1
|
Li H, Dai X, Zhou J, Wang Y, Zhang S, Guo J, Shen L, Yan H, Jiang H. Mitochondrial dynamics in pulmonary disease: Implications for the potential therapeutics. J Cell Physiol 2024; 239:e31370. [PMID: 38988059 DOI: 10.1002/jcp.31370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Mitochondria are dynamic organelles that continuously undergo fusion/fission to maintain normal cell physiological activities and energy metabolism. When mitochondrial dynamics is unbalanced, mitochondrial homeostasis is broken, thus damaging mitochondrial function. Accumulating evidence demonstrates that impairment in mitochondrial dynamics leads to lung tissue injury and pulmonary disease progression in a variety of disease models, including inflammatory responses, apoptosis, and barrier breakdown, and that the role of mitochondrial dynamics varies among pulmonary diseases. These findings suggest that modulation of mitochondrial dynamics may be considered as a valid therapeutic strategy in pulmonary diseases. In this review, we discuss the current evidence on the role of mitochondrial dynamics in pulmonary diseases, with a particular focus on its underlying mechanisms in the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis (PF), pulmonary arterial hypertension (PAH), lung cancer and bronchopulmonary dysplasia (BPD), and outline effective drugs targeting mitochondrial dynamics-related proteins, highlighting the great potential of targeting mitochondrial dynamics in the treatment of pulmonary disease.
Collapse
Affiliation(s)
- Hui Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinyan Dai
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
2
|
Sinha JK, Jorwal K, Singh KK, Han SS, Bhaskar R, Ghosh S. The Potential of Mitochondrial Therapeutics in the Treatment of Oxidative Stress and Inflammation in Aging. Mol Neurobiol 2024:10.1007/s12035-024-04474-0. [PMID: 39230868 DOI: 10.1007/s12035-024-04474-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
Mitochondria are central to cellular energy production, and their dysfunction is a major contributor to oxidative stress and chronic inflammation, pivotal factors in aging, and related diseases. With aging, mitochondrial efficiency declines, leading to an increase in ROS and persistent inflammatory responses. Therapeutic interventions targeting mitochondrial health show promise in mitigating these detrimental effects. Antioxidants such as MitoQ and MitoVitE, and supplements like coenzyme Q10 and NAD + precursors, have demonstrated potential in reducing oxidative stress. Additionally, gene therapy aimed at enhancing mitochondrial function, alongside lifestyle modifications such as regular exercise and caloric restriction can ameliorate age-related mitochondrial decline. Exercise not only boosts mitochondrial biogenesis but also improves mitophagy. Enhancing mitophagy is a key strategy to prevent the accumulation of dysfunctional mitochondria, which is crucial for cellular homeostasis and longevity. Pharmacological agents like sulforaphane, SS-31, and resveratrol indirectly promote mitochondrial biogenesis and improve cellular resistance to oxidative damage. The exploration of mitochondrial therapeutics, including emerging techniques like mitochondrial transplantation, offers significant avenues for extending health span and combating age-related diseases. However, translating these findings into clinical practice requires overcoming challenges in precisely targeting dysfunctional mitochondria and optimizing delivery mechanisms for therapeutic agents. Continued research is essential to refine these approaches and fully understand the interplay between mitochondrial dynamics and aging.
Collapse
Affiliation(s)
| | - Khanak Jorwal
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh, 201301, India
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology, Symbiosis International (Deemed University), Pune, Maharashtra, 411057, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang, 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeonsang, 38541, Republic of Korea
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang, 38541, Republic of Korea.
- Research Institute of Cell Culture, Yeungnam University, Gyeonsang, 38541, Republic of Korea.
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh, 201301, India.
| |
Collapse
|
3
|
Choi EH, Kim MH, Park SJ. Targeting Mitochondrial Dysfunction and Reactive Oxygen Species for Neurodegenerative Disease Treatment. Int J Mol Sci 2024; 25:7952. [PMID: 39063194 PMCID: PMC11277296 DOI: 10.3390/ijms25147952] [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: 06/30/2024] [Revised: 07/18/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common neurodegenerative diseases, and they affect millions of people worldwide, particularly older individuals. Therefore, there is a clear need to develop novel drug targets for the treatment of age-related neurodegenerative diseases. Emerging evidence suggests that mitochondrial dysfunction and reactive oxygen species (ROS) generation play central roles in the onset and progression of neurodegenerative diseases. Mitochondria are key regulators of respiratory function, cellular energy adenosine triphosphate production, and the maintenance of cellular redox homeostasis, which are essential for cell survival. Mitochondrial morphology and function are tightly regulated by maintaining a balance among mitochondrial fission, fusion, biogenesis, and mitophagy. In this review, we provide an overview of the main functions of mitochondria, with a focus on recent progress highlighting the critical role of ROS-induced oxidative stress, dysregulated mitochondrial dynamics, mitochondrial apoptosis, mitochondria-associated inflammation, and impaired mitochondrial function in the pathogenesis of age-related neurodegenerative diseases, such as AD and PD. We also discuss the potential of mitochondrial fusion and biogenesis enhancers, mitochondrial fission inhibitors, and mitochondria-targeted antioxidants as novel drugs for the treatment of these diseases.
Collapse
Affiliation(s)
| | | | - Sun-Ji Park
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Republic of Korea; (E.-H.C.); (M.-H.K.)
| |
Collapse
|
4
|
Liu Q, Zhao JN, Fang ZT, Wang X, Zhang BG, He Y, Liu RJ, Chen J, Liu GP. BGP-15 alleviates LPS-induced depression-like behavior by promoting mitophagy. Brain Behav Immun 2024; 119:648-664. [PMID: 38677623 DOI: 10.1016/j.bbi.2024.04.036] [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: 12/19/2023] [Revised: 03/22/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024] Open
Abstract
The high prevalence of major depressive disorder (MDD) frequently imposes severe constraints on psychosocial functioning and detrimentally impacts overall well-being. Despite the growing interest in the hypothesis of mitochondrial dysfunction, the precise mechanistic underpinnings and therapeutic strategies remain unclear and require further investigation. In this study, an MDD model was established in mice using lipopolysaccharide (LPS). Our research findings demonstrated that LPS exposure induced depressive-like behaviors and disrupted mitophagy by diminishing the mitochondrial levels of PINK1/Parkin in the brains of mice. Furthermore, LPS exposure evoked the activation of the NLRP3 inflammasome, accompanied by a notable elevation in the concentrations of pro-inflammatory factors (TNF-α, IL-1β, and IL-6). Additionally, neuronal apoptosis was stimulated through the JNK/p38 pathway. The administration of BGP-15 effectively nullified the impact of LPS, corresponding to the amelioration of depressive-like phenotypes and restoration of mitophagy, prevention of neuronal injury and inflammation, and suppression of reactive oxygen species (ROS)-mediated NLRP3 inflammasome activation. Furthermore, we elucidated the involvement of mitophagy in BGP-15-attenuated depressive-like behaviors using the inhibitors targeting autophagy (3-MA) and mitophagy (Mdivi-1). Notably, these inhibitors notably counteracted the antidepressant and anti-inflammatory effects exerted by BGP-15. Based on the research findings, it can be inferred that the antidepressant properties of BGP-15 in LPS-induced depressive-like behaviors could potentially be attributed to the involvement of the mitophagy pathway. These findings offer a potential novel therapeutic strategy for managing MDD.
Collapse
Affiliation(s)
- Qian Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun-Ning Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhi-Ting Fang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Bing-Ge Zhang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ye He
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rui-Juan Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jian Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
5
|
Liu BH, Xu CZ, Liu Y, Lu ZL, Fu TL, Li GR, Deng Y, Luo GQ, Ding S, Li N, Geng Q. Mitochondrial quality control in human health and disease. Mil Med Res 2024; 11:32. [PMID: 38812059 PMCID: PMC11134732 DOI: 10.1186/s40779-024-00536-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 05/07/2024] [Indexed: 05/31/2024] Open
Abstract
Mitochondria, the most crucial energy-generating organelles in eukaryotic cells, play a pivotal role in regulating energy metabolism. However, their significance extends beyond this, as they are also indispensable in vital life processes such as cell proliferation, differentiation, immune responses, and redox balance. In response to various physiological signals or external stimuli, a sophisticated mitochondrial quality control (MQC) mechanism has evolved, encompassing key processes like mitochondrial biogenesis, mitochondrial dynamics, and mitophagy, which have garnered increasing attention from researchers to unveil their specific molecular mechanisms. In this review, we present a comprehensive summary of the primary mechanisms and functions of key regulators involved in major components of MQC. Furthermore, the critical physiological functions regulated by MQC and its diverse roles in the progression of various systemic diseases have been described in detail. We also discuss agonists or antagonists targeting MQC, aiming to explore potential therapeutic and research prospects by enhancing MQC to stabilize mitochondrial function.
Collapse
Affiliation(s)
- Bo-Hao Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, 130021, China
| | - Chen-Zhen Xu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yi Liu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zi-Long Lu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ting-Lv Fu
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Rui Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yu Deng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Guo-Qing Luo
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Song Ding
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
6
|
Zhu JY, van de Leemput J, Han Z. Promoting mitochondrial dynamics by inhibiting the PINK1-PRKN pathway to relieve diabetic nephropathy. Dis Model Mech 2024; 17:dmm050471. [PMID: 38602042 PMCID: PMC11095637 DOI: 10.1242/dmm.050471] [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: 08/23/2023] [Accepted: 03/28/2024] [Indexed: 04/12/2024] Open
Abstract
Diabetes is a metabolic disorder characterized by high blood glucose levels and is a leading cause of kidney disease. Diabetic nephropathy has been attributed to dysfunctional mitochondria. However, many questions remain about the exact mechanism. The structure, function and molecular pathways are highly conserved between mammalian podocytes and Drosophila nephrocytes; therefore, we used flies on a high-sucrose diet to model type 2 diabetic nephropathy. The nephrocytes from flies on a high-sucrose diet showed a significant functional decline and decreased cell size, associated with a shortened lifespan. Structurally, the nephrocyte filtration structure, known as the slit diaphragm, was disorganized. At the cellular level, we found altered mitochondrial dynamics and dysfunctional mitochondria. Regulating mitochondrial dynamics by either genetic modification of the Pink1-Park (mammalian PINK1-PRKN) pathway or treatment with BGP-15, mitigated the mitochondrial defects and nephrocyte functional decline. These findings support a role for Pink1-Park-mediated mitophagy and associated control of mitochondrial dynamics in diabetic nephropathy, and demonstrate that targeting this pathway might provide therapeutic benefits for type 2 diabetic nephropathy.
Collapse
Affiliation(s)
- Jun-yi Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes, and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
7
|
Zhang W, Li M, Ye X, Jiang M, Wu X, Tang Z, Hu L, Zhang H, Li Y, Pan J. Disturbance of mitochondrial dynamics in myocardium of broilers with pulmonary hypertension syndrome. Br Poult Sci 2024; 65:154-164. [PMID: 38380624 DOI: 10.1080/00071668.2024.2308277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 12/05/2023] [Indexed: 02/22/2024]
Abstract
1. The following study investigated the relationship between pulmonary hypertension syndrome (PHS) and mitochondrial dynamics in broiler cardiomyocytes.2. An animal model for PHS was established by injecting broiler chickens with CM-32 cellulose particles. Broiler myocardial cells were cultured under hypoxic conditions to establish an in vitro model. The ascites heart index, histomorphology, mitochondrial ultrastructure, and mitochondrial dynamic-related gene and protein expression were evaluated.3. The myocardial fibres from PHS broilers had wider spaces and were wavy and twisted and the number of mitochondria increased. Compared with the control group, the gene and protein expression levels were decreased for Opa1, Mfn1, and Mfn2 in the myocardium of PHS broilers. The gene and protein expression was significantly increased for Drp1 and Mff.4. This study showed that PHS in broilers may cause myocardial mitochondrial dysfunction, specifically by diminishing mitochondrial fusion and enhancing fission, causing disturbances in the mitochondrial dynamics of the heart.
Collapse
Affiliation(s)
- W Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - M Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - X Ye
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - M Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - X Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - Z Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - L Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - H Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - Y Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| | - J Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
8
|
Zhang JF, Fang ZT, Zhao JN, Liu GP, Shen X, Jiang GF, Liu Q. Acetylated tau exacerbates apoptosis by disturbing mitochondrial dynamics in HEK293 cells. J Neurochem 2024; 168:288-302. [PMID: 38275215 DOI: 10.1111/jnc.16053] [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: 08/14/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/27/2024]
Abstract
An increase in tau acetylation at K274 and K281 and abnormal mitochondrial dynamics have been observed in the brains of Alzheimer's disease (AD) patients. Here, we constructed three types of tau plasmids, TauKQ (acetylated tau mutant, by mutating its K274/K281 into glutamine to mimic disease-associated lysine acetylation), TauKR (non-acetylated tau mutant, by mutating its K274/K281 into arginine), and TauWT (wild-type human full-length tau). By transfecting these tau plasmids in HEK293 cells, we found that TauWT and TauKR induced mitochondrial fusion by increasing the level of mitochondrial fusion proteins. Conversely, TauKQ induced mitochondrial fission by reducing mitochondrial fusion proteins, exacerbating mitochondrial dysfunction and apoptosis. BGP-15 ameliorated TauKQ-induced mitochondrial dysfunction and apoptosis by improving mitochondrial dynamics. Our findings suggest that acetylation of K274/281 represents an important post-translational modification site regulating mitochondrial dynamics, and that BGP-15 holds potential as a therapeutic agent for mitochondria-associated diseases such as AD.
Collapse
Affiliation(s)
- Jun-Fei Zhang
- Department of Pathology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhi-Ting Fang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jun-Ning Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xin Shen
- School of Stomatology and Ophthalmology, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Gao-Feng Jiang
- Center for Translational Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Qian Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
9
|
Huang Y, Ji W, Zhang J, Huang Z, Ding A, Bai H, Peng B, Huang K, Du W, Zhao T, Li L. The involvement of the mitochondrial membrane in drug delivery. Acta Biomater 2024; 176:28-50. [PMID: 38280553 DOI: 10.1016/j.actbio.2024.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/23/2023] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
Treatment effectiveness and biosafety are critical for disease therapy. Bio-membrane modification facilitates the homologous targeting of drugs in vivo by exploiting unique antibodies or antigens, thereby enhancing therapeutic efficacy while ensuring biosafety. To further enhance the precision of disease treatment, future research should shift focus from targeted cellular delivery to targeted subcellular delivery. As the cellular powerhouses, mitochondria play an indispensable role in cell growth and regulation and are closely involved in many diseases (e.g., cancer, cardiovascular, and neurodegenerative diseases). The double-layer membrane wrapped on the surface of mitochondria not only maintains the stability of their internal environment but also plays a crucial role in fundamental biological processes, such as energy generation, metabolite transport, and information communication. A growing body of evidence suggests that various diseases are tightly related to mitochondrial imbalance. Moreover, mitochondria-targeted strategies hold great potential to decrease therapeutic threshold dosage, minimize side effects, and promote the development of precision medicine. Herein, we introduce the structure and function of mitochondrial membranes, summarize and discuss the important role of mitochondrial membrane-targeting materials in disease diagnosis/treatment, and expound the advantages of mitochondrial membrane-assisted drug delivery for disease diagnosis, treatment, and biosafety. This review helps readers understand mitochondria-targeted therapies and promotes the application of mitochondrial membranes in drug delivery. STATEMENT OF SIGNIFICANCE: Bio-membrane modification facilitates the homologous targeting of drugs in vivo by exploiting unique antibodies or antigens, thereby enhancing therapeutic efficacy while ensuring biosafety. Compared to cell-targeted treatment, targeting of mitochondria for drug delivery offers higher efficiency and improved biosafety and will promote the development of precision medicine. As a natural material, the mitochondrial membrane exhibits excellent biocompatibility and can serve as a carrier for mitochondria-targeted delivery. This review provides an overview of the structure and function of mitochondrial membranes and explores the potential benefits of utilizing mitochondrial membrane-assisted drug delivery for disease treatment and biosafety. The aim of this review is to enhance readers' comprehension of mitochondrial targeted therapy and to advance the utilization of mitochondrial membrane in drug delivery.
Collapse
Affiliation(s)
- Yinghui Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Wenhui Ji
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Jiaxin Zhang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Ze Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China; Future Display Institute in Xiamen, Xiamen 361005, China
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Kai Huang
- Future Display Institute in Xiamen, Xiamen 361005, China
| | - Wei Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Tingting Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China; Future Display Institute in Xiamen, Xiamen 361005, China.
| |
Collapse
|
10
|
Alvarez S, Vanasco V, Adán Areán JS, Magnani N, Evelson P. Mitochondrial Mechanisms in Immunity and Inflammatory Conditions: Beyond Energy Management. Antioxid Redox Signal 2024. [PMID: 38062738 DOI: 10.1089/ars.2023.0367] [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] [Indexed: 01/25/2024]
Abstract
Significance: The growing importance of mitochondria in the immune response and inflammation is multifaceted. Unraveling the different mechanisms by which mitochondria have a relevant role in the inflammatory response beyond the energy management of the process is necessary for improving our understanding of the host immune defense and the pathogenesis of various inflammatory diseases and syndromes. Critical Issues: Mitochondria are relevant in the immune response at different levels, including releasing activation molecules, changing its structure and function to accompany the immune response, and serving as a structural base for activating intermediates as NLRP3 inflammasome. In this scientific journey of dissecting mitochondrial mechanisms, new questions and interesting aspects arise, such as the involvement of mitochondrial-derived vesicles in the immune response with the putative role of preventing uncontrolled situations. Recent Advances: Researchers are continuously rethinking the role of mitochondria in acute and chronic inflammation and related disorders. As such, mitochondria have important roles as centrally positioned signaling hubs in regulating inflammatory and immune responses. In this review, we present the current understanding of mitochondrial mechanisms involved, beyond the largely known mitochondrial dysfunction, in the onset and development of inflammatory situations. Future Directions: Mitochondria emerge as an interesting and multifaceted platform for studying and developing pharmaceutical and therapeutic approaches. There are many ongoing studies aimed to describe the effects of specific mitochondrial targeted molecules and treatments to ameliorate the consequences of exacerbated inflammatory components of pathologies and syndromes, resulting in an open area of increasing research interest.
Collapse
Affiliation(s)
- Silvia Alvarez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Fisicoquímica, CABA, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
| | - Virginia Vanasco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Fisicoquímica, CABA, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
| | - Juan Santiago Adán Areán
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Fisicoquímica, CABA, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, CABA, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, CABA, Argentina
| |
Collapse
|
11
|
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
|
12
|
Li C, Liu Q, Chang Q, Xie M, Weng J, Wang X, Li M, Chen J, Huang Y, Yang X, Wang K, Zhang N, Chung KF, Adcock IM, Zhang H, Li F. Role of mitochondrial fusion proteins MFN2 and OPA1 on lung cellular senescence in chronic obstructive pulmonary disease. Respir Res 2023; 24:319. [PMID: 38110986 PMCID: PMC10726594 DOI: 10.1186/s12931-023-02634-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 12/10/2023] [Indexed: 12/20/2023] Open
Abstract
BACKGROUND Mitochondrial dysfunction and lung cellular senescence are significant features involved in the pathogenesis of chronic obstructive pulmonary disease (COPD). Cigarette smoke (CS) stands as the primary contributing factor to COPD. This study examined mitochondrial dynamics, mitophagy and lung cellular senescence in COPD patients and investigated the effects of modulation of mitochondrial fusion [mitofusin2 (MFN2) and Optic atrophy 1 (OPA1)] on CS extract (CSE)-induced lung cellular senescence. METHODS Senescence-associated secretory phenotype (SASP) component mRNAs (IL-1β, IL-6, CXCL1 and CXCL8), mitochondrial morphology, mitophagy and mitochondria-related proteins (including phosphorylated-DRP1(p-DRP1), DRP1, MFF, MNF2, OPA1, PINK1, PARK2, SQSTM1/p62 and LC3b) and senescence-related proteins (including P16, H2A.X and Klotho) were measured in lung tissues or primary alveolar type II (ATII) cells of non-smokers, smokers and COPD patients. Alveolar epithelial (A549) cells were exposed to CSE with either pharmacologic inducer (leflunomide and BGP15) or genetic induction of MFN2 and OPA1 respectively. RESULTS There were increases in mitochondrial number, and decreases in mitochondrial size and activity in lung tissues from COPD patients. SASP-related mRNAs, DRP1 phosphorylation, DRP1, MFF, PARK2, SQSTM1/p62, LC3B II/LC3B I, P16 and H2A.X protein levels were increased, while MFN2, OPA1, PINK1 and Klotho protein levels were decreased in lung tissues from COPD patients. Some similar results were identified in primary ATII cells of COPD patients. CSE induced increases in oxidative stress, SASP-related mRNAs, mitochondrial damage and dysfunction, mitophagy and cellular senescence in A549 cells, which were ameliorated by both pharmacological inducers and genetic overexpression of MFN2 and OPA1. CONCLUSIONS Impaired mitochondrial fusion, enhanced mitophagy and lung cellular senescence are observed in the lung of COPD patients. Up-regulation of MFN2 and OPA1 attenuates oxidative stress, mitophagy and lung cellular senescence, offering potential innovative therapeutic targets for COPD therapy.
Collapse
Affiliation(s)
- Chenfei Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China
| | - Qi Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China
| | - Qing Chang
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China
| | - Meiqin Xie
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China
| | - Jiali Weng
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China
| | - Xiaohui Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China
| | - Mengnan Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China
| | - Jiani Chen
- College of Public Health, University of South China, NO.28, West Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China
| | - Yan Huang
- School of Pharmacy, Anhui Medical University, Meishan Road, Hefei, 230032, Anhui, People's Republic of China
| | - Xiaohua Yang
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, Shanghai, 200030, People's Republic of China
| | - Kai Wang
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, Shanghai, 200030, People's Republic of China
| | - Na Zhang
- Department of Central Laboratory, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, Shanghai, 200030, People's Republic of China
| | - Kian Fan Chung
- Airway Disease Section, National Heart and Lung Institute, Imperial College, Dovehouse Street, London, SW3 6LY, UK
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, Dovehouse Street, London, SW3 6LY, UK
| | - Hai Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China.
| | - Feng Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, NO.241, West HuaiHai Road, 200030, Shanghai, People's Republic of China.
| |
Collapse
|
13
|
Cabrera AR, Deaver JW, Lim S, Morena da Silva F, Schrems ER, Saling LW, Tsitkanou S, Rosa-Caldwell ME, Wiggs MP, Washington TA, Greene NP. Females display relatively preserved muscle quality compared with males during the onset and early stages of C26-induced cancer cachexia. J Appl Physiol (1985) 2023; 135:655-672. [PMID: 37535708 PMCID: PMC10642509 DOI: 10.1152/japplphysiol.00196.2023] [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: 03/28/2023] [Revised: 07/05/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023] Open
Abstract
Cancer cachexia is clinically defined by involuntary weight loss >5% in <6 mo, primarily affecting skeletal muscle. Here, we aimed to identify sex differences in the onset of colorectal cancer cachexia with specific consideration to skeletal muscle contractile and metabolic functions. Eight-weeks old BALB/c mice (69 males, 59 females) received subcutaneous C26 allografts or PBS vehicle. Tumors were developed for 10-, 15-, 20-, or 25 days. Muscles and organs were collected, in vivo muscle contractility, protein synthesis rate, mitochondrial function, and protein turnover markers were assessed. One-way ANOVA within sex and trend analysis between sexes were performed, P < 0.05. Gastrocnemius and tibialis anterior (TA) muscles became atrophic in male mice at 25 days, whereas female mice exhibited no significant differences in muscle weights at endpoints despite presenting hallmarks of cancer cachexia (fat loss, hepatosplenomegaly). We observed lowered muscle contractility and protein synthesis concomitantly to muscle mass decay in males, with higher proteolytic markers in muscles of both sexes. mRNA of Opa1 was lower in TA, whereas Bnip3 was higher in gastrocnemius after 25 days in male mice, with no significant effect in female mice. Our data suggest relative protections to skeletal muscle in females compared with males despite other canonical signs of cancer cachexia and increased protein degradation markers; suggesting we should place onus upon nonmuscle tissues during early stages of cancer cachexia in females. We noted potential protective mechanisms relating to skeletal muscle contractile and mitochondrial functions. Our findings underline possible heterogeneity in onset of cancer cachexia between biological sexes, suggesting the need for sex-specific approaches to treat cancer cachexia.NEW & NOTEWORTHY Our study demonstrates biological-sex differences in phenotypic characteristics of cancer cachexia between male and female mice, whereby females display many common characteristics of cachexia (gonadal fat loss and hepatosplenomegaly), protein synthesis markers alterations, and common catabolic markers in skeletal muscle despite relatively preserved muscle mass in early-stage cachexia compared with males. Mechanisms of cancer cachexia appear to differ between sexes. Data suggest need to place onus of early cancer cachexia detection and treatment on nonmuscle tissues in females.
Collapse
Affiliation(s)
- Ana Regina Cabrera
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - J William Deaver
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Seongkyun Lim
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Francielly Morena da Silva
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Eleanor R Schrems
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Landen W Saling
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Stavroula Tsitkanou
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Megan E Rosa-Caldwell
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, United States
| | - Michael P Wiggs
- Department of Health, Human Performance and Recreation, Baylor University, Waco, Texas, United States
| | - Tyrone A Washington
- Exercise Muscle Biology Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| | - Nicholas P Greene
- Cachexia Research Laboratory, Exercise Science Research Center, Department of Health, Human Performance and Recreation, University of Arkansas, Fayetteville, Arkansas, United States
| |
Collapse
|
14
|
Baldassarro VA, Alastra G, Lorenzini L, Giardino L, Calzà L. Photobiomodulation at Defined Wavelengths Regulates Mitochondrial Membrane Potential and Redox Balance in Skin Fibroblasts. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7638223. [PMID: 37663921 PMCID: PMC10471456 DOI: 10.1155/2023/7638223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023]
Abstract
Starting from the discovery of phototherapy in the beginning of the last century, photobiomodulation (PBM) has been defined in late 1960s and, since then, widely described in different in vitro models. Robust evidence indicates that the effect of light exposure on the oxidative state of the cells and on mitochondrial dynamics, suggesting a great therapeutic potential. The translational scale-up of PBM, however, has often given contrasting and confusing results, mainly due to light exposure protocols which fail to adequately control or define factors such as emitting device features, emitted light characteristics, exposure time, cell target, and readouts. In this in vitro study, we describe the effects of a strictly controlled light-emitting diode (LED)-based PBM protocol on human fibroblasts, one of the main cells involved in skin care, regeneration, and repair. We used six emitter probes at different wavelengths (440, 525, 645, 660, 780, and 900 nm) with the same irradiance value of 0.1 mW/cm2, evenly distributed over the entire surface of the cell culture well. The PBM was analyzed by three main readouts: (i) mitochondrial potential (MitoTracker Orange staining), (ii) reactive oxygen species (ROS) production (CellROX staining); and (iii) cell death (nuclear morphology). The assay was also implemented by cell-based high-content screening technology, further increasing the reliability of the data. Different exposure protocols were also tested (one, two, or three subsequent 20 s pulsed exposures at 24 hr intervals), and the 645 nm wavelength and single exposure chosen as the most efficient protocol based on the mitochondrial potential readout, further confirmed by mitochondrial fusion quantification. This protocol was then tested for its potential to prevent H2O2-induced oxidative stress, including modulation of the light wave frequency. Finally, we demonstrated that the controlled PBM induced by the LED light exposure generates a preconditioning stimulation of the mitochondrial potential, which protects the cell from oxidative stress damage.
Collapse
Affiliation(s)
- Vito Antonio Baldassarro
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, Ozzano dell'Emilia (Bologna) 40064, Italy
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Via Tolara di Sopra 41/E, Ozzano dell'Emilia (Bologna) 40064, Italy
| | - Giuseppe Alastra
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, Ozzano dell'Emilia (Bologna) 40064, Italy
| | - Luca Lorenzini
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, Ozzano dell'Emilia (Bologna) 40064, Italy
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Via Tolara di Sopra 41/E, Ozzano dell'Emilia (Bologna) 40064, Italy
| | - Luciana Giardino
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra 50, Ozzano dell'Emilia (Bologna) 40064, Italy
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Via Tolara di Sopra 41/E, Ozzano dell'Emilia (Bologna) 40064, Italy
| | - Laura Calzà
- Interdepartmental Centre for Industrial Research in Health Sciences and Technologies, University of Bologna, Via Tolara di Sopra 41/E, Ozzano dell'Emilia (Bologna) 40064, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, Via Belmeloro 6, Bologna 40126, Italy
- IRET Fundation, Via Tolara di Sopra 41/E, Ozzano dell'Emilia (Bologna) 40064, Italy
| |
Collapse
|
15
|
Grel H, Woznica D, Ratajczak K, Kalwarczyk E, Anchimowicz J, Switlik W, Olejnik P, Zielonka P, Stobiecka M, Jakiela S. Mitochondrial Dynamics in Neurodegenerative Diseases: Unraveling the Role of Fusion and Fission Processes. Int J Mol Sci 2023; 24:13033. [PMID: 37685840 PMCID: PMC10487704 DOI: 10.3390/ijms241713033] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 09/10/2023] Open
Abstract
Neurodegenerative diseases (NDs) are a diverse group of disorders characterized by the progressive degeneration and death of neurons, leading to a range of neurological symptoms. Despite the heterogeneity of these conditions, a common denominator is the implication of mitochondrial dysfunction in their pathogenesis. Mitochondria play a crucial role in creating biomolecules, providing energy through adenosine triphosphate (ATP) generated by oxidative phosphorylation (OXPHOS), and producing reactive oxygen species (ROS). When they're not functioning correctly, becoming fragmented and losing their membrane potential, they contribute to these diseases. In this review, we explore how mitochondria fuse and undergo fission, especially in the context of NDs. We discuss the genetic and protein mutations linked to these diseases and how they impact mitochondrial dynamics. We also look at the key regulatory proteins in fusion (MFN1, MFN2, and OPA1) and fission (DRP1 and FIS1), including their post-translational modifications. Furthermore, we highlight potential drugs that can influence mitochondrial dynamics. By unpacking these complex processes, we aim to direct research towards treatments that can improve life quality for people with these challenging conditions.
Collapse
Affiliation(s)
- Hubert Grel
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Damian Woznica
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Katarzyna Ratajczak
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Ewelina Kalwarczyk
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Julia Anchimowicz
- Department of Biochemistry and Microbiology, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Weronika Switlik
- Department of Biochemistry and Microbiology, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Piotr Olejnik
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Piotr Zielonka
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Magdalena Stobiecka
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| | - Slawomir Jakiela
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences, 02-787 Warsaw, Poland
| |
Collapse
|
16
|
Liu Q, Weng J, Li C, Feng Y, Xie M, Wang X, Chang Q, Li M, Chung KF, Adcock IM, Huang Y, Zhang H, Li F. Attenuation of PM 2.5-induced alveolar epithelial cells and lung injury through regulation of mitochondrial fission and fusion. Part Fibre Toxicol 2023; 20:28. [PMID: 37464447 DOI: 10.1186/s12989-023-00534-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 06/05/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Exposure to particulate matter (PM) with an aerodynamic diameter less than 2.5 μm (PM2.5) is a risk factor for developing pulmonary diseases and the worsening of ongoing disease. Mitochondrial fission and fusion are essential processes underlying mitochondrial homeostasis in health and disease. We examined the role of mitochondrial fission and fusion in PM2.5-induced alveolar epithelial cell damage and lung injury. Key genes in these processes include dystrophin-related protein 1 (DRP1) and optic atrophy 1 (OPA1) respectively. METHODS Alveolar epithelial (A549) cells were treated with PM2.5 (32 µg/ml) in the presence and absence of Mdivi-1 (10µM, a DRP1 inhibitor) or BGP-15 (10µM, an OPA1 activator). Results were validated using DRP1-knockdown (KD) and OPA1-overexpression (OE). Mice were injected intraperitoneally with Mdivi-1 (20 mg/kg), BGP-15 (20 mg/kg) or distilled water (control) one hour before intranasal instillation of PM2.5 (7.8 mg/kg) or distilled water for two consecutive days. RESULTS PM2.5 exposure of A549 cells caused oxidative stress, enhanced inflammation, necroptosis, mitophagy and mitochondrial dysfunction indicated by abnormal mitochondrial morphology, decreased mitochondrial membrane potential (ΔΨm), reduced mitochondrial respiration and disrupted mitochondrial fission and fusion. Regulating mitochondrial fission and fusion pharmacologically using Mdivi-1 and BGP-15 and genetically using DRP1-KD and OPA1-OE prevented PM2.5-induced celluar damage in A549 cells. Mdivi-1 and BGP-15 attenuated PM2.5-induced acute lung injury in mice. CONCLUSION Increased mitochondrial fission and decreased mitochondrial fusion may underlie PM2.5-induced alveolar epithelial cell damage in vitro and lung injury in vivo.
Collapse
Affiliation(s)
- Qi Liu
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China
| | - Jiali Weng
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China
| | - Chenfei Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China
| | - Yi Feng
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China
| | - Meiqin Xie
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China
| | - Xiaohui Wang
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China
| | - Qing Chang
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China
| | - Mengnan Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China
| | - Kian Fan Chung
- Airway Disease Section, National Heart and Lung Institute, Imperial College, Dovehouse Street, SW3 6LY, London, UK
| | - Ian M Adcock
- Airway Disease Section, National Heart and Lung Institute, Imperial College, Dovehouse Street, SW3 6LY, London, UK
| | - Yan Huang
- School of Pharmacy, Anhui Medical University, 230022, Hefei, Anhui, China
| | - Hai Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China.
| | - Feng Li
- Department of Pulmonary and Critical Care Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of medicine, NO.241, West Huaihai Road, 200030, Shanghai, P.R. China.
| |
Collapse
|
17
|
Liu Q, Wang X, Hu Y, Zhao JN, Huang CH, Li T, Zhang BG, He Y, Wu YQ, Zhang ZJ, Wang GP, Liu GP. Acetylated tau exacerbates learning and memory impairment by disturbing with mitochondrial homeostasis. Redox Biol 2023; 62:102697. [PMID: 37037158 PMCID: PMC10114242 DOI: 10.1016/j.redox.2023.102697] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/22/2023] [Accepted: 04/05/2023] [Indexed: 04/09/2023] Open
Abstract
Increased tau acetylation at K274 and K281 has been observed in the brains of Alzheimer's disease (AD) patients and animal models, and mitochondrial dysfunction are noticeable and early features of AD. However, the effect of acetylated tau on mitochondria has been unclear until now. Here, we constructed three type of tau forms, acetylated tau mutant by mutating its K274/K281 into Glutamine (TauKQ) to mimic disease-associated lysine acetylation, the non-acetylation tau mutant by mutating its K274/K281 into Arginine (TauKR) and the wild-type human full-length tau (TauWT). By overexpression of these tau forms in vivo and in vitro, we found that, TauKQ induced more severe cognitive deficits with neuronal loss, dendritic plasticity damage and mitochondrial dysfunctions than TauWT. Unlike TauWT induced mitochondria fusion, TauKQ not only induced mitochondria fission by decreasing mitofusion proteins, but also inhibited mitochondrial biogenesis via reduction of PGC-1a/Nrf1/Tfam levels. TauKR had no significant difference in the cognitive and mitochondrial abnormalities compared with TauWT. Treatment with BGP-15 rescued impaired learning and memory by attenuation of mitochondrial dysfunction, neuronal loss and dendritic complexity damage, which caused by TauKQ. Our data suggested that, acetylation at K274/281 was an important post translational modification site for tau neurotoxicity, and BGP-15 is a potential therapeutic drug for AD.
Collapse
Affiliation(s)
- Qian Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Yu Hu
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun-Ning Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chun-Hui Huang
- Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research, Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Ting Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bing-Ge Zhang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ye He
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan-Qing Wu
- Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Zai-Jun Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research, Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Guo-Ping Wang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
18
|
Huang Y, Zhou B. Mitochondrial Dysfunction in Cardiac Diseases and Therapeutic Strategies. Biomedicines 2023; 11:biomedicines11051500. [PMID: 37239170 DOI: 10.3390/biomedicines11051500] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Mitochondria are the main site of intracellular synthesis of ATP, which provides energy for various physiological activities of the cell. Cardiomyocytes have a high density of mitochondria and mitochondrial damage is present in a variety of cardiovascular diseases. In this paper, we describe mitochondrial damage in mitochondrial cardiomyopathy, congenital heart disease, coronary heart disease, myocardial ischemia-reperfusion injury, heart failure, and drug-induced cardiotoxicity, in the context of the key roles of mitochondria in cardiac development and homeostasis. Finally, we discuss the main current therapeutic strategies aimed at alleviating mitochondrial impairment-related cardiac dysfunction, including pharmacological strategies, gene therapy, mitochondrial replacement therapy, and mitochondrial transplantation. It is hoped that this will provide new ideas for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yafei Huang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing 100037, China
| | - Bingying Zhou
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, 167 North Lishi Road, Xicheng District, Beijing 100037, China
| |
Collapse
|
19
|
Wu A, Zhang W, Zhang G, Ding X, Kang L, Zhou T, Ji M, Guan H. Age-related cataract: GSTP1 ubiquitination and degradation by Parkin inhibits its anti-apoptosis in lens epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119450. [PMID: 36871745 DOI: 10.1016/j.bbamcr.2023.119450] [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: 12/01/2022] [Revised: 02/14/2023] [Accepted: 02/23/2023] [Indexed: 03/07/2023]
Abstract
PURPOSE Oxidative stress-induced apoptosis of lens epithelial cells (LECs) contributes to the pathogenesis of age-related cataract (ARC). The purpose of this research is to underlie the potential mechanism of E3 ligase Parkin and its oxidative stress-associated substrate in cataractogenesis. METHODS The central anterior capsules were obtained from patients with ARC, Emory mice, and corresponding controls. SRA01/04 cells were exposed to H2O2 combined with cycloheximide (a translational inhibitor), MG-132 (a proteasome inhibitor), chloroquine (an autophagy inhibitor), Mdivi-1 (a mitochondrial division inhibitor), respectively. Co-immunoprecipitation was employed to detect protein-protein interactions and ubiquitin-tagged protein products. Levels of proteins and mRNA were evaluated by western blotting and quantitative RT-PCR assays. RESULTS Glutathione-S-transferase P1 (GSTP1) was identified as a novel Parkin substrate. Compared with corresponding controls, GSTP1 was significantly decreased in the anterior lens capsules obtained from human cataracts and Emory mice. Similarly, GSTP1 was declined in H2O2-stimulated SRA01/04 cells. Ectopic expression of GSTP1 mitigated H2O2-induced apoptosis, whereas silencing GSTP1 aggregated apoptosis. In addition, H2O2 stimulation and Parkin overexpression could promote the degradation of GSTP1 through the ubiquitin-proteasome system, autophagy-lysosome pathway, and mitophagy. After co-transfection with Parkin, the non-ubiquitinatable GSTP1 mutant maintained its anti-apoptotic function, while wildtype GSTP1 failed. Mechanistically, GSTP1 might promote mitochondrial fusion through upregulating Mitofusins 1/2 (MFN1/2). CONCLUSION Oxidative stress induces LECs apoptosis via Parkin-regulated degradation of GSTP1, which may provide potential targets for ARC therapy.
Collapse
Affiliation(s)
- Anran Wu
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Wenyi Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Guowei Zhang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Xuemeng Ding
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Lihua Kang
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Tianqiu Zhou
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China
| | - Min Ji
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| | - Huaijin Guan
- Eye Institute, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
20
|
Dharaskar SP, Amere Subbarao S. The mitochondrial chaperone TRAP-1 regulates the glutamine metabolism in tumor cells. Mitochondrion 2023; 69:159-170. [PMID: 36828164 DOI: 10.1016/j.mito.2023.02.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/24/2023]
Abstract
Understanding cancer cell metabolism always provides information on hidden dimensions of tumor adaptations. Warburg's theory that cancer cells opt for aerobic glycolysis over the mitochondrial oxidative phosphorylation (OXPHOS) system is widely accepted. However, the hypothesis does not explain the mitochondrion's role in these cells. Here, we demonstrate that intact mitochondria are used for anaplerotic functions and ATP production by utilizing glutamine with the help of mitochondrial chaperone TRAP-1 (Tumor Necrosis Factor Receptor-associated Protein 1). TRAP-1 otherwise promotes aerobic glycolysis by lowering the mitochondrial OXPHOS in the presence of glucose. Here, we show that TRAP-1 maintains mitochondrial integrity and augments glutamine metabolism upon glucose deprivation to meet the cellular energy demand. The enhanced PER and ECAR correlating with increased ATP production suggest that glutamine fuels mitochondria in the presence of TRAP-1. We also found that TRAP1-dependent glutamine utilization involves the HIF2α-SLC1A5-GLS axis and is independent of hypoxia. Subsequently, we show that the metastatic potential of tumor cells is linked with glucose utilization, whereas the proliferative potential is linked with both glucose and glutamine utilization. Our findings establish that TRAP-1 contributes to enhanced glutamine utilization through the HIF2α-SLC1A5-GLS axis. Our results endow that TRAP-1 inhibitors can be potential drug candidates to combat tumor metabolism. Therefore, their use, either alone or in combination with existing chemotherapeutic agents, may target tumor metabolism and improve anticancer treatment response.
Collapse
Affiliation(s)
- Shrikant Purushottam Dharaskar
- CSIR - Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, Telangana, India; Academy of Scientific & Innovative Research (AcSIR), Government of India, Ghaziabad 201002, Uttar Pradesh, India
| | - Sreedhar Amere Subbarao
- CSIR - Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500007, Telangana, India; Academy of Scientific & Innovative Research (AcSIR), Government of India, Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
21
|
Targeting mitochondrial impairment for the treatment of cardiovascular diseases: From hypertension to ischemia-reperfusion injury, searching for new pharmacological targets. Biochem Pharmacol 2023; 208:115405. [PMID: 36603686 DOI: 10.1016/j.bcp.2022.115405] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023]
Abstract
Mitochondria and mitochondrial proteins represent a group of promising pharmacological target candidates in the search of new molecular targets and drugs to counteract the onset of hypertension and more in general cardiovascular diseases (CVDs). Indeed, several mitochondrial pathways result impaired in CVDs, showing ATP depletion and ROS production as common traits of cardiac tissue degeneration. Thus, targeting mitochondrial dysfunction in cardiomyocytes can represent a successful strategy to prevent heart failure. In this context, the identification of new pharmacological targets among mitochondrial proteins paves the way for the design of new selective drugs. Thanks to the advances in omics approaches, to a greater availability of mitochondrial crystallized protein structures and to the development of new computational approaches for protein 3D-modelling and drug design, it is now possible to investigate in detail impaired mitochondrial pathways in CVDs. Furthermore, it is possible to design new powerful drugs able to hit the selected pharmacological targets in a highly selective way to rescue mitochondrial dysfunction and prevent cardiac tissue degeneration. The role of mitochondrial dysfunction in the onset of CVDs appears increasingly evident, as reflected by the impairment of proteins involved in lipid peroxidation, mitochondrial dynamics, respiratory chain complexes, and membrane polarization maintenance in CVD patients. Conversely, little is known about proteins responsible for the cross-talk between mitochondria and cytoplasm in cardiomyocytes. Mitochondrial transporters of the SLC25A family, in particular, are responsible for the translocation of nucleotides (e.g., ATP), amino acids (e.g., aspartate, glutamate, ornithine), organic acids (e.g. malate and 2-oxoglutarate), and other cofactors (e.g., inorganic phosphate, NAD+, FAD, carnitine, CoA derivatives) between the mitochondrial and cytosolic compartments. Thus, mitochondrial transporters play a key role in the mitochondria-cytosol cross-talk by leading metabolic pathways such as the malate/aspartate shuttle, the carnitine shuttle, the ATP export from mitochondria, and the regulation of permeability transition pore opening. Since all these pathways are crucial for maintaining healthy cardiomyocytes, mitochondrial carriers emerge as an interesting class of new possible pharmacological targets for CVD treatments.
Collapse
|
22
|
Zhang W, Liu B, Wang Y, Zhang H, He L, Wang P, Dong M. Mitochondrial dysfunction in pulmonary arterial hypertension. Front Physiol 2022; 13:1079989. [PMID: 36589421 PMCID: PMC9795033 DOI: 10.3389/fphys.2022.1079989] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/29/2022] [Indexed: 01/03/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is characterized by the increased pulmonary vascular resistance due to pulmonary vasoconstriction and vascular remodeling. PAH has high disability, high mortality and poor prognosis, which is becoming a more common global health issue. There is currently no drug that can permanently cure PAH patients. The pathogenesis of PAH is still not fully elucidated. However, the role of metabolic theory in the pathogenesis of PAH is becoming clearer, especially mitochondrial metabolism. With the deepening of mitochondrial researches in recent years, more and more studies have shown that the occurrence and development of PAH are closely related to mitochondrial dysfunction, including the tricarboxylic acid cycle, redox homeostasis, enhanced glycolysis, and increased reactive oxygen species production, calcium dysregulation, mitophagy, etc. This review will further elucidate the relationship between mitochondrial metabolism and pulmonary vasoconstriction and pulmonary vascular remodeling. It might be possible to explore more comprehensive and specific treatment strategies for PAH by understanding these mitochondrial metabolic mechanisms.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Bo Liu
- Department of Cardiovascular, Geratric Diseases Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Yazhou Wang
- Department of Cardiothoracic, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Hengli Zhang
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China
| | - Lang He
- Department of Oncology, Cancer Prevention and Treatment Institute of Chengdu, Chengdu Fifth People’s Hospital (The Second Clinical Medical College Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China,Correspondence: Mingqing Dong, ; Lang He, ; Pan Wang,
| | - Pan Wang
- Department of Critical Care Medicine, The Traditional Chinese Medicine Hospital of Wenjiang District, Chengdu, China,Correspondence: Mingqing Dong, ; Lang He, ; Pan Wang,
| | - Mingqing Dong
- Center for Medicine Research and Translation, Chengdu Fifth People’s Hospital (The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine), Chengdu, China,Correspondence: Mingqing Dong, ; Lang He, ; Pan Wang,
| |
Collapse
|
23
|
Jurcău MC, Andronie-Cioara FL, Jurcău A, Marcu F, Ţiț DM, Pașcalău N, Nistor-Cseppentö DC. The Link between Oxidative Stress, Mitochondrial Dysfunction and Neuroinflammation in the Pathophysiology of Alzheimer's Disease: Therapeutic Implications and Future Perspectives. Antioxidants (Basel) 2022; 11:2167. [PMID: 36358538 PMCID: PMC9686795 DOI: 10.3390/antiox11112167] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 08/26/2023] Open
Abstract
Alzheimer's disease (AD), the most common form of dementia, has increasing incidence, increasing mortality rates, and poses a huge burden on healthcare. None of the currently approved drugs for the treatment of AD influence disease progression. Many clinical trials aiming at inhibiting amyloid plaque formation, increasing amyloid beta clearance, or inhibiting neurofibrillary tangle pathology yielded inconclusive results or failed. Meanwhile, research has identified many interlinked vicious cascades implicating oxidative stress, mitochondrial dysfunction, and chronic neuroinflammation, and has pointed to novel therapeutic targets such as improving mitochondrial bioenergetics and quality control, diminishing oxidative stress, or modulating the neuroinflammatory pathways. Many novel molecules tested in vitro or in animal models have proven efficient, but their translation into clinic needs further research regarding appropriate doses, delivery routes, and possible side effects. Cell-based therapies and extracellular vesicle-mediated delivery of messenger RNAs and microRNAs seem also promising strategies allowing to target specific signaling pathways, but need further research regarding the most appropriate harvesting and culture methods as well as control of the possible tumorigenic side effects. The rapidly developing area of nanotechnology could improve drug delivery and also be used in early diagnosis.
Collapse
Affiliation(s)
| | - Felicia Liana Andronie-Cioara
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Anamaria Jurcău
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Florin Marcu
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Delia Mirela Ţiț
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Nicoleta Pașcalău
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Delia Carmen Nistor-Cseppentö
- Department of Psycho-Neuroscience and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| |
Collapse
|
24
|
Tracy EP, Nair R, Rowe G, Beare JE, Beyer A, LeBlanc AJ. Adipose stromal vascular fraction reverses mitochondrial dysfunction and hyperfission in aging-induced coronary microvascular disease. Am J Physiol Heart Circ Physiol 2022; 323:H749-H762. [PMID: 36018760 PMCID: PMC9529257 DOI: 10.1152/ajpheart.00311.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 01/28/2023]
Abstract
Aging is associated with blunted coronary microvascular vasodilatory function. Previously, systemically administered adipose stromal vascular fraction (SVF) therapy reversed aging-induced attenuation of β1-adrenergic- and flow-mediated dilation dependent on reducing mitochondrial reactive oxygen species. We hypothesized that SVF-mediated recovery of microvascular dilatory function is dependent on recovery of mitochondrial function, specifically by reducing mitochondrial hyperfission. Female Fischer-344 rats were allocated into young control, old control, and old + SVF therapy groups. Pressure myography, immunofluorescent staining, Western blot analysis, and RNA sequencing were performed to determine coronary microvascular mitochondrial dynamics and function. Gene and protein expression of fission-mediator DRP-1 was enhanced with aging but reversed by SVF therapy. SVF facilitated an increase in fusion-mediator MFN-1 gene and protein expression. Mitochondrial morphology was characterized as rod-like and densely networked in young controls, isolated circular and punctate with aging, and less circularity with partially restored mitochondrial branch density with SVF therapy. Decreased mitochondrial membrane potential and ATP bioavailability in aged animals at baseline and during flow-mediated dilation were reversed by SVF and accompanied with enhanced oxygen consumption. Dilation to norepinephrine and flow in young controls were dependent on uninhibited mitochondrial fusion, whereas inhibiting fission did not restore aged microvessel response to norepinephrine or flow. SVF-mediated recovery of β-adrenergic function was dependent on uninhibited mitochondrial fusion, whereas recovery of flow-mediated dilation was dependent on maintained mitochondrial fission. Impaired dilation in aging is mitigated by SVF therapy, which recovers mitochondrial function and fission/fusion balance.NEW & NOTEWORTHY We elucidated the consequences of aging on coronary microvascular mitochondrial health as well as SVF's ability to reverse these effects. Aging shifts gene/protein expression and mitochondrial morphology indicating hyperfission, alongside attenuated mitochondrial membrane potential and ATP bioavailability, all reversed using SVF therapy. Mitochondrial membrane potential and ATP levels correlated with vasodilatory efficiency. Mitochondrial dysfunction is a contributing pathological factor in aging that can be targeted by therapeutic SVF to preserve microvascular dilative function.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Rajeev Nair
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Gabrielle Rowe
- Department of Physiology, University of Louisville, Louisville, Kentucky
| | - Jason E Beare
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, Kentucky
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky
| | - Andreas Beyer
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Amanda Jo LeBlanc
- Department of Cardiovascular and Thoracic Surgery, University of Louisville, Louisville, Kentucky
| |
Collapse
|
25
|
Gonzalez M, Prashar T, Connaughton H, Barry M, Robker R, Rose R. Restoring Sperm Quality Post-Cryopreservation Using Mitochondrial-Targeted Compounds. Antioxidants (Basel) 2022; 11:antiox11091808. [PMID: 36139882 PMCID: PMC9495717 DOI: 10.3390/antiox11091808] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/03/2022] [Accepted: 09/05/2022] [Indexed: 11/16/2022] Open
Abstract
While critical for male fertility preservation, cryopreservation damage reduces sperm quality and fertilization potential. This study investigated whether the addition of mitochondrial-targeted, antioxidant compounds, also known as Mitochondrial activators, to the cryopreservation medium could protect sperm quality during cryopreservation. For this, semen samples from men undergoing IVF/ICSI treatment, which were donated for research, underwent cryopreservation in the absence or presence of BGP-15, MitoQ and L-carnitine. Fresh semen and thawed sperm samples from the same participant were analyzed for indicators of sperm quality: sperm viability, kinetics, mitochondrial reactive oxygen species (ROS) levels, Mitochondrial Membrane Potential (MMP) and DNA damage. Cryopreservation significantly reduced sperm viability and motility and predicted mucous penetration. BGP-15, MitoQ and L-carnitine improved sperm motility, whilst the addition of L-Carnitine prevented the loss of sperm viability during cryopreservation. Both BGP-15 and L-carnitine reduced sperm DNA oxidative damage, but only BGP-15 significantly reduced DNA fragmentation. More importantly, BGP-15 increased sperm predictive mucous penetration and MMP and reduced DNA oxidation. Our results show that the addition of BGP-15 or L-carnitine to the cryopreservation medium improves sperm quality post-thawing, highlighting the potential of mitochondrial antioxidants to improve long-term fertility preservation in males.
Collapse
Affiliation(s)
- Macarena Gonzalez
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide 5000, Australia
- Correspondence:
| | - Tanisha Prashar
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide 5000, Australia
| | - Haley Connaughton
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide 5000, Australia
| | - Michael Barry
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide 5000, Australia
- Fertility SA, St. Andrew’s Hospital, Adelaide 5000, Australia
| | - Rebecca Robker
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide 5000, Australia
| | - Ryan Rose
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide 5000, Australia
- Fertility SA, St. Andrew’s Hospital, Adelaide 5000, Australia
| |
Collapse
|
26
|
Lin J, Duan J, Wang Q, Xu S, Zhou S, Yao K. Mitochondrial Dynamics and Mitophagy in Cardiometabolic Disease. Front Cardiovasc Med 2022; 9:917135. [PMID: 35783853 PMCID: PMC9247260 DOI: 10.3389/fcvm.2022.917135] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/20/2022] [Indexed: 12/17/2022] Open
Abstract
Mitochondria play a key role in cellular metabolism. Mitochondrial dynamics (fusion and fission) and mitophagy, are critical to mitochondrial function. Fusion allows organelles to share metabolites, proteins, and mitochondrial DNA, promoting complementarity between damaged mitochondria. Fission increases the number of mitochondria to ensure that they are passed on to their offspring during mitosis. Mitophagy is a process of selective removal of excess or damaged mitochondria that helps improve energy metabolism. Cardiometabolic disease is characterized by mitochondrial dysfunction, high production of reactive oxygen species, increased inflammatory response, and low levels of ATP. Cardiometabolic disease is closely related to mitochondrial dynamics and mitophagy. This paper reviewed the mechanisms of mitochondrial dynamics and mitophagy (focus on MFN1, MFN2, OPA1, DRP1, and PINK1 proteins) and their roles in diabetic cardiomyopathy, myocardial infarction, cardiac hypertrophy, heart failure, atherosclerosis, and obesity.
Collapse
Affiliation(s)
- Jianguo Lin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinlong Duan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingqing Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Siyu Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Simin Zhou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Kuiwu Yao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Eye Hospital China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Kuiwu Yao
| |
Collapse
|
27
|
Umehara T, Winstanley YE, Andreas E, Morimoto A, Williams EJ, Smith KM, Carroll J, Febbraio MA, Shimada M, Russell DL, Robker RL. Female reproductive life span is extended by targeted removal of fibrotic collagen from the mouse ovary. SCIENCE ADVANCES 2022; 8:eabn4564. [PMID: 35714185 PMCID: PMC9205599 DOI: 10.1126/sciadv.abn4564] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The female ovary contains a finite number of oocytes, and their release at ovulation becomes sporadic and disordered with aging and with obesity, leading to loss of fertility. Understanding the molecular defects underpinning this pathology is essential as age of childbearing and obesity rates increase globally. We identify that fibrosis within the ovarian stromal compartment is an underlying mechanism responsible for impaired oocyte release, which is initiated by mitochondrial dysfunction leading to diminished bioenergetics, oxidative damage, inflammation, and collagen deposition. Furthermore, antifibrosis drugs (pirfenidone and BGP-15) eliminate fibrotic collagen and restore ovulation in reproductively old and obese mice, in association with dampened M2 macrophage polarization and up-regulated MMP13 protease. This is the first evidence that ovarian fibrosis is reversible and indicates that drugs targeting mitochondrial metabolism may be a viable therapeutic strategy for women with metabolic disorders or advancing age to maintain ovarian function and extend fertility.
Collapse
Affiliation(s)
- Takashi Umehara
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Yasmyn E. Winstanley
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Eryk Andreas
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Atsushi Morimoto
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Elisha J. Williams
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Kirsten M. Smith
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - John Carroll
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Mark A. Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Masayuki Shimada
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Darryl L. Russell
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Rebecca L. Robker
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Corresponding author.
| |
Collapse
|
28
|
Jiang HL, Yang HH, Liu YB, Zhang CY, Zhong WJ, Guan XX, Jin L, Hong JR, Yang JT, Tan XH, Li Q, Zhou Y, Guan CX. L-OPA1 deficiency aggravates necroptosis of alveolar epithelial cells through impairing mitochondrial function during ALI in mice. J Cell Physiol 2022; 237:3030-3043. [PMID: 35478455 DOI: 10.1002/jcp.30766] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 11/11/2022]
Abstract
Necroptosis, a recently described form of programmed cell death, is the main way of alveolar epithelial cells (AECs) death in acute lung injury (ALI). While the mechanism of how to trigger necroptosis in AECs during ALI has been rarely evaluated. Long optic atrophy protein 1 (L-OPA1) is a crucial mitochondrial inner membrane fusion protein, and its deficiency impairs mitochondrial function. This study aimed to investigate the role of L-OPA1 deficiency-mediated mitochondrial dysfunction in AECs necroptosis. We comprehensively investigated the detailed contribution and molecular mechanism of L-OPA1 deficiency in AECs necroptosis by inhibiting or activating L-OPA1. Firstly, our data showed that L-OPA1 expression was down-regulated in the lungs and AECs under the lipopolysaccharide (LPS) challenge. Furthermore, inhibition of L-OPA1 aggravated the pathological injury, inflammatory response, and necroptosis in the lungs of LPS-induced ALI mice. In vitro, inhibition of L-OPA1 induced necroptosis of AECs, while activation of L-OPA1 alleviated necroptosis of AECs under the LPS challenge. Mechanistically, inhibition of L-OPA1 aggravated necroptosis of AECs by inducing mitochondrial fragmentation and reducing mitochondrial membrane potential. While activation of L-OPA1 had the opposite effects. In summary, these findings indicate for the first time that L-OPA1 deficiency mediates mitochondrial fragmentation, induces necroptosis of AECs, and exacerbates ALI in mice. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hui-Ling Jiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Xin-Xin Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jie-Ru Hong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jin-Tong Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Xiao-Hua Tan
- Experimental Center of Medical Morphology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Qing Li
- Department of Physiology, Hunan University of Medicine, Huaihua, Hunan, 418000, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| |
Collapse
|
29
|
Metabolism, Mitochondrial Dysfunction, and Redox Homeostasis in Pulmonary Hypertension. Antioxidants (Basel) 2022; 11:antiox11020428. [PMID: 35204311 PMCID: PMC8869288 DOI: 10.3390/antiox11020428] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 02/05/2023] Open
Abstract
Pulmonary hypertension (PH) represents a group of disorders characterized by elevated mean pulmonary artery (PA) pressure, progressive right ventricular failure, and often death. Some of the hallmarks of pulmonary hypertension include endothelial dysfunction, intimal and medial proliferation, vasoconstriction, inflammatory infiltration, and in situ thrombosis. The vascular remodeling seen in pulmonary hypertension has been previously linked to the hyperproliferation of PA smooth muscle cells. This excess proliferation of PA smooth muscle cells has recently been associated with changes in metabolism and mitochondrial biology, including changes in glycolysis, redox homeostasis, and mitochondrial quality control. In this review, we summarize the molecular mechanisms that have been reported to contribute to mitochondrial dysfunction, metabolic changes, and redox biology in PH.
Collapse
|
30
|
Identifying Potential Mitochondrial Proteome Signatures Associated with the Pathogenesis of Pulmonary Arterial Hypertension in the Rat Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8401924. [PMID: 35237384 PMCID: PMC8885180 DOI: 10.1155/2022/8401924] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 01/12/2022] [Accepted: 02/05/2022] [Indexed: 01/09/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a severe and progressive disease that affects the heart and lungs and a global health concern that impacts individuals and society. Studies have reported that some proteins related to mitochondrial metabolic functions could play an essential role in the pathogenesis of PAH, and their specific expression and biological function are still unclear. We successfully constructed a monocrotaline- (MCT-) induced PAH rat model in the present research. Then, the label-free quantification proteomic technique was used to determine mitochondrial proteins between the PAH group (n = 6) and the normal group (n = 6). Besides, we identified 1346 mitochondrial differentially expressed proteins (DEPs) between these two groups. Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) were used to analyze the mainly mitochondrial DEPs' biological functions and the signal pathways. Based on the protein-protein interaction (PPI) network construction and functional enrichment, we screened 19 upregulated mitochondrial genes (Psmd1, Psmc4, Psmd13, Psmc2, etc.) and 123 downregulated mitochondrial genes (Uqcrfs1, Uqcrc1, Atp5c1, Atp5a1, Uqcrc2, etc.) in rats with PAH. Furthermore, in an independent cohort dataset and experiments with rat lung tissue using qPCR, validation results consistently showed that 6 upregulated mitochondrial genes (Psmd2, Psmc4, Psmc3, Psmc5, Psmd13, and Psmc2) and 3 downregulated mitochondrial genes (Lipe, Cat, and Prkce) were significantly differentially expressed in the lung tissue of PAH rats. Using the RNAInter database, we predict potential miRNA target hub mitochondrial genes at the transcriptome level. We also identified bortezomib and carfilzomib as the potential drugs for treatment in PAH. Finally, this study provides us with a new perspective on critical biomarkers and treatment strategies in PAH.
Collapse
|
31
|
Zhang S, Rao S, Yang M, Ma C, Hong F, Yang S. Role of Mitochondrial Pathways in Cell Apoptosis during He-Patic Ischemia/Reperfusion Injury. Int J Mol Sci 2022; 23:ijms23042357. [PMID: 35216473 PMCID: PMC8877300 DOI: 10.3390/ijms23042357] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/13/2022] [Accepted: 02/17/2022] [Indexed: 12/15/2022] Open
Abstract
Hepatic ischemia-reperfusion injury is a major cause of post-operative hepatic dysfunction and liver failure after transplantation. Mitochondrial pathways can be either beneficial or detrimental to hepatic cell apoptosis during hepatic ischemia/reperfusion injury, depending on multiple factors. Hepatic ischemia/reperfusion injury may be induced by opened mitochondrial permeability transition pore, released apoptosis-related proteins, up-regulated B-cell lymphoma-2 gene family proteins, unbalanced mitochondrial dynamics, and endoplasmic reticulum stress, which are integral parts of mitochondrial pathways. In this review, we discuss the role of mitochondrial pathways in apoptosis that account for the most deleterious effect of hepatic ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Sen Zhang
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Sijing Rao
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Meiwen Yang
- Department of Surgery, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China;
| | - Chen Ma
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
| | - Fengfang Hong
- Experimental Center of Pathogen Biology, College of Medicine, Nanchang University, Nanchang 330006, China; (S.Z.); (S.R.); (C.M.)
- Correspondence: (F.H.); or (S.Y.)
| | - Shulong Yang
- Department of Physiology, College of Medicine, Nanchang University, Nanchang 330006, China
- Department of Physiology, Fuzhou Medical College, Nanchang University, Fuzhou 344099, China
- Correspondence: (F.H.); or (S.Y.)
| |
Collapse
|
32
|
Sun J, Yu F, Wang T, Bian J, Liu Z, Zou H. The role of DRP1- PINK1-Parkin-mediated mitophagy in early cadmium-induced liver damage. Toxicology 2021; 466:153082. [PMID: 34952138 DOI: 10.1016/j.tox.2021.153082] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022]
Abstract
Cadmium (Cd) is an important environmental pollutant that causes varying degrees of damage to multiple systems of the body. However, the specific mechanism of Cd-induced liver mitophagy remains unclear. In the present study, 5-week-old BALB/c mice and a mouse liver parenchyma cell line (AML12) were studied using a combination of in vivo and in vitro studies. We found that Cd damaged liver cells, destroy the structure and function of mitochondria, and increased the production of superoxide anions. This study further examined the effect of Cd on mitochondrial dynamics and mitophagy and showed that Cd increased mitochondrial division and induced mitophagy. The PINK1-Parkin pathway is a classical mitophagy pathway. Cd-induced mitophagy was inhibited after significantly knocking down Pink1. Mdivi-1 can effectively inhibit mitochondrial division. In this study, Mdivi-1 inhibited the expression of DRP1 and significantly inhibited the occurrence of mitophagy induced by Cd. We further examined the effect of Cd on mitophagy flux. Cd did not increase lysosomal colocalization with mitochondria. In summary, Cd increase the level of oxidative stress, destroy the structure and function of mitochondria, destroy the homeostasis of mitochondrial division and fusion, induce mitophagy through the PINK1-Parkin pathway. Mitophagy plays a protective role in early cadmium-induced liver damage.
Collapse
Affiliation(s)
- Jian Sun
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Fan Yu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jianchun Bian
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Zongping Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Hui Zou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.
| |
Collapse
|
33
|
Morio A, Tsutsumi R, Satomi S, Kondo T, Miyoshi H, Kato T, Kuroda M, Kitamura T, Hara K, Saeki N, Sakaue H, Tsutsumi YM. Leucine imparts cardioprotective effects by enhancing mTOR activity and mitochondrial fusion in a myocardial ischemia/reperfusion injury murine model. Diabetol Metab Syndr 2021; 13:139. [PMID: 34801078 PMCID: PMC8606064 DOI: 10.1186/s13098-021-00755-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 11/04/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Coronary artery disease is a leading cause of morbidity and mortality among patients with diabetes. Previously, we demonstrated that branched-chain amino acids (BCAAs) showed cardioprotective effects against cardiac ischemia/reperfusion (I/R) injury. A recent study suggested that leucine (Leu), a BCAA, is a key amino acid involved in mammalian target of rapamycin (mTOR) activity and mitochondrial function. However, whether Leu has cardioprotective effects on diabetic hearts is unclear. In this study, we examined the preconditioning effect of Leu treatment on high-fat diet (HFD)-induced obese mouse which simulate prediabetic heart. METHODS In vivo mice models of I/R injury were divided into the following groups: control, mTOR+/-, and high-fat diet (HFD)-induced obese groups. Mice were randomly administered with Leu, the mTOR inhibitor rapamycin (Rap), or Leu with Rap. Isolated rat cardiomyocytes were subjected to simulated I/R injury. Biochemical and mitochondrial functional assays were performed to evaluate the changes in mTOR activity and mitochondrial dynamics caused by Leu treatment. RESULTS Leu-treated mice showed a significant reduction in infarct size when compared with the control group (34.8% ± 3.8% vs. 43.1% ± 2.4%, n = 7, p < 0.05), whereas Rap-treated mice did not show the protective effects of Leu. This preconditioning effect of Leu was attenuated in mTOR+/- mice. Additionally, Leu increased the percentage of fused mitochondria and the mitochondrial volume, and decreased the number of mitochondria per cell in isolated cardiomyocytes. In HFD-induced obese mice, Leu treatment significantly reduced infarct size (41.0% ± 1.1% vs. 51.0% ± 1.4%, n = 7, p < 0.05), which was not induced by ischemic preconditioning, and this effect was inhibited by Rap. Furthermore, we observed enhanced mTOR protein expression and mitochondrial fusion with decreased reactive oxygen species production with Leu treatment in HFD-induced obese mice, but not in mTOR+/- mice. CONCLUSIONS Leu treatment improved the damage caused by myocardial I/R injury by promoting mTOR activity and mitochondrial fusion on prediabetic hearts in mice.
Collapse
Affiliation(s)
- Atsushi Morio
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Shiho Satomi
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Takashi Kondo
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Hirotsugu Miyoshi
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Takahiro Kato
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Masashi Kuroda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Tadahiro Kitamura
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma, 371-8512, Japan
| | - Kenta Hara
- Kita Harima Medical Center, 926-250 Ichiba, Ono, Hyogo, 675-1392, Japan
| | - Noboru Saeki
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, 3-18-15 Kuramoto, Tokushima, 770-8503, Japan
| | - Yasuo M Tsutsumi
- Department of Anesthesiology and Critical Care, Hiroshima University, 1-2-3 Kasumi, Minami, Hiroshima, 734-8551, Japan.
| |
Collapse
|
34
|
S-Carboxymethyl Cysteine Protects against Oxidative Stress and Mitochondrial Impairment in a Parkinson's Disease In Vitro Model. Biomedicines 2021; 9:biomedicines9101467. [PMID: 34680584 PMCID: PMC8533464 DOI: 10.3390/biomedicines9101467] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 10/12/2021] [Indexed: 11/21/2022] Open
Abstract
The mucolytic agent S-carboxymethylcysteine is widely used as an expectorant for the treatment of numerous respiratory disorders. The metabolic fate of S-carboxymethyl-L-cysteine is complex. Several clinical studies have demonstrated that the metabolism of this agent differs within the same individual, with sulfur oxygenated metabolites generated upon night-time administration. It has been indicated that this drug behaves like a free radical scavenger and that, in this regard, the sulfide is the active species with sulphoxide metabolites (already oxidized) being inactive. Consequently, a night-time consumption of the drug should be more effective upon daytime administration. Still, this diurnal variation in biotransformation (deactivation) is dependent on the genetic polymorphism on which relies the patient population capacities of S-carboxymethyl-L-cysteine sulphoxidation. It has been reported that those cohorts who are efficient sulfur oxidizers will generate inactive oxygenated metabolites. In contrast, those who have a relative deficiency in this mechanism will be subjected to the active sulfide for a more extended period. In this regard, it is noteworthy that 38–39% of Parkinson’s disease patients belong to the poor sulphoxide cohort, being exposed to higher levels of active sulfide, the active antioxidant metabolite of S-carboxymethyl-L-cysteine. Parkinson’s disease is a neurodegenerative disorder that affects predominately dopaminergic neurons. It has been demonstrated that oxidative stress and mitochondrial dysfunction play a crucial role in the degeneration of dopaminergic neurons. Based on this evidence, in this study, we evaluated the effects of S-carboxymethyl cysteine in an in vitro model of Parkinson’s disease in protecting against oxidative stress injury. The data obtained suggested that an S-carboxymethylcysteine-enriched diet could be beneficial during aging to protect neurons from oxidative imbalance and mitochondrial dysfunction, thus preventing the progression of neurodegenerative processes.
Collapse
|
35
|
Zhou M, Yu Y, Luo X, Wang J, Lan X, Liu P, Feng Y, Jian W. Myocardial Ischemia-Reperfusion Injury: Therapeutics from a Mitochondria-Centric Perspective. Cardiology 2021; 146:781-792. [PMID: 34547747 DOI: 10.1159/000518879] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 07/01/2021] [Indexed: 11/19/2022]
Abstract
Coronary arterial disease is the most common cardiovascular disease. Myocardial ischemia-reperfusion injury caused by the initial interruption of organ blood flow and subsequent restoration of organ blood flow is an important clinical problem with various cardiac reperfusion strategies after acute myocardial infarction. Even though blood flow recovery is necessary for oxygen and nutrient supply, reperfusion causes pathological sequelae that lead to the aggravation of ischemic injury. At present, although it is known that injury will occur after reperfusion, clinical treatment always focuses on immediate recanalization. Mitochondrial fusion, fission, biogenesis, autophagy, and their intricate interaction constitute an effective mitochondrial quality control system. The mitochondrial quality control system plays an important role in maintaining cell homeostasis and cell survival. The removal of damaged, aging, and dysfunctional mitochondria is mediated by mitochondrial autophagy. With the help of appropriate changes in mitochondrial dynamics, new mitochondria are produced through mitochondrial biogenesis to meet the energy needs of cells. Mitochondrial dysfunction and the resulting oxidative stress have been associated with the pathogenesis of ischemia/reperfusion (I/R) injury, which play a crucial role in the pathophysiological process of myocardial injury. This review aimed at elucidating the mitochondrial quality control system and establishing the possibility of using mitochondria as a potential therapeutic target in the treatment of I/R injuries.
Collapse
Affiliation(s)
- Manli Zhou
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China,
| | - Yunfeng Yu
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Xiaoxin Luo
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Jianzhang Wang
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Xiaodong Lan
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Pei Liu
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Yu Feng
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China
| | - Weixiong Jian
- College of Traditional Chinese Medicine, Hunan University of Traditional Chinese Medicine, Changsha, China.,National Key Discipline of Traditional Chinese Medicine Diagnostics, Hunan Provincial Key Laboratory, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
36
|
A Novel Plant-Produced Asialo-rhuEPO Protects Brain from Ischemic Damage Without Erythropoietic Action. Transl Stroke Res 2021; 13:338-354. [PMID: 34553324 PMCID: PMC10068895 DOI: 10.1007/s12975-021-00943-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/09/2021] [Accepted: 09/03/2021] [Indexed: 12/14/2022]
Abstract
Mammalian cell-produced recombinant human erythropoietin (rhuEPOM) has been shown to be a multimodal neuroprotectant targeting an array of key pathological mechanisms in experimental stroke models. However, the rhuEPOM clinical trials were terminated due to increased risk of thrombosis, largely ascribed to its erythropoietic function. We recently took advantage of a plant-based expression system lacking sialylation capacity to produce asialo-rhuEPOP, a rhuEPO derivative without sialic acid residues. In the present study, we proved that asialo-rhuEPOP is non-erythropoietic by repeated intravenous injection (44 μg/kg bw) in mice showing no increase in hemoglobin levels and red blood cell counts, and confirmed that it is non-immunogenic by measuring humoral response after immunizing the mice. We demonstrate that it is neuroprotective in a cerebral ischemia and reperfusion (I/R) mouse model, exhibiting ~ 50% reduction in cerebral infarct volume and edema, and significant improvement in neurological deficits and histopathological outcome. Our studies further revealed that asialo-rhuEPOP, like rhuEPOM, displays pleiotropic neuroprotective effects, including restoring I/R-interrupted mitochondrial fission and fusion proteins, preventing I/R injury-induced increase in mitophagy and autophagy markers, and inhibiting apoptosis to benefit nerve cell survival. Most importantly, asialo-rhuEPOP lacking erythropoietic activity and immunogenicity holds great translational potential as a multimodal neuroprotectant for stroke treatment.
Collapse
|
37
|
L-Methionine Protects against Oxidative Stress and Mitochondrial Dysfunction in an In Vitro Model of Parkinson's Disease. Antioxidants (Basel) 2021; 10:antiox10091467. [PMID: 34573099 PMCID: PMC8469212 DOI: 10.3390/antiox10091467] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 01/31/2023] Open
Abstract
Methionine is an aliphatic, sulfur-containing, essential amino acid that has been demonstrated to have crucial roles in metabolism, innate immunity, and activation of endogenous antioxidant enzymes, including methionine sulfoxide reductase A/B and the biosynthesis of glutathione to counteract oxidative stress. Still, methionine restriction avoids altered methionine/transmethylation metabolism, thus reducing DNA damage and possibly avoiding neurodegenerative processes. In this study, we wanted to study the preventive effects of methionine in counteracting 6-hydroxydopamine (6-OHDA)-induced injury. In particular, we analyzed the protective effects of the amino acid L-methionine in an in vitro model of Parkinson's disease and dissected the underlying mechanisms compared to the known antioxidant taurine to gain insights into the potential of methionine treatment in slowing the progression of the disease by maintaining mitochondrial functionality. In addition, to ascribe the effects of methionine on mitochondria and oxidative stress, methionine sulfoxide was used in place of methionine. The data obtained suggested that an L-methionine-enriched diet could be beneficial during aging to protect neurons from oxidative imbalance and mitochondrial dysfunction, thus preventing the progression of neurodegenerative processes.
Collapse
|
38
|
Cervantes‐Silva MP, Cox SL, Curtis AM. Alterations in mitochondrial morphology as a key driver of immunity and host defence. EMBO Rep 2021; 22:e53086. [PMID: 34337844 PMCID: PMC8447557 DOI: 10.15252/embr.202153086] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/09/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
Mitochondria are dynamic organelles whose architecture changes depending on the cell's energy requirements and other signalling events. These structural changes are collectively known as mitochondrial dynamics. Mitochondrial dynamics are crucial for cellular functions such as differentiation, energy production and cell death. Importantly, it has become clear in recent years that mitochondrial dynamics are a critical control point for immune cell function. Mitochondrial remodelling allows quiescent immune cells to rapidly change their metabolism and become activated, producing mediators, such as cytokines, chemokines and even metabolites to execute an effective immune response. The importance of mitochondrial dynamics in immunity is evident, as numerous pathogens have evolved mechanisms to manipulate host cell mitochondrial remodelling in order to promote their own survival. In this review, we comprehensively address the roles of mitochondrial dynamics in immune cell function, along with modulation of host cell mitochondrial morphology during viral and bacterial infections to facilitate either pathogen survival or host immunity. We also speculate on what the future may hold in terms of therapies targeting mitochondrial morphology for bacterial and viral control.
Collapse
Affiliation(s)
- Mariana P Cervantes‐Silva
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| | - Shannon L Cox
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| | - Annie M Curtis
- School of Pharmacy and Biomedical Sciences and Tissue Engineering Research GroupRoyal College of Surgeons in IrelandDublinIreland
| |
Collapse
|
39
|
Maremanda KP, Sundar IK, Rahman I. Role of inner mitochondrial protein OPA1 in mitochondrial dysfunction by tobacco smoking and in the pathogenesis of COPD. Redox Biol 2021; 45:102055. [PMID: 34214709 PMCID: PMC8258692 DOI: 10.1016/j.redox.2021.102055] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/19/2021] [Accepted: 06/20/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Chronic lung diseases, such as chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF) are linked to several mitochondrial alterations. Cigarette smoke (CS) alters the structure and function of mitochondria. OPA1 is the main inner mitochondrial GTPase responsible for the fusion events. OPA1 undergoes proteolytic cleavage from long to short forms during acute stress and mitophagy. However, the exact role of OPA1 isoforms and related proteins during CS-induced mitophagy and COPD is not clear. METHODS Lung tissues from non-smokers, smokers, COPD and IPF were used to determine the relative expression of OPA1 and related proteins. Additionally, we used mouse lungs from chronic (6 months) CS exposure to evaluate the status of OPA1. Primary lung fibroblasts from normal and COPD patients and naked mole rat (NMR) lung fibroblasts, human fetal lung fibroblast (HFL1), mouse embryonic fibroblast from wild type (WT), OPA1-/-, MFN1 and MFN2-/- were used to determine the effect of CS on OPA1 isoforms. Various mitochondrial fusion promoters/activators (BGP-15, leflunomide, M1) and fission inhibitor (DRP1) were used to determine their effect on OPA1 status and cigarette smoke extract (CSE)-induced lung epithelial (BEAS2B) cell damage, respectively. Seahorse flux analyzer was used to determine the effect of these compounds in BEAS2B cells with and without CSE exposure. FINDINGS Short OPA1 isoforms were predominantly detected and significantly increased in COPD subjects. Acute CSE treatment in various cell lines except NMR was found to increase the conversion of long to short OPA1 isoforms. CSE treatment significantly increased mitochondrial stress-related protein SLP2 in all the cells used. OPA1 interacting partners like prohibitins (PHB1 and 2) were also altered depending on the CS exposure. Finally, BGP-15 and leflunomide treatment were able to preserve the long OPA1 isoform in cells treated with CSE. INTERPRETATION/CONCLUSION The long OPA1 isoform along with SLP2 and prohibitins play a crucial role in CS-induced lung damage, causing mitophagy/mitochondrial dysfunction in COPD, which may be used as a novel therapeutic target in COPD.
Collapse
Affiliation(s)
| | - Isaac Kirubakaran Sundar
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
40
|
Ji MH, Kreymerman A, Belle K, Ghiam BK, Muscat SR, Mahajan VB, Enns GM, Mercola M, Wood EH. The Present and Future of Mitochondrial-Based Therapeutics for Eye Disease. Transl Vis Sci Technol 2021; 10:4. [PMID: 34232272 PMCID: PMC8267180 DOI: 10.1167/tvst.10.8.4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Translational Relevance Mitochondria are viable therapeutic targets for a broad spectrum of ocular diseases.
Collapse
Affiliation(s)
- Marco H Ji
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Alexander Kreymerman
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Kinsley Belle
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Benjamin K Ghiam
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Stephanie R Muscat
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Vinit B Mahajan
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Gregory M Enns
- Department of Pediatrics, Division of Medical Genetics, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mark Mercola
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Edward H Wood
- Department of Ophthalmology, Byers Eye Institute, Stanford University School of Medicine, Palo Alto, CA, USA.,Stanford Cardiovascular Institute, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
41
|
Lopez-Crisosto C, Arias-Carrasco R, Sepulveda P, Garrido-Olivares L, Maracaja-Coutinho V, Verdejo HE, Castro PF, Lavandero S. Novel molecular insights and public omics data in pulmonary hypertension. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166200. [PMID: 34144090 DOI: 10.1016/j.bbadis.2021.166200] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 06/02/2021] [Accepted: 06/07/2021] [Indexed: 12/21/2022]
Abstract
Pulmonary hypertension is a rare disease with high morbidity and mortality which mainly affects women of reproductive age. Despite recent advances in understanding the pathogenesis of pulmonary hypertension, the high heterogeneity in the presentation of the disease among different patients makes it difficult to make an accurate diagnosis and to apply this knowledge to effective treatments. Therefore, new studies are required to focus on translational and personalized medicine to overcome the lack of specificity and efficacy of current management. Here, we review the majority of public databases storing 'omics' data of pulmonary hypertension studies, from animal models to human patients. Moreover, we review some of the new molecular mechanisms involved in the pathogenesis of pulmonary hypertension, including non-coding RNAs and the application of 'omics' data to understand this pathology, hoping that these new approaches will provide insights to guide the way to personalized diagnosis and treatment.
Collapse
Affiliation(s)
- Camila Lopez-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile; Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile
| | - Raul Arias-Carrasco
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile
| | - Pablo Sepulveda
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luis Garrido-Olivares
- Cardiovascular Surgery, Division of Surgery, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile
| | - Hugo E Verdejo
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo F Castro
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8380492, Chile; Division of Cardiovascular Diseases, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago 8380492, Chile; Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA.
| |
Collapse
|
42
|
Metronomic 5-Fluorouracil Delivery Primes Skeletal Muscle for Myopathy but Does Not Cause Cachexia. Pharmaceuticals (Basel) 2021; 14:ph14050478. [PMID: 34067869 PMCID: PMC8156038 DOI: 10.3390/ph14050478] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 12/22/2022] Open
Abstract
Skeletal myopathy encompasses both atrophy and dysfunction and is a prominent event in cancer and chemotherapy-induced cachexia. Here, we investigate the effects of a chemotherapeutic agent, 5-fluorouracil (5FU), on skeletal muscle mass and function, and whether small-molecule therapeutic candidate, BGP-15, could be protective against the chemotoxic challenge exerted by 5FU. Additionally, we explore the molecular signature of 5FU treatment. Male Balb/c mice received metronomic tri-weekly intraperitoneal delivery of 5FU (23 mg/kg), with and without BGP-15 (15 mg/kg), 6 times in total over a 15 day treatment period. We demonstrated that neither 5FU, nor 5FU combined with BGP-15, affected body composition indices, skeletal muscle mass or function. Adjuvant BGP-15 treatment did, however, prevent the 5FU-induced phosphorylation of p38 MAPK and p65 NF-B subunit, signalling pathways involved in cell stress and inflammatory signalling, respectively. This as associated with mitoprotection. 5FU reduced the expression of the key cytoskeletal proteins, desmin and dystrophin, which was not prevented by BGP-15. Combined, these data show that metronomic delivery of 5FU does not elicit physiological consequences to skeletal muscle mass and function but is implicit in priming skeletal muscle with a molecular signature for myopathy. BGP-15 has modest protective efficacy against the molecular changes induced by 5FU.
Collapse
|
43
|
Zou W, Chen Q, Slone J, Yang L, Lou X, Diao J, Huang T. Nanoscopic quantification of sub-mitochondrial morphology, mitophagy and mitochondrial dynamics in living cells derived from patients with mitochondrial diseases. J Nanobiotechnology 2021; 19:136. [PMID: 33985528 PMCID: PMC8120746 DOI: 10.1186/s12951-021-00882-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
SLC25A46 mutations have been found to lead to mitochondrial hyper-fusion and reduced mitochondrial respiratory function, which results in optic atrophy, cerebellar atrophy, and other clinical symptoms of mitochondrial disease. However, it is generally believed that mitochondrial fusion is attributable to increased mitochondrial oxidative phosphorylation (OXPHOS), which is inconsistent with the decreased OXPHOS of highly-fused mitochondria observed in previous studies. In this paper, we have used the live-cell nanoscope to observe and quantify the structure of mitochondrial cristae, and the behavior of mitochondria and lysosomes in patient-derived SLC25A46 mutant fibroblasts. The results show that the cristae have been markedly damaged in the mutant fibroblasts, but there is no corresponding increase in mitophagy. This study suggests that severely damaged mitochondrial cristae might be the predominant cause of reduced OXPHOS in SLC25A46 mutant fibroblasts. This study demonstrates the utility of nanoscope-based imaging for realizing the sub-mitochondrial morphology, mitophagy and mitochondrial dynamics in living cells, which may be particularly valuable for the quick evaluation of pathogenesis of mitochondrial morphological abnormalities. ![]()
Collapse
Affiliation(s)
- Weiwei Zou
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Qixin Chen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.,Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Jesse Slone
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA
| | - Li Yang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Xiaoting Lou
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,School of Laboratory Medicine and Life sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| | - Taosheng Huang
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA. .,Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
44
|
Adlimoghaddam A, Odero GG, Glazner G, Turner RS, Albensi BC. Nilotinib Improves Bioenergetic Profiling in Brain Astroglia in the 3xTg Mouse Model of Alzheimer's Disease. Aging Dis 2021; 12:441-465. [PMID: 33815876 PMCID: PMC7990369 DOI: 10.14336/ad.2020.0910] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/10/2020] [Indexed: 12/27/2022] Open
Abstract
Current treatments targeting amyloid beta in Alzheimer's disease (AD) have minimal efficacy, which results in a huge unmet medical need worldwide. Accumulating data suggest that brain mitochondrial dysfunction play a critical role in AD pathogenesis. Targeting cellular mechanisms associated with mitochondrial dysfunction in AD create a novel approach for drug development. This study investigated the effects of nilotinib, as a selective tyrosine kinase inhibitor, in astroglia derived from 3xTg-AD mice versus their C57BL/6-controls. Parameters included oxygen consumption rates (OCR), ATP, cytochrome c oxidase (COX), citrate synthase (CS) activity, alterations in oxidative phosphorylation (OXPHOS), nuclear factor kappa B (NF-κB), key regulators of mitochondrial dynamics (mitofusin (Mfn1), dynamin-related protein 1 (Drp1)), and mitochondrial biogenesis (peroxisome proliferator-activated receptor gamma coactivator1-alpha (PGC-1α), calcium/calmodulin-dependent protein kinase II (CaMKII), and nuclear factor (erythroid-derived 2)-like 2 (Nrf2)). Nilotinib increased OCR, ATP, COX, Mfn1, and OXPHOS levels in 3xTg astroglia. No significant differences were detected in levels of Drp1 protein and CS activity. Nilotinib enhanced mitochondrial numbers, potentially through a CaMKII-PGC1α-Nrf2 pathway in 3xTg astroglia. Additionally, nilotinib-induced OCR increases were reduced in the presence of the NF-κB inhibitor, Bay11-7082. The data suggest that NF-κB signaling is intimately involved in nilotinib-induced changes in bioenergetics in 3xTg brain astroglia. Nilotinib increased translocation of the NF-κB p50 subunit into the nucleus of 3xTg astroglia that correlates with an increased expression and activation of NF-κB. The current findings support a role for nilotinib in improving mitochondrial function and suggest that astroglia may be a key therapeutic target in treating AD.
Collapse
Affiliation(s)
- Aida Adlimoghaddam
- 1Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada
| | - Gary G Odero
- 1Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada
| | - Gordon Glazner
- 1Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada.,2Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - R Scott Turner
- 3Department of Neurology, Georgetown University, Washington, DC, USA
| | - Benedict C Albensi
- 1Division of Neurodegenerative Disorders, St. Boniface Hospital Research, Winnipeg, MB, Canada.,2Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
45
|
Jia J, Jin H, Nan D, Yu W, Huang Y. New insights into targeting mitochondria in ischemic injury. Apoptosis 2021; 26:163-183. [PMID: 33751318 DOI: 10.1007/s10495-021-01661-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
Stroke is the leading cause of adult disability and death worldwide. Mitochondrial dysfunction has been recognized as a marker of neuronal death during ischemic stroke. Maintaining the function of mitochondria is important for improving the survival of neurons and maintaining neuronal function. Damaged mitochondria induce neuronal cell apoptosis by releasing reactive oxygen species (ROS) and pro-apoptotic factors. Mitochondrial fission and fusion processes and mitophagy are of great importance to mitochondrial quality control. This paper reviews the dynamic changes in mitochondria, the roles of mitochondria in different cell types, and related signaling pathways in ischemic stroke. This review describes in detail the role of mitochondria in the process of neuronal injury and protection in cerebral ischemia, and integrates neuroprotective drugs targeting mitochondria in recent years, which may provide a theoretical basis for the progress of treatment of ischemic stroke. The potential of mitochondrial-targeted therapy is also emphasized, which provides valuable insights for clinical research.
Collapse
Affiliation(s)
- Jingjing Jia
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Ding Nan
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Weiwei Yu
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China
| | - Yining Huang
- Department of Neurology, Peking University First Hospital, No.8 Xishiku Street, Xicheng District, Beijing, 100034, China.
| |
Collapse
|
46
|
Horvath O, Ordog K, Bruszt K, Kalman N, Kovacs D, Radnai B, Gallyas F, Toth K, Halmosi R, Deres L. Modulation of Mitochondrial Quality Control Processes by BGP-15 in Oxidative Stress Scenarios: From Cell Culture to Heart Failure. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6643871. [PMID: 33728024 PMCID: PMC7937466 DOI: 10.1155/2021/6643871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/15/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
Heart failure (HF) is a complex chronic clinical disease characterized by among others the damage of the mitochondrial network. The disruption of the mitochondrial quality control and the imbalance in fusion-fission processes lead to a lack of energy supply and, finally, to cell death. BGP-15 (O-[3-piperidino-2-hydroxy-1-propyl]-nicotinic acid amidoxime dihydrochloride) is an insulin sensitizer molecule and has a cytoprotective effect in a wide variety of experimental models. In our recent work, we aimed to clarify the mitochondrial protective effects of BGP-15 in a hypertension-induced heart failure model and "in vitro." Spontaneously hypertensive rats (SHRs) received BGP-15 or placebo for 18 weeks. BGP-15 treatment preserved the normal mitochondrial ultrastructure and enhanced the mitochondrial fusion. Neonatal rat cardiomyocytes (NRCMs) were stressed by hydrogen-peroxide. BGP-15 treatment inhibited the mitochondrial fission processes, promoted mitochondrial fusion, maintained the integrity of the mitochondrial genome, and moreover enhanced the de novo biogenesis of the mitochondria. As a result of these effects, BGP-15 treatment also supports the maintenance of mitochondrial function through the preservation of the mitochondrial structure during hydrogen peroxide-induced oxidative stress as well as in an "in vivo" heart failure model. It offers the possibility, which pharmacological modulation of mitochondrial quality control under oxidative stress could be a novel therapeutic approach in heart failure.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cell Culture Techniques
- Citrate (si)-Synthase/metabolism
- DNA/metabolism
- DNA Damage
- DNA, Mitochondrial/genetics
- Dynamins/metabolism
- Electron Transport/drug effects
- Energy Metabolism/drug effects
- Genome, Mitochondrial
- Heart Failure/etiology
- Heart Failure/pathology
- Hypertension/complications
- Male
- Membrane Potential, Mitochondrial/drug effects
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/ultrastructure
- Mitochondrial Dynamics
- Mitochondrial Proteins/metabolism
- Myocardium/pathology
- Myocardium/ultrastructure
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/ultrastructure
- Natriuretic Peptide, Brain/metabolism
- Organelle Biogenesis
- Oxidative Stress/drug effects
- Oximes/administration & dosage
- Oximes/chemistry
- Oximes/pharmacology
- Oxygen Consumption/drug effects
- Piperidines/administration & dosage
- Piperidines/chemistry
- Piperidines/pharmacology
- Rats, Inbred SHR
- Rats, Inbred WKY
- Rats
Collapse
Affiliation(s)
- Orsolya Horvath
- 1st Department of Medicine, University of Pecs, Medical School, Pecs, Hungary
- Szentágothai Research Centre, University of Pecs, Pecs, Hungary
| | - Katalin Ordog
- 1st Department of Medicine, University of Pecs, Medical School, Pecs, Hungary
- Szentágothai Research Centre, University of Pecs, Pecs, Hungary
| | - Kitti Bruszt
- 1st Department of Medicine, University of Pecs, Medical School, Pecs, Hungary
- Szentágothai Research Centre, University of Pecs, Pecs, Hungary
| | - Nikoletta Kalman
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Dominika Kovacs
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Balazs Radnai
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
| | - Ferenc Gallyas
- Szentágothai Research Centre, University of Pecs, Pecs, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Pecs, Hungary
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
| | - Kalman Toth
- 1st Department of Medicine, University of Pecs, Medical School, Pecs, Hungary
- Szentágothai Research Centre, University of Pecs, Pecs, Hungary
| | - Robert Halmosi
- 1st Department of Medicine, University of Pecs, Medical School, Pecs, Hungary
- Szentágothai Research Centre, University of Pecs, Pecs, Hungary
| | - Laszlo Deres
- 1st Department of Medicine, University of Pecs, Medical School, Pecs, Hungary
- Szentágothai Research Centre, University of Pecs, Pecs, Hungary
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
| |
Collapse
|
47
|
Misrani A, Tabassum S, Yang L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer's Disease. Front Aging Neurosci 2021; 13:617588. [PMID: 33679375 PMCID: PMC7930231 DOI: 10.3389/fnagi.2021.617588] [Citation(s) in RCA: 256] [Impact Index Per Article: 85.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Mitochondria play a pivotal role in bioenergetics and respiratory functions, which are essential for the numerous biochemical processes underpinning cell viability. Mitochondrial morphology changes rapidly in response to external insults and changes in metabolic status via fission and fusion processes (so-called mitochondrial dynamics) that maintain mitochondrial quality and homeostasis. Damaged mitochondria are removed by a process known as mitophagy, which involves their degradation by a specific autophagosomal pathway. Over the last few years, remarkable efforts have been made to investigate the impact on the pathogenesis of Alzheimer’s disease (AD) of various forms of mitochondrial dysfunction, such as excessive reactive oxygen species (ROS) production, mitochondrial Ca2+ dyshomeostasis, loss of ATP, and defects in mitochondrial dynamics and transport, and mitophagy. Recent research suggests that restoration of mitochondrial function by physical exercise, an antioxidant diet, or therapeutic approaches can delay the onset and slow the progression of AD. In this review, we focus on recent progress that highlights the crucial role of alterations in mitochondrial function and oxidative stress in the pathogenesis of AD, emphasizing a framework of existing and potential therapeutic approaches.
Collapse
Affiliation(s)
- Afzal Misrani
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Sidra Tabassum
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| |
Collapse
|
48
|
Horvath O, Ordog K, Bruszt K, Deres L, Gallyas F, Sumegi B, Toth K, Halmosi R. BGP-15 Protects against Heart Failure by Enhanced Mitochondrial Biogenesis and Decreased Fibrotic Remodelling in Spontaneously Hypertensive Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1250858. [PMID: 33564362 PMCID: PMC7867468 DOI: 10.1155/2021/1250858] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/18/2020] [Accepted: 01/15/2021] [Indexed: 01/06/2023]
Abstract
Heart failure (HF) is a complex clinical syndrome with poor clinical outcomes despite the growing number of therapeutic approaches. It is characterized by interstitial fibrosis, cardiomyocyte hypertrophy, activation of various intracellular signalling pathways, and damage of the mitochondrial network. Mitochondria are responsible for supplying the energy demand of cardiomyocytes; therefore, the damage of the mitochondrial network causes cellular dysfunction and finally leads to cell death. BGP-15, a hydroxylamine derivative, is an insulin-sensitizer molecule and has a wide range of cytoprotective effects in animal as well as in human studies. Our recent work was aimed at examining the effects of BGP-15 in a chronic hypertension-induced heart failure model. 15-month-old male SHRs were used in our experiment. The SHR-Baseline group represented the starting point (n = 7). Animals received BGP-15 (SHR-B, n = 7) or placebo (SHR-C, n = 7) for 18 weeks. WKY rats were used as age-matched normotensive controls (n = 7). The heart function was monitored by echocardiography. Histological preparations were made from cardiac tissue. The levels of signalling proteins were determined by Western blot. At the end of the study, systolic and diastolic cardiac function was preserved in the BGP-treated animals. BGP-15 decreased the interstitial collagen deposition via decreasing the activity of TGFβ/Smad signalling factors and prevented the cardiomyocyte hypertrophy in hypertensive animals. BGP-15 enhanced the prosurvival signalling pathways (Akt/Gsk3β). The treatment increased the activity of MKP1 and decreased the activity of p38 and JNK signalling routes. The mitochondrial mass of cardiomyocytes was also increased in BGP-15-treated SHR animals due to the activation of mitochondrial biogenesis. The mitigation of remodelling processes and the preserved systolic cardiac function in hypertension-induced heart failure can be a result-at least partly-of the enhanced mitochondrial biogenesis caused by BGP-15.
Collapse
Affiliation(s)
- Orsolya Horvath
- 1st Department of Medicine, University of Pecs, Medical School, Hungary
- Szentágothai Research Centre, University of Pecs, Hungary
| | - Katalin Ordog
- 1st Department of Medicine, University of Pecs, Medical School, Hungary
- Szentágothai Research Centre, University of Pecs, Hungary
| | - Kitti Bruszt
- 1st Department of Medicine, University of Pecs, Medical School, Hungary
- Szentágothai Research Centre, University of Pecs, Hungary
| | - Laszlo Deres
- 1st Department of Medicine, University of Pecs, Medical School, Hungary
- Szentágothai Research Centre, University of Pecs, Hungary
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
| | - Ferenc Gallyas
- Szentágothai Research Centre, University of Pecs, Hungary
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Hungary
| | - Balazs Sumegi
- Szentágothai Research Centre, University of Pecs, Hungary
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
- Department of Biochemistry and Medical Chemistry, University of Pecs, Medical School, Hungary
| | - Kalman Toth
- 1st Department of Medicine, University of Pecs, Medical School, Hungary
- Szentágothai Research Centre, University of Pecs, Hungary
| | - Robert Halmosi
- 1st Department of Medicine, University of Pecs, Medical School, Hungary
- Szentágothai Research Centre, University of Pecs, Hungary
| |
Collapse
|
49
|
He Z, Zhang Y, Khan AR, Ji J, Yu A, Zhai G. A novel progress of drug delivery system for organelle targeting in tumour cells. J Drug Target 2020; 29:12-28. [PMID: 32698651 DOI: 10.1080/1061186x.2020.1797051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
At present, malignant tumours have become one of the most serious diseases that endanger human health. According to a survey on causes of death in Chinese population in early 1990s, the malignant tumours were the second leading cause of death. In the treatment of tumours, the ideal situation is that drugs should target and accumulate at tumour sites and destroy tumour cells specifically, without affecting normal cells and stem cells with regenerative capacity. This requires drugs to be specifically transported to the target organs, tissues, cells, and even specific organelles, like mitochondria, nuclei, lysosomes, endoplasmic reticulum (ER), and Golgi apparatus (GA). The nano drug delivery system can not only protect drugs from degradation but also facilitate functional modification and targeted drug delivery to the tumour site. This article mainly reviews the targeting of nano drug delivery systems to tumour cytoplasmic matrix, nucleus, mitochondria, ER, and lysosomes. Organelle-specific drug delivery system will be a major mean of targeting drug delivery with lower toxicity, less dosage and higher drug concentration in tumour cells.
Collapse
Affiliation(s)
- Zhijing He
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Yanan Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Abdur Rauf Khan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Aihua Yu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
50
|
Axonal transport dysfunction of mitochondria in traumatic brain injury: A novel therapeutic target. Exp Neurol 2020; 329:113311. [PMID: 32302676 DOI: 10.1016/j.expneurol.2020.113311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/27/2020] [Accepted: 04/10/2020] [Indexed: 01/05/2023]
|