/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) represents a debilitating vascular disease characterized by aortic dilatation and wall rupture if it remains untreated. We aimed to determine the effects of Ang 1-7 in a murine model of AAA and to investigate the molecular mechanisms involved. Eight- to 10-week-old apolipoprotein E-deficient mice (ApoEKO) were infused with Ang II (1.44 mg/kg/day, s.c.) and treated with Ang 1-7 (0.576 mg/kg/day, i.p.). Echocardiographic and histological analyses showed abdominal aortic dilatation and extracellular matrix remodeling in Ang II-infused mice. Treatment with Ang 1-7 led to suppression of Ang II-induced aortic dilatation in the abdominal aorta. The immunofluorescence imaging exhibited reduced smooth muscle cell (SMC) density in the abdominal aorta. The abdominal aortic SMCs from ApoEKO mice exhibited markedly increased apoptosis in response to Ang II. Ang 1-7 attenuated cell death, as evident by increased SMC density in the aorta and reduced annexin V/propidium iodide-positive cells in flow cytometric analysis. Gene expression analysis for contractile and synthetic phenotypes of abdominal SMCs showed preservation of contractile phenotype by Ang 1-7 treatment. Molecular analyses identified increased mitochondrial fission, elevated cellular and mitochondrial reactive oxygen species (ROS) levels, and apoptosis-associated proteins, including cytochrome c, in Ang II-treated aortic SMCs. Ang 1-7 mitigated Ang II-induced mitochondrial fission, ROS generation, and levels of pro-apoptotic proteins, resulting in decreased cell death of aortic SMCs. These results highlight a critical vasculo-protective role of Ang 1-7 in a degenerative aortic disease; increased Ang 1-7 activity may provide a promising therapeutic strategy against the progression of AAA.
Collapse
Affiliation(s)
- Anshul S. Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Karina P. Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Noura N. Ballasy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Tishani Methsala Wijesuriya
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Darrell Belke
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W.M. Fedak
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Vaibhav B. Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
41
|
Atici AE, Crother TR, Noval Rivas M. Mitochondrial quality control in health and cardiovascular diseases. Front Cell Dev Biol 2023; 11:1290046. [PMID: 38020895 PMCID: PMC10657886 DOI: 10.3389/fcell.2023.1290046] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Cardiovascular diseases (CVDs) are one of the primary causes of mortality worldwide. An optimal mitochondrial function is central to supplying tissues with high energy demand, such as the cardiovascular system. In addition to producing ATP as a power source, mitochondria are also heavily involved in adaptation to environmental stress and fine-tuning tissue functions. Mitochondrial quality control (MQC) through fission, fusion, mitophagy, and biogenesis ensures the clearance of dysfunctional mitochondria and preserves mitochondrial homeostasis in cardiovascular tissues. Furthermore, mitochondria generate reactive oxygen species (ROS), which trigger the production of pro-inflammatory cytokines and regulate cell survival. Mitochondrial dysfunction has been implicated in multiple CVDs, including ischemia-reperfusion (I/R), atherosclerosis, heart failure, cardiac hypertrophy, hypertension, diabetic and genetic cardiomyopathies, and Kawasaki Disease (KD). Thus, MQC is pivotal in promoting cardiovascular health. Here, we outline the mechanisms of MQC and discuss the current literature on mitochondrial adaptation in CVDs.
Collapse
Affiliation(s)
- Asli E. Atici
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Timothy R. Crother
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
42
|
Li AL, Lian L, Chen XN, Cai WH, Fan XB, Fan YJ, Li TT, Xie YY, Zhang JP. The role of mitochondria in myocardial damage caused by energy metabolism disorders: From mechanisms to therapeutics. Free Radic Biol Med 2023; 208:236-251. [PMID: 37567516 DOI: 10.1016/j.freeradbiomed.2023.08.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023]
Abstract
Myocardial damage is the most serious pathological consequence of cardiovascular diseases and an important reason for their high mortality. In recent years, because of the high prevalence of systemic energy metabolism disorders (e.g., obesity, diabetes mellitus, and metabolic syndrome), complications of myocardial damage caused by these disorders have attracted widespread attention. Energy metabolism disorders are independent of traditional injury-related risk factors, such as ischemia, hypoxia, trauma, and infection. An imbalance of myocardial metabolic flexibility and myocardial energy depletion are usually the initial changes of myocardial injury caused by energy metabolism disorders, and abnormal morphology and functional destruction of the mitochondria are their important features. Specifically, mitochondria are the centers of energy metabolism, and recent evidence has shown that decreased mitochondrial function, caused by an imbalance in mitochondrial quality control, may play a key role in myocardial injury caused by energy metabolism disorders. Under chronic energy stress, mitochondria undergo pathological fission, while mitophagy, mitochondrial fusion, and biogenesis are inhibited, and mitochondrial protein balance and transfer are disturbed, resulting in the accumulation of nonfunctional and damaged mitochondria. Consequently, damaged mitochondria lead to myocardial energy depletion and the accumulation of large amounts of reactive oxygen species, further aggravating the imbalance in mitochondrial quality control and forming a vicious cycle. In addition, impaired mitochondria coordinate calcium homeostasis imbalance, and epigenetic alterations participate in the pathogenesis of myocardial damage. These pathological changes induce rapid progression of myocardial damage, eventually leading to heart failure or sudden cardiac death. To intervene more specifically in the myocardial damage caused by metabolic disorders, we need to understand the specific role of mitochondria in this context in detail. Accordingly, promising therapeutic strategies have been proposed. We also summarize the existing therapeutic strategies to provide a reference for clinical treatment and developing new therapies.
Collapse
Affiliation(s)
- Ao-Lin Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Lu Lian
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xin-Nong Chen
- Department of Traditional Chinese Medicine, Tianjin First Central Hospital, Tianjin, 300190, China
| | - Wen-Hui Cai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Xin-Biao Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ya-Jie Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ting-Ting Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China; Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Ying-Yu Xie
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
| | - Jun-Ping Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300183, China.
| |
Collapse
|
43
|
Park JS, Lee C, Cheon SY, Lee Y, Jeon H, Lee D, Kim SH, Lim SG, Koo H. Efficient drug supply in stem cell cytosol via pore-forming saponin nanoparticles promotes in vivo osteogenesis and bone regeneration. Biomaterials 2023; 302:122342. [PMID: 37804721 DOI: 10.1016/j.biomaterials.2023.122342] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/22/2023] [Accepted: 09/27/2023] [Indexed: 10/09/2023]
Abstract
Directional differentiation of stem cells is a key step in stem cell therapy. In this study, we developed saponin-based nanoparticles (Ad-SNPs) containing dexamethasone (Dex) and alpha-lipoic acid (ALA) to promote osteogenic differentiation of human mesenchymal stem cells (hMSCs) and bone regeneration. The Ad-SNPs can achieve rapid cellular uptake through a pore-forming effect without cytotoxic cationic charges. They also provide extended retention in cell cytosol due to their uptake route. These properties are advantageous in efficiently supplying drugs to the hMSCs. The combination of Dex and ALA facilitated mitochondrial fusion and prevented reactive oxygen species-induced DNA damage. It also helped to preserve mitochondrial dynamics, and the efficient supply of it provided by the Ad-SNPs induced differentiation of hMSCs into osteoblasts. The Ad-SNPs showed outstanding performance in osteoblast differentiation, maturation, and mineralization in 3D culture compared with NPs without saponin and with free drugs. When Ad-SNP-treated hMSCs were tested in a rat femoral bone defect model, they showed the fastest regeneration of bones and complete repair in the shortest period among all groups. To the best of our knowledge, this study is the first application of pore-forming saponin-based NPs with rapid cellular uptake and extended retention to stem cell therapy, and we demonstrated their promising potential in bone regeneration and efficient delivery of Dex and ALA.
Collapse
Affiliation(s)
- Ji Sun Park
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Chaehyun Lee
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Seo Young Cheon
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Yeeun Lee
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Hayoung Jeon
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Se Hee Kim
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Seong Gi Lim
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.
| |
Collapse
|
44
|
Yin L, Qi S, Zhu Z. Advances in mitochondria-centered mechanism behind the roles of androgens and androgen receptor in the regulation of glucose and lipid metabolism. Front Endocrinol (Lausanne) 2023; 14:1267170. [PMID: 37900128 PMCID: PMC10613047 DOI: 10.3389/fendo.2023.1267170] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023] Open
Abstract
An increasing number of studies have reported that androgens and androgen receptors (AR) play important roles in the regulation of glucose and lipid metabolism. Impaired glucose and lipid metabolism and the development of obesity-related diseases have been found in either hypogonadal men or male rodents with androgen deficiency. Exogenous androgens supplementation can effectively improve these disorders, but the mechanism by which androgens regulate glucose and lipid metabolism has not been fully elucidated. Mitochondria, as powerhouses within cells, are key organelles influencing glucose and lipid metabolism. Evidence from both pre-clinical and clinical studies has reported that the regulation of glucose and lipid metabolism by androgens/AR is strongly associated with the impact on the content and function of mitochondria, but few studies have systematically reported the regulatory effect and the molecular mechanism. In this paper, we review the effect of androgens/AR on mitochondrial content, morphology, quality control system, and function, with emphases on molecular mechanisms. Additionally, we discuss the sex-dimorphic effect of androgens on mitochondria. This paper provides a theoretical basis for shedding light on the influence and mechanism of androgens on glucose and lipid metabolism and highlights the mitochondria-based explanation for the sex-dimorphic effect of androgens on glucose and lipid metabolism.
Collapse
Affiliation(s)
- Lijun Yin
- School of Sport, Shenzhen University, Shenzhen, China
| | - Shuo Qi
- School of Sport Health, Shandong Sport University, Jinan, China
| | - Zhiqiang Zhu
- School of Sport, Shenzhen University, Shenzhen, China
| |
Collapse
|
45
|
Miao L, Liu YL. Research Advances in Targeted Therapy for Heart Failure. Rev Cardiovasc Med 2023; 24:276. [PMID: 39077559 PMCID: PMC11262442 DOI: 10.31083/j.rcm2410276] [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: 02/21/2023] [Revised: 04/16/2023] [Accepted: 05/17/2023] [Indexed: 07/31/2024] Open
Abstract
Cardiovascular disease is one of the major diseases threatening the health of Chinese residents, and the death rate has long been the highest on the disease spectrum in China. With the progress of population aging, the prevalence and mortality of cardiovascular diseases remain on the rise, and the current treatment effect on and prognosis of heart failure (HF) are not satisfactory. It is particularly important to explore the potential pathogenic mechanisms of HF and identify new therapeutic targets.
Collapse
Affiliation(s)
- Liu Miao
- Department of Cardiology, Liuzhou People's Hospital, Affiliated of Guangxi
Medical University, 545006 Liuzhou, Guangxi, China
| | - Yan-Li Liu
- Department of Cardiology, Liuzhou People's Hospital, Affiliated of Guangxi
Medical University, 545006 Liuzhou, Guangxi, China
| |
Collapse
|
46
|
Ge J, Shelby SL, Wang Y, Morse PD, Coffey K, Li J, Geng T, Huang Y. Cardioprotective properties of quercetin in fescue toxicosis-induced cardiotoxicity via heart-gut axis in lambs (Ovis Aries). JOURNAL OF HAZARDOUS MATERIALS 2023; 458:131843. [PMID: 37379607 DOI: 10.1016/j.jhazmat.2023.131843] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/31/2023] [Accepted: 06/11/2023] [Indexed: 06/30/2023]
Abstract
The present study investigated whether quercetin mitigated fescue toxicosis-induced cardiovascular injury via the heart-gut axis. Twenty-four commercial Dorper lambs were stratified by body weight and assigned randomly to diets in one of four groups: endophyte-free without quercetin (E-,Q-), endophyte-positive without quercetin (E+,Q-), endophyte-positive plus 4 g/kg quercetin (E+,Q+) or endophyte-free plus 4 g/kg quercetin (E-,Q+) for 42 days. Body weight and average daily feed intake (ADFI) of lambs fed the endophyte-positive diets showed significant decreases. However, in the groups treated with quercetin, there were significant alterations of cardiac enzymes. Furthermore, reduced fescue toxicosis-induced histopathological lesions of heart and aorta were demonstrated in the E+,Q+ lambs. Results also suggested quercetin eased cardiovascular oxidative injury by inhibiting the increase of oxidative metabolites, and enhancing the levels of antioxidases. Quercetin reduced the inflammation response through suppressing NF-κB signaling pathway activation. Additionally, quercetin ameliorated fescue toxicosis-induced mitochondria dysfunction and improved mitochondrial quality control through enhancing PGC-1α-mediated mitochondrial biogenesis, maintaining the mitochondrial dynamics, and relieving aberrant Parkin/PINK-mediated mitophagy. Quercetin enhanced gastrointestinal microbial alpha and beta diversity, alleviated gut microbiota and microbiome derived metabolites-SCFAs dysbiosis by fescue toxicosis. These findings signified that quercetin may play a cardio-protective role via regulating the heart-gut microbiome axis.
Collapse
Affiliation(s)
- Jing Ge
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, United States
| | - Sarah Layne Shelby
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, United States
| | - Yongjie Wang
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, United States
| | - Palika Dias Morse
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, United States
| | - Ken Coffey
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, United States
| | - Jinlong Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, PR China, Northeast Agricultural University, Harbin 150030, PR China
| | - Tuoyu Geng
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, PR China; Joint International Research Laboratory of Agriculture and Agri-Product Safety of the Ministry of Education of China, Yangzhou 225009, PR China.
| | - Yan Huang
- Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, United States.
| |
Collapse
|
47
|
Kubat GB, Bouhamida E, Ulger O, Turkel I, Pedriali G, Ramaccini D, Ekinci O, Ozerklig B, Atalay O, Patergnani S, Nur Sahin B, Morciano G, Tuncer M, Tremoli E, Pinton P. Mitochondrial dysfunction and skeletal muscle atrophy: Causes, mechanisms, and treatment strategies. Mitochondrion 2023; 72:33-58. [PMID: 37451353 DOI: 10.1016/j.mito.2023.07.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 07/02/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Skeletal muscle, which accounts for approximately 40% of total body weight, is one of the most dynamic and plastic tissues in the human body and plays a vital role in movement, posture and force production. More than just a component of the locomotor system, skeletal muscle functions as an endocrine organ capable of producing and secreting hundreds of bioactive molecules. Therefore, maintaining healthy skeletal muscles is crucial for supporting overall body health. Various pathological conditions, such as prolonged immobilization, cachexia, aging, drug-induced toxicity, and cardiovascular diseases (CVDs), can disrupt the balance between muscle protein synthesis and degradation, leading to skeletal muscle atrophy. Mitochondrial dysfunction is a major contributing mechanism to skeletal muscle atrophy, as it plays crucial roles in various biological processes, including energy production, metabolic flexibility, maintenance of redox homeostasis, and regulation of apoptosis. In this review, we critically examine recent knowledge regarding the causes of muscle atrophy (disuse, cachexia, aging, etc.) and its contribution to CVDs. Additionally, we highlight the mitochondrial signaling pathways involvement to skeletal muscle atrophy, such as the ubiquitin-proteasome system, autophagy and mitophagy, mitochondrial fission-fusion, and mitochondrial biogenesis. Furthermore, we discuss current strategies, including exercise, mitochondria-targeted antioxidants, in vivo transfection of PGC-1α, and the potential use of mitochondrial transplantation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Gokhan Burcin Kubat
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey.
| | - Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Oner Ulger
- Department of Mitochondria and Cellular Research, Gulhane Health Sciences Institute, University of Health Sciences, 06010 Ankara, Turkey
| | - Ibrahim Turkel
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Ozgur Ekinci
- Department of Pathology, Gazi University, 06500 Ankara, Turkey
| | - Berkay Ozerklig
- Department of Exercise and Sport Sciences, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey
| | - Ozbeyen Atalay
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Beyza Nur Sahin
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Meltem Tuncer
- Department of Physiology, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy; Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
48
|
Cao X, Yao F, Zhang B, Sun X. Mitochondrial dysfunction in heart diseases: Potential therapeutic effects of Panax ginseng. Front Pharmacol 2023; 14:1218803. [PMID: 37547332 PMCID: PMC10399631 DOI: 10.3389/fphar.2023.1218803] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/12/2023] [Indexed: 08/08/2023] Open
Abstract
Heart diseases have a high incidence and mortality rate, and seriously affect people's quality of life. Mitochondria provide energy for the heart to function properly. The process of various heart diseases is closely related to mitochondrial dysfunction. Panax ginseng (P. ginseng), as a traditional Chinese medicine, is widely used to treat various cardiovascular diseases. Many studies have confirmed that P. ginseng and ginsenosides can regulate and improve mitochondrial dysfunction. Therefore, the role of mitochondria in various heart diseases and the protective effect of P. ginseng on heart diseases by regulating mitochondrial function were reviewed in this paper, aiming to gain new understanding of the mechanisms, and promote the clinical application of P. ginseng.
Collapse
Affiliation(s)
- Xinxin Cao
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Fan Yao
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
49
|
Colpman P, Dasgupta A, Archer SL. The Role of Mitochondrial Dynamics and Mitotic Fission in Regulating the Cell Cycle in Cancer and Pulmonary Arterial Hypertension: Implications for Dynamin-Related Protein 1 and Mitofusin2 in Hyperproliferative Diseases. Cells 2023; 12:1897. [PMID: 37508561 PMCID: PMC10378656 DOI: 10.3390/cells12141897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/14/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Mitochondria, which generate ATP through aerobic respiration, also have important noncanonical functions. Mitochondria are dynamic organelles, that engage in fission (division), fusion (joining) and translocation. They also regulate intracellular calcium homeostasis, serve as oxygen-sensors, regulate inflammation, participate in cellular and organellar quality control and regulate the cell cycle. Mitochondrial fission is mediated by the large GTPase, dynamin-related protein 1 (Drp1) which, when activated, translocates to the outer mitochondrial membrane (OMM) where it interacts with binding proteins (Fis1, MFF, MiD49 and MiD51). At a site demarcated by the endoplasmic reticulum, fission proteins create a macromolecular ring that divides the organelle. The functional consequence of fission is contextual. Physiological fission in healthy, nonproliferating cells mediates organellar quality control, eliminating dysfunctional portions of the mitochondria via mitophagy. Pathological fission in somatic cells generates reactive oxygen species and triggers cell death. In dividing cells, Drp1-mediated mitotic fission is critical to cell cycle progression, ensuring that daughter cells receive equitable distribution of mitochondria. Mitochondrial fusion is regulated by the large GTPases mitofusin-1 (Mfn1) and mitofusin-2 (Mfn2), which fuse the OMM, and optic atrophy 1 (OPA-1), which fuses the inner mitochondrial membrane. Mitochondrial fusion mediates complementation, an important mitochondrial quality control mechanism. Fusion also favors oxidative metabolism, intracellular calcium homeostasis and inhibits cell proliferation. Mitochondrial lipids, cardiolipin and phosphatidic acid, also regulate fission and fusion, respectively. Here we review the role of mitochondrial dynamics in health and disease and discuss emerging concepts in the field, such as the role of central versus peripheral fission and the potential role of dynamin 2 (DNM2) as a fission mediator. In hyperproliferative diseases, such as pulmonary arterial hypertension and cancer, Drp1 and its binding partners are upregulated and activated, positing mitochondrial fission as an emerging therapeutic target.
Collapse
Affiliation(s)
- Pierce Colpman
- Department of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Asish Dasgupta
- Department of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, ON K7L 3N6, Canada
| |
Collapse
|
50
|
Marginean CM, Pirscoveanu D, Popescu M, Vasile CM, Docea AO, Mitruț R, Mărginean IC, Iacob GA, Firu DM, Mitruț P. Challenges in Diagnosis and Therapeutic Approach of Acute on Chronic Liver Failure-A Review of Current Evidence. Biomedicines 2023; 11:1840. [PMID: 37509478 PMCID: PMC10376368 DOI: 10.3390/biomedicines11071840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/23/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Acute-on-chronic liver failure (ACLF) is a syndrome characterized by acute and severe decompensation of chronic liver disease (CLD) correlated with multiple organ failure, poor prognosis, and increased mortality. In 40-50% of ACLF cases, the trigger is not recognized; for many of these patients, bacterial translocation associated with systemic inflammation is thought to be the determining factor; in the other 50% of patients, sepsis, alcohol consumption, and reactivation of chronic viral hepatitis are the most frequently described trigger factors. Other conditions considered precipitating factors are less common, including acute alcoholic hepatitis, major surgery, TIPS insertion, or inadequate paracentesis without albumin substitution. Host response is likely the primary factor predicting ACLF severity and prognosis, the host immune response having a particular significance in this syndrome, together with the inflammatory cascade. The management of ACLF includes both the prevention of the precipitating factors that lead to acute liver decompensation and the support of vital functions, the prevention and management of complications, the estimation of prognosis, and the opportunity for liver transplantation.
Collapse
Affiliation(s)
- Cristina Maria Marginean
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Denisa Pirscoveanu
- Department of Neurology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Mihaela Popescu
- Department of Endocrinology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Corina Maria Vasile
- Department of Pediatric Cardiology, "Marie Curie" Emergency Children's Hospital, 041451 Bucharest, Romania
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Radu Mitruț
- Department of Cardiology, University and Emergency Hospital, 050098 Bucharest, Romania
| | | | - George Alexandru Iacob
- Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Dan Mihai Firu
- Ph.D. School Department, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Paul Mitruț
- Department of Internal Medicine, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|