1
|
Skawratananond S, Xiong DX, Zhang C, Tonk S, Pinili A, Delacruz B, Pham P, Smith SC, Navab R, Reddy PH. Mitophagy in Alzheimer's disease and other metabolic disorders: A focus on mitochondrial-targeted therapeutics. Ageing Res Rev 2025; 108:102732. [PMID: 40122398 DOI: 10.1016/j.arr.2025.102732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025]
Abstract
Mitochondria, as central regulators of cellular processes such as energy production, apoptosis, and metabolic homeostasis, are essential to cellular function and health. The maintenance of mitochondrial integrity, especially through mitophagy-the selective removal of impaired mitochondria-is crucial for cellular homeostasis. Dysregulation of mitochondrial function, dynamics, and biogenesis is linked to neurodegenerative and metabolic diseases, notably Alzheimer's disease (AD), which is increasingly recognized as a metabolic disorder due to its shared pathophysiologic features: insulin resistance, oxidative stress, and chronic inflammation. In this review, we highlight recent advancements in pharmacological interventions, focusing on agents that modulate mitophagy, mitochondrial uncouplers that reduce oxidative phosphorylation, compounds that directly scavenge reactive oxygen species to alleviate oxidative stress, and molecules that ameliorate amyloid beta plaque accumulation and phosphorylated tau pathology. Additionally, we explore dietary and lifestyle interventions-MIND and ketogenic diets, caloric restriction, physical activity, hormone modulation, and stress management-that complement pharmacological approaches and support mitochondrial health. Our review underscores mitochondria's central role in the pathogenesis and potential treatment of neurodegenerative and metabolic diseases, particularly AD. By advocating for an integrated therapeutic model that combines pharmacological and lifestyle interventions, we propose a comprehensive approach aimed at mitigating mitochondrial dysfunction and improving clinical outcomes in these complex, interrelated diseases.
Collapse
Affiliation(s)
- Shadt Skawratananond
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Daniel X Xiong
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX 78712, United States.
| | - Charlie Zhang
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Sahil Tonk
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Aljon Pinili
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Brad Delacruz
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Patrick Pham
- Honors College, Texas Tech University, Lubbock, TX 79401, United States; Department of Biology, Texas Tech University, Lubbock, TX 79401, USA, Texas Tech University, Lubbock, TX 79401, United States.
| | - Shane C Smith
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States.
| | - Rahul Navab
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Internal Medicine, PES Institute of Medical Sciences and Research, Kuppam, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, United States; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, United States; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
2
|
Nuñez-Selles AJ, Nuñez-Musa RA, Guillen-Marmolejos RA. Linking oxidative stress biomarkers to disease progression and antioxidant therapy in hypertension and diabetes mellitus. Front Mol Biosci 2025; 12:1611842. [PMID: 40492113 PMCID: PMC12146192 DOI: 10.3389/fmolb.2025.1611842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 06/11/2025] Open
Abstract
Oxidative stress (OS) is increasingly recognized as a key factor linking hypertension (HTN) and diabetes mellitus (DM). This review summarizes recent evidence regarding the dual role of OS as both an instigator and an amplifier of cardiometabolic dysfunction. In HTN, reactive oxygen species (ROS) produced by NADPH oxidases (NOXs) and mitochondrial dysfunction contribute to endothelial impairment and vascular remodeling. In DM, hyperglycemia-induced ROS production worsens beta-cell failure and insulin resistance through pathways such as the AGE-RAGE signaling, protein kinase C (PKC) activation, and the polyol pathway. Clinically validated biomarkers of OS, such as F2-isoprostanes (which indicate lipid peroxidation), 8-OHdG (which indicates DNA damage), and the activities of redox enzymes like superoxide dismutase (SOD) and glutathione peroxidase (GPx), show strong correlations with disease progression and end-organ complications. Despite promising preclinical results, the application of antioxidant therapies in clinical settings has faced challenges due to inconsistent outcomes, highlighting the need for targeted approaches. Emerging strategies include: 1. Mitochondria-targeted antioxidants to enhance vascular function in resistant HTN; 2. Nrf2 activators to restore redox balance in early diabetes; and 3. Specific inhibitors of NOX isoforms. We emphasize three transformative areas of research: (i) the interaction between the microbiome and ROS, where modifying gut microbiota can reduce systemic OS; (ii) the use of nanotechnology to deliver antioxidants directly to pancreatic islets or atherosclerotic plaques; and (iii) phenotype-specific diagnosis and therapy guided by redox biomarkers and genetic profiling (for example, KEAP1/NRF2 polymorphisms). Integrating these advances with lifestyle modifications, such as following a Mediterranean diet and exercising regularly, may provide additional benefits. This review outlines a mechanistic framework for targeting OS in the comorbidity of HTN and DM while identifying critical knowledge gaps, particularly regarding the timing of antioxidant signaling and the development of personalized redox medicine, which may serve as a reference for researchers and clinicians working in this area.
Collapse
Affiliation(s)
- Alberto J. Nuñez-Selles
- Research Division, Universidad Nacional “Pedro Henríquez Ureña” (UNPHU), Santo Domingo, Dominican Republic
| | | | | |
Collapse
|
3
|
Tian Y, Kang L, Ha NT, Deng J, Liu D. Hydrogen peroxide in midbrain sleep neurons regulates sleep homeostasis. Cell Metab 2025:S1550-4131(25)00254-2. [PMID: 40378838 DOI: 10.1016/j.cmet.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 01/13/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
Sleep could protect animals from oxidative damage, yet the dynamic interplay between the redox state and sleep homeostasis remains unclear. Here, we show that acute sleep deprivation (SD) in mice caused a general increase in brain oxidation, particularly in sleep-promoting regions. In vivo imaging of intracellular hydrogen peroxide (H2O2) real-time dynamics revealed that in nigra sleep neurons, the increase in cytosolic but not mitochondrial H2O2 reflects sleep debt and tracks spontaneous wakefulness by positively correlating with wake duration. By controllably manipulating intraneuronal H2O2, we discovered that H2O2 elevation is required for compensatory sleep and causally promotes sleep initiation, at least partly dependent on transient receptor potential melastatin 2 (TRPM2) channel. However, excessive H2O2 induced brain inflammation and sleep fragmentation. Together, our study demonstrates intraneuronal H2O2 as a crucial signaling molecule that translates brain redox imbalance into sleep drive and underscores the significance of oxidative eustress in sleep homeostasis.
Collapse
Affiliation(s)
- Yujing Tian
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Luwei Kang
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ngoc T Ha
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Danqian Liu
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
4
|
Losarwar S, Pancholi B, Babu R, Garabadu D. Mitochondria-dependent innate immunity: A potential therapeutic target in Flavivirus infection. Int Immunopharmacol 2025; 154:114551. [PMID: 40158432 DOI: 10.1016/j.intimp.2025.114551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/22/2025] [Indexed: 04/02/2025]
Abstract
Mitochondria, known as the powerhouse of cells, play a crucial role in host innate immunity during flavivirus infections such as Dengue, Zika, West Nile, and Japanese Encephalitis Virus. Mitochondrial antiviral signaling protein (MAVS) resides on the outer mitochondrial membrane which is triggered by viral RNA recognition by RIG-I-like receptors (RLRs). This activation induces IRF3 and NF-κB signaling, resulting in type I interferon (IFN) production and antiviral responses. Upon flavivirus infection, mitochondrial stress and dysfunction may lead to the release of mitochondrial DNA (mtDNA) into the cytoplasm, which serves as a damage-associated molecular pattern (DAMP). Cytosolic mtDNA is sensed by cGAS (cyclic GMP-AMP synthase), leading to the activation of the STING (Stimulator of Interferon Genes) pathway to increase IFN production and expand inflammation. Flaviviral proteins control mitochondrial morphology by controlling mitochondrial fission (MF) and fusion (MFu), disrupting mitochondrial dynamics (MD) to inhibit MAVS signaling and immune evasion. Flaviviral proteins also cause oxidative stress, resulting in the overproduction of reactive oxygen species (ROS), which triggers NLRP3 inflammasome activation and amplifies inflammation. Additionally, flaviviruses drive metabolic reprogramming by shifting host cell metabolism from oxidative phosphorylation (OxPhos) to glycolysis and fatty acid synthesis, creating a pro-replicative environment that supports viral replication and persistence. Thus, the present review explores the complex interaction between MAVS, mtDNA, and the cGAS-STING pathway, which is key to the innate immune response against flavivirus infections. Understanding these mechanisms opens new avenues in therapeutic interventions in targeting mitochondrial pathways to enhance antiviral immunity and mitigate viral infection.
Collapse
Affiliation(s)
- Saurabh Losarwar
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | | | - Raja Babu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India
| | - Debapriya Garabadu
- Department of Pharmacology, Central University of Punjab, Bhatinda 151401, India.
| |
Collapse
|
5
|
Pang M, Wang S, Shi T, Chen J. Overview of MitoQ on prevention and management of cardiometabolic diseases: a scoping review. Front Cardiovasc Med 2025; 12:1506460. [PMID: 40134978 PMCID: PMC11934253 DOI: 10.3389/fcvm.2025.1506460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Background The exploration of mitochondrial-targeted antioxidants represented a burgeoning field of research with significant implications for cardiometabolic diseases (CMD). The studies reviewed in this scoping analysis collectively highlighted the effect of MitoQ on prevention and management of CMD and underlying mechanisms were discussed, mainly including cardiovascular diseases (CVDs), liver health and others. Methods This scoping review aimed to synthesize current research on the health impacts of MitoQ on CMD, focusing primarily on human-based clinical trials. While the primary focus was on human trials, in vivo and in vitro studies were referenced as supplementary material to provide a broader understanding of MitoQ's mechanisms and potential effects. Results This scoping review had synthesized the findings that collectively contributed to the understanding of mitochondrial-targeted antioxidants and their role in CMD. Conclusion The synthesis of these findings illustrated a broad spectrum of benefits ranging from enhanced insulin secretion to improved lipid profiles and mitochondrial function, yet the path to clinical application required further investigation on appropriate doses and populations.
Collapse
Affiliation(s)
- Mingli Pang
- School of Public Affairs, Zhejiang University, Hangzhou, China
- National Institute for Health Innovation, School of Population Health, The University of Auckland, Auckland, New Zealand
| | - Shidi Wang
- Department of Social Medicine and Health Care Management, Fudan University, Shanghai, China
| | - Tianyi Shi
- Faculty of Medical and Health Sciences, School of Population Health, The University of Auckland, Auckland, New Zealand
| | - Jinsong Chen
- School of Public Affairs, Zhejiang University, Hangzhou, China
- National Institute for Health Innovation, School of Population Health, The University of Auckland, Auckland, New Zealand
- Faculty of Public Administration, School of Law, Hangzhou City University, Hangzhou, China
| |
Collapse
|
6
|
Du T, Su H, Cao D, Meng Q, Zhang M, Liu Z, Li H. Mitochondria-targeted antioxidant mitoquinone mitigates vitrification-induced damage in mouse ovarian tissue by maintaining mitochondrial homeostasis via the p38 MAPK pathway. Eur J Med Res 2024; 29:593. [PMID: 39696534 DOI: 10.1186/s40001-024-02181-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024] Open
Abstract
OBJECTIVE Ovarian tissue cryopreservation has become a promising alternative for fertility preservation in cancer patients, allowing ovarian tissue to be stored for future autotransplantation. Oxidative stress damage occurring during the cryopreservation process may impact tissue quality and function. This study aims to investigate the protective effects and potential mechanisms of Mitoquinone (MitoQ), a mitochondria-targeted derivative of the antioxidant ubiquinone, during the vitrification of ovarian tissue in mice. METHODS KGN cells were treated with various concentrations (0.1, 1, 10, and 50 μM) of MitoQ to determine the optimal concentration. Female ICR mice were divided into three groups: control, conventional vitrification, and MitoQ-supplemented vitrification. Ovarian samples were cryopreserved, thawed, and assessed for tissue morphology using Hematoxylin and Eosin (H&E) staining, and mitochondrial changes using immunofluorescence, transmission electron microscopy, and Western blot analysis. RNA sequencing (RNA-seq) was employed to explore potential protective mechanisms. Autotransplantation experiments were conducted, and the long-term effects of MitoQ on ovarian function were evaluated by counting follicle numbers through H&E staining and measuring serum estradiol and AMH levels using ELISA. RESULTS MitoQ at 1 μM was found to be the optimal concentration for maintaining follicular morphology after vitrification. It effectively reduced mitochondrial oxidative damage, preserved mitochondrial morphology, and regulated the expression of mitochondrial dynamics proteins (Drp1 and Mfn2). RNA-seq and Western blot analyses revealed that MitoQ inhibited the p38 MAPK pathway, thereby reducing apoptosis. Additionally, autotransplantation experiments showed that MitoQ treatment significantly increased follicle counts, estradiol (E2), and anti-Müllerian hormone (AMH) levels compared to conventional vitrification. CONCLUSIONS MitoQ effectively mitigates vitrification-induced oxidative damage, maintains mitochondrial homeostasis, and preserves both follicular reserve and endocrine function. These findings suggest that MitoQ is a valuable adjunct in ovarian tissue cryopreservation and could significantly improve fertility preservation outcomes for cancer patients.
Collapse
Affiliation(s)
- Tianqi Du
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China
| | - Han Su
- Key Laboratory of Reproductive Medicine and Offspring Health, Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhouing, China
- Obstetrics and Gynecology Department, BENQ Medical Center, Nanjing, China
| | - Dan Cao
- Department of Pathology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhouing, China
| | - Qingxia Meng
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China
| | - Ming Zhang
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China
- Key Laboratory of Reproductive Medicine and Offspring Health, Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhouing, China
| | - Zhenxing Liu
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China
| | - Hong Li
- Center for Human Reproduction and Genetics, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhouing, China.
| |
Collapse
|
7
|
Liu JC, Zhao XY, Wu ML, Shi YF, Huang ZP, Fang LP, Zhu C, Peng X, Shi ZL, Lan LJ, Ji WL, Luo L, Feng L, Zhang ZL, Xu DE, Li S, Qin ZH, Sun YY, Schachner M, Ma QH. GPR50 regulates neuronal development as a mitophagy receptor. Cell Death Dis 2024; 15:591. [PMID: 39143050 PMCID: PMC11324738 DOI: 10.1038/s41419-024-06978-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Abstract
Neurons rely heavily on high mitochondrial metabolism to provide sufficient energy for proper development. However, it remains unclear how neurons maintain high oxidative phosphorylation (OXPHOS) during development. Mitophagy plays a pivotal role in maintaining mitochondrial quality and quantity. We herein describe that G protein-coupled receptor 50 (GPR50) is a novel mitophagy receptor, which harbors the LC3-interacting region (LIR) and is required in mitophagy under stress conditions. Although it does not localize in mitochondria under normal culturing conditions, GPR50 is recruited to the depolarized mitochondrial membrane upon mitophagy stress, which marks the mitochondrial portion and recruits the assembling autophagosomes, eventually facilitating the mitochondrial fragments to be engulfed by the autophagosomes. Mutations Δ502-505 and T532A attenuate GPR50-mediated mitophagy by disrupting the binding of GPR50 to LC3 and the mitochondrial recruitment of GPR50. Deficiency of GPR50 causes the accumulation of damaged mitochondria and disrupts OXPHOS, resulting in insufficient ATP production and excessive ROS generation, eventually impairing neuronal development. GPR50-deficient mice exhibit impaired social recognition, which is rescued by prenatal treatment with mitoQ, a mitochondrially antioxidant. The present study identifies GPR50 as a novel mitophagy receptor that is required to maintain mitochondrial OXPHOS in developing neurons.
Collapse
Affiliation(s)
- Ji-Chuan Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Xiu-Yun Zhao
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Ming-Lei Wu
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Yi-Fan Shi
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Ze-Ping Huang
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Li-Pao Fang
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Chao Zhu
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Xuan Peng
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
- School of Public Health, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Zi-Ling Shi
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Li-Jun Lan
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Wen-Li Ji
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Li Luo
- School of Physical Education and Sports Science, Soochow University, Suzhou, 215021, China
| | - Lei Feng
- Monash Suzhou Research Institute, Suzhou, 215000, China
| | - Zeng-Li Zhang
- School of Public Health, Soochow University, Suzhou, Jiangsu, 215021, China
| | - De-En Xu
- The Wuxi No.2 People Hospital, Wuxi, 214002, Jiangsu, China
| | - Shao Li
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R & D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, 116044, China
| | - Zheng-Hong Qin
- Institute of Health Technology, Suzhou Gaobo Vocational College, Suzhou High-Technology District, Science & Technology Town, 5 Qingshan Road, Suzhou, Jiangsu, 215163, PR China
| | - Yan-Yun Sun
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong, 515041, China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08854, USA
| | - Quan-Hong Ma
- Department of Neurology and Clinical Research Center of Neurological Disease, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215021, China.
- Institute of Neuroscience & Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-psycho-Diseases, Soochow University, Suzhou, Jiangsu, 215021, China.
| |
Collapse
|
8
|
Pan X, Hao E, Zhang F, Wei W, Du Z, Yan G, Wang X, Deng J, Hou X. Diabetes cardiomyopathy: targeted regulation of mitochondrial dysfunction and therapeutic potential of plant secondary metabolites. Front Pharmacol 2024; 15:1401961. [PMID: 39045049 PMCID: PMC11263127 DOI: 10.3389/fphar.2024.1401961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a specific heart condition in diabetic patients, which is a major cause of heart failure and significantly affects quality of life. DCM is manifested as abnormal cardiac structure and function in the absence of ischaemic or hypertensive heart disease in individuals with diabetes. Although the development of DCM involves multiple pathological mechanisms, mitochondrial dysfunction is considered to play a crucial role. The regulatory mechanisms of mitochondrial dysfunction mainly include mitochondrial dynamics, oxidative stress, calcium handling, uncoupling, biogenesis, mitophagy, and insulin signaling. Targeting mitochondrial function in the treatment of DCM has attracted increasing attention. Studies have shown that plant secondary metabolites contribute to improving mitochondrial function and alleviating the development of DCM. This review outlines the role of mitochondrial dysfunction in the pathogenesis of DCM and discusses the regulatory mechanism for mitochondrial dysfunction. In addition, it also summarizes treatment strategies based on plant secondary metabolites. These strategies targeting the treatment of mitochondrial dysfunction may help prevent and treat DCM.
Collapse
Affiliation(s)
- Xianglong Pan
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Erwei Hao
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Fan Zhang
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wei Wei
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zhengcai Du
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Guangli Yan
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Xijun Wang
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jiagang Deng
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Xiaotao Hou
- Department of Pharmaceutical, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Collaborative Innovation Center for Research on Functional Ingredients of Agricultural Residues, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of TCM Formulas Theory and Transformation for Damp Diseases, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
9
|
Xiao Liang K. Interplay of mitochondria and diabetes: Unveiling novel therapeutic strategies. Mitochondrion 2024; 75:101850. [PMID: 38331015 DOI: 10.1016/j.mito.2024.101850] [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: 08/16/2023] [Revised: 12/26/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
The interplay between mitochondrial function and diabetes has gained significant attention due to its crucial role in the pathogenesis and progression of the disease. Mitochondria, known as the cellular powerhouses, are essential for glucose metabolism. Dysfunction of these organelles has been implicated in the development of insulin resistance and beta-cell failure, both prominent features of diabetes. This comprehensive review explores the intricate mechanisms involved, including the generation of reactive oxygen species and the impact of mitochondrial DNA (mtDNA) mutations. Moreover, the review delves into emerging therapeutic strategies that specifically target mitochondria, such as mitochondria-targeted antioxidants, agents promoting mitochondrial biogenesis, and compounds modulating mitochondrial dynamics. The potential of these novel approaches is critically evaluated, taking into account their benefits and limitations, to provide a well-rounded perspective. Ultimately, this review emphasizes the importance of advancing our understanding of mitochondrial biology to revolutionize the treatment of diabetes.
Collapse
|
10
|
Ding Z, Wei Y, Peng J, Wang S, Chen G, Sun J. The Potential Role of C-Reactive Protein in Metabolic-Dysfunction-Associated Fatty Liver Disease and Aging. Biomedicines 2023; 11:2711. [PMID: 37893085 PMCID: PMC10603830 DOI: 10.3390/biomedicines11102711] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), recently redefined as metabolic-dysfunction-associated fatty liver disease (MASLD), is liver-metabolism-associated steatohepatitis caused by nonalcoholic factors. NAFLD/MASLD is currently the most prevalent liver disease in the world, affecting one-fourth of the global population, and its prevalence increases with age. Current treatments are limited; one important reason hindering drug development is the insufficient understanding of the onset and pathogenesis of NAFLD/MASLD. C-reactive protein (CRP), a marker of inflammation, has been linked to NAFLD and aging in recent studies. As a conserved acute-phase protein, CRP is widely characterized for its host defense functions, but the link between CRP and NAFLD/MASLD remains unclear. Herein, we discuss the currently available evidence for the involvement of CRP in MASLD to identify areas where further research is needed. We hope this review can provide new insights into the development of aging-associated NAFLD biomarkers and suggest that modulation of CRP signaling is a potential therapeutic target.
Collapse
Affiliation(s)
- Zheng Ding
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Yuqiu Wei
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Jing Peng
- College of Biological Sciences, China Agricultural University, Beijing 100193, China
| | - Siyu Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Guixi Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| | - Jiazeng Sun
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100190, China
| |
Collapse
|
11
|
Tong W, Leng L, Wang Y, Guo J, Owusu FB, Zhang Y, Wang F, Li R, Li Y, Chang Y, Wang Y, Wang Q. Buyang huanwu decoction inhibits diabetes-accelerated atherosclerosis via reduction of AMPK-Drp1-mitochondrial fission axis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116432. [PMID: 37003404 DOI: 10.1016/j.jep.2023.116432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/26/2023] [Accepted: 03/22/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese drugs, including Buyang Huanwu decoction (BYHWD), have been used in traditional practice to manage cardiovascular and cerebrovascular diseases. However, the effect and mechanisms by which this decoction alleviates diabetes-accelerated atherosclerosis are unknown and require exploration. AIM OF THE STUDY This study aims to investigate the pharmacological effects of BYHWD on preventing diabetes-accelerated atherosclerosis, and elucidate its underlying mechanism. MATERIALS AND METHODS Streptozotocin (STZ)-induced diabetic ApoE-/- mice were treated with BYHWD. Atherosclerotic aortic lesions, endothelial function, mitochondrial morphology, and mitochondrial dynamics-related proteins were evaluated in isolated aortas. High glucose-exposed human umbilical endothelial cells (HUVECs) were treated with BYHWD and its components. AMPK siRNA transfection, Drp1 molecular docking, Drp1 enzyme activity measurement, and so on were used to explore and verify the mechanism. RESULT BYHWD treatment inhibited the worsening of diabetes-accelerated atherosclerosis by lessening atherosclerotic lesions in diabetic ApoE-/- mice, by impeding endothelial dysfunction under diabetic conditions, and by inhibiting mitochondrial fragmentation by lowering protein expression levels of Drp1 and mitochondrial fission-1 protein (Fis1) in diabetic aortic endothelium. In high glucose-exposed HUVECs, BYHWD treatment also downgraded reactive oxygen species, promoted nitric oxide levels, and abated mitochondrial fission by reducing protein expression levels of Drp1 and fis1, but not mitofusin-1 and optic atrophy-1. Interestingly, we found that BYHWD's protective effect against mitochondrial fission is mediated by AMPK activation-dependent reduction of Drp1 levels. The main serum chemical components of BYHWD, ferulic acid, and calycosin-7-glucoside, can reduce the expression of Drp1 by regulating AMPK, and can inhibit the activity of GTPase of Drp1. CONCLUSION The above findings support the conclusion that BYHWD suppresses diabetes-accelerated atherosclerosis by reducing mitochondrial fission through modulation of the AMPK/Drp1 pathway.
Collapse
Affiliation(s)
- Wanyu Tong
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ling Leng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, 301617, China
| | - Yucheng Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jingwen Guo
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Felix Boahen Owusu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yue Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fang Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ruiqiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, 301617, China
| | - Yuhong Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, 301617, China
| | - Yanxu Chang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Yuefei Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
12
|
Liu Y, Liu Q, Zhang Z, Yang Y, Zhou Y, Yan H, Wang X, Li X, Zhao J, Hu J, Yang S, Tian Y, Yao Y, Qiu Z, Song Y, Yang Y. The regulatory role of PI3K in ageing-related diseases. Ageing Res Rev 2023; 88:101963. [PMID: 37245633 DOI: 10.1016/j.arr.2023.101963] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 05/30/2023]
Abstract
Ageing is a physiological/pathological process accompanied by the progressive damage of cell function, triggering various ageing-related disorders. Phosphatidylinositol 3-kinase (PI3K), which serves as one of the central regulators of ageing, is closely associated with cellular characteristics or molecular features, such as genome instability, telomere erosion, epigenetic alterations, and mitochondrial dysfunction. In this review, the PI3K signalling pathway was firstly thoroughly explained. The link between ageing pathogenesis and the PI3K signalling pathway was then summarized. Finally, the key regulatory roles of PI3K in ageing-related illnesses were investigated and stressed. In summary, we revealed that drug development and clinical application targeting PI3K is one of the focal points for delaying ageing and treating ageing-related diseases in the future.
Collapse
Affiliation(s)
- Yanqing Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Qiong Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Zhe Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yaru Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yazhe Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Huanle Yan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Xin Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Xiaoru Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Jing Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Jingyan Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Shulin Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yifan Tian
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yu Yao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Zhenye Qiu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China
| | - Yanbin Song
- Department of Cardiology, Affiliated Hospital, Yan'an University, 43 North Street, Yan'an 716000, China.
| | - Yang Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an 710069, China.
| |
Collapse
|
13
|
López-Lluch G. Coenzyme Q-related compounds to maintain healthy mitochondria during aging. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 136:277-308. [PMID: 37437981 DOI: 10.1016/bs.apcsb.2023.02.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Mitochondrial dysfunction is one of the main factors that affects aging progression and many age-related diseases. Accumulation of dysfunctional mitochondria can be driven by unbalanced mito/autophagy or by decrease in mitochondrial biosynthesis and turnover. Coenzyme Q is an essential component of the mitochondrial electron transport chain and a key factor in the protection of membrane and mitochondrial DNA against oxidation. Coenzyme Q levels decay during aging and this can be considered an accelerating factor in mitochondrial dysfunction and aging progression. Supplementation with coenzyme Q is successful for some tissues and organs but not for others. For this reason, the role of coenzyme Q in systemic aging is a complex picture that needs different strategies depending on the organ considered the main objective to be addressed. In this chapter we focus on the different effects of coenzyme Q and related compounds and the probable strategies to induce endogenous synthesis to maintain healthy aging.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo, CABD-CSIC, CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide, Sevilla, Spain.
| |
Collapse
|
14
|
Barbero NM, Oller J, Sanz AB, Ramos AM, Ortiz A, Ruiz-Ortega M, Rayego-Mateos S. Mitochondrial Dysfunction in the Cardio-Renal Axis. Int J Mol Sci 2023; 24:ijms24098209. [PMID: 37175915 PMCID: PMC10179675 DOI: 10.3390/ijms24098209] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 05/15/2023] Open
Abstract
Cardiovascular disease (CVD) frequently complicates chronic kidney disease (CKD). The risk of all-cause mortality increases from 20% to 500% in patients who suffer both conditions; this is referred to as the so-called cardio-renal syndrome (CRS). Preclinical studies have described the key role of mitochondrial dysfunction in cardiovascular and renal diseases, suggesting that maintaining mitochondrial homeostasis is a promising therapeutic strategy for CRS. In this review, we explore the malfunction of mitochondrial homeostasis (mitochondrial biogenesis, dynamics, oxidative stress, and mitophagy) and how it contributes to the development and progression of the main vascular pathologies that could be affected by kidney injury and vice versa, and how this knowledge may guide the development of novel therapeutic strategies in CRS.
Collapse
Affiliation(s)
- Nerea Mendez Barbero
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Jorge Oller
- Laboratory of Vascular Pathology, IIS-Fundación Jiménez Díaz, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, 28037 Madrid, Spain
| | - Ana B Sanz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Adrian M Ramos
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Alberto Ortiz
- Spain Nephrology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
| | - Marta Ruiz-Ortega
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| | - Sandra Rayego-Mateos
- REDINREN Spain/Ricors2040, 28029 Madrid, Spain
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma, 28040 Madrid, Spain
| |
Collapse
|
15
|
Pant T, Uche N, Juric M, Bosnjak ZJ. Clinical Relevance of lncRNA and Mitochondrial Targeted Antioxidants as Therapeutic Options in Regulating Oxidative Stress and Mitochondrial Function in Vascular Complications of Diabetes. Antioxidants (Basel) 2023; 12:antiox12040898. [PMID: 37107272 PMCID: PMC10135521 DOI: 10.3390/antiox12040898] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic imbalances and persistent hyperglycemia are widely recognized as driving forces for augmented cytosolic and mitochondrial reactive oxygen species (ROS) in diabetes mellitus (DM), fostering the development of vascular complications such as diabetic nephropathy, diabetic cardiomyopathy, diabetic neuropathy, and diabetic retinopathy. Therefore, specific therapeutic approaches capable of modulating oxidative milieu may provide a preventative and/or therapeutic benefit against the development of cardiovascular complications in diabetes patients. Recent studies have demonstrated epigenetic alterations in circulating and tissue-specific long non-coding RNA (lncRNA) signatures in vascular complications of DM regulating mitochondrial function under oxidative stress. Intriguingly, over the past decade mitochondria-targeted antioxidants (MTAs) have emerged as a promising therapeutic option for managing oxidative stress-induced diseases. Here, we review the present status of lncRNA as a diagnostic biomarker and potential regulator of oxidative stress in vascular complications of DM. We also discuss the recent advances in using MTAs in different animal models and clinical trials. We summarize the prospects and challenges for the use of MTAs in treating vascular diseases and their application in translation medicine, which may be beneficial in MTA drug design development, and their application in translational medicine.
Collapse
Affiliation(s)
- Tarun Pant
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Nnamdi Uche
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Matea Juric
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Zeljko J Bosnjak
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
16
|
Akalay S, Hosgood SA. How to Best Protect Kidneys for Transplantation-Mechanistic Target. J Clin Med 2023; 12:jcm12051787. [PMID: 36902572 PMCID: PMC10003664 DOI: 10.3390/jcm12051787] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
The increasing number of patients on the kidney transplant waiting list underlines the need to expand the donor pool and improve kidney graft utilization. By protecting kidney grafts adequately from the initial ischemic and subsequent reperfusion injury occurring during transplantation, both the number and quality of kidney grafts could be improved. The last few years have seen the emergence of many new technologies to abrogate ischemia-reperfusion (I/R) injury, including dynamic organ preservation through machine perfusion and organ reconditioning therapies. Although machine perfusion is gradually making the transition to clinical practice, reconditioning therapies have not yet progressed from the experimental setting, pointing towards a translational gap. In this review, we discuss the current knowledge on the biological processes implicated in I/R injury and explore the strategies and interventions that are being proposed to either prevent I/R injury, treat its deleterious consequences, or support the reparative response of the kidney. Prospects to improve the clinical translation of these therapies are discussed with a particular focus on the need to address multiple aspects of I/R injury to achieve robust and long-lasting protective effects on the kidney graft.
Collapse
Affiliation(s)
- Sara Akalay
- Department of Development and Regeneration, Laboratory of Pediatric Nephrology, KU Leuven, 3000 Leuven, Belgium
| | - Sarah A. Hosgood
- Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
- Correspondence:
| |
Collapse
|
17
|
Ye W, Wen C, Zeng A, Hu X. Increased levels of circulating oxidized mitochondrial DNA contribute to chronic inflammation in metabolic syndrome, and MitoQ-based antioxidant therapy alleviates this DNA-induced inflammation. Mol Cell Endocrinol 2023; 560:111812. [PMID: 36334615 DOI: 10.1016/j.mce.2022.111812] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Here, the aim was to investigate the role of circulating oxidized mitochondrial DNA (ox-mtDNA) in metabolic syndrome (MetS)-associated chronic inflammation and evaluate the effect of Mito-Quinone (MitoQ)-based antioxidant therapy on inflammation. A total of 112 MetS patients and 111 healthy control individuals (HCs) were recruited. Peripheral blood was collected, and mononuclear cells (PBMCs) were separated. In a preclinical study, MitoQ, a mitochondrial-targeted antioxidant, was administered to Sprague-Dawley (SD) rats fed a high-fat diet (HFD). In vitro, H2O2- or MitoQ-treated HUVECs served as the oxidative or antioxidative cell models to detect the cell-free ox-mtDNA level. Plasma or cell-free ox-mtDNA levels were measured by qPCR. Additionally, THP-1 cells were incubated with plasma cell-free DNA (cfDNA) from MetS patients and HCs or cell-free ox-mtDNA to detect TLR9-NF-κB pathway activation. Plasma ox-mtDNA levels and TLR9 expression levels in PBMCs were increased in MetS patients. In vivo, HFD-fed rats showed elevated plasma ox-mtDNA and TLR9 expression levels in cardiac-residing immune cells, but MitoQ administration attenuated these increases. In vitro, a significant lower level of cell-free ox-mtDNA was detected in MitoQ-treated cells, compared with H2O2-treated cells. Coincubation of plasma cfDNA from MetS patients or cell-free ox-mtDNA and THP-1 cells increased TLR9-NF-κB p65 expression, and promoted IL-1β, IL-6 and IL-8 secretion in THP-1 cells. In conclusion, increased circulating ox-mtDNA contributes to chronic inflammation in MetS by activating the TLR9-NF-κB pathway. MitoQ-based antioxidant therapy effectively alleviates inflammation by reducing ox-mtDNA release.
Collapse
Affiliation(s)
- Wei Ye
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Chaowei Wen
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Aibing Zeng
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Xingzhong Hu
- Department of Clinical Laboratory Medicine, Wenzhou Central Hospital, Dingli Clinical School of Wenzhou Medical University, Wenzhou, 325000, China
| |
Collapse
|
18
|
Oxidative stress in metabolic diseases: current scenario and therapeutic relevance. Mol Cell Biochem 2023; 478:185-196. [PMID: 35764861 DOI: 10.1007/s11010-022-04496-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 06/01/2022] [Indexed: 01/17/2023]
Abstract
The metabolic syndrome is a clustering condition of increased abdominal obesity in concert with hyperglycemia, insulin resistance, hypertension, and dyslipidemia. It confers higher risk of metabolic diseases such as diabetes and ischemic heart disease and has been observed to be associated with high morbidity and mortality. It is a progressive pathological process for diabetes-induced complications and appears to be multifactorial in origin. Several preclinical, clinical, and epidemiological reports have shown a persistent link between the metabolic syndrome and oxidative stress. There is pronounced imbalance between pro-oxidants and anti-oxidants with increased production of oxidizing molecules, depletion of anti-oxidants, and consequently accumulation of protein and lipid oxidation products in the cell in metabolic syndrome. The increased cellular pro-oxidant activity also results in altered molecular pathways, mitochondrial dysfunction, deregulation in cell cycle control, chromosomal aberrations, inflammation, and overall decreased biological activity as well as impairment of the antioxidant systems. Here, the focus of our review article will be on the formation of oxidative species, the interplay between metabolic syndrome and oxidative stress, and its potential implications in therapeutic approaches.
Collapse
|
19
|
Zhu L, Luo M, Zhang Y, Fang F, Li M, An F, Zhao D, Zhang J. Free radical as a double-edged sword in disease: Deriving strategic opportunities for nanotherapeutics. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Zhang Y, Zhang J, Duan L. The role of microbiota-mitochondria crosstalk in pathogenesis and therapy of intestinal diseases. Pharmacol Res 2022; 186:106530. [DOI: 10.1016/j.phrs.2022.106530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/17/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022]
|
21
|
Yuan Q, Zeng ZL, Yang S, Li A, Zu X, Liu J. Mitochondrial Stress in Metabolic Inflammation: Modest Benefits and Full Losses. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8803404. [PMID: 36457729 PMCID: PMC9708372 DOI: 10.1155/2022/8803404] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 09/02/2023]
Abstract
Energy intake and metabolic balance are the pillars of health preservation. Overnutrition causes nonspecific, persistently low inflammatory state known as metabolic inflammation. This condition contributes to the pathophysiology of various metabolic disorders, such as atherosclerosis, obesity, diabetes mellitus, and metabolic syndrome. The mitochondria maintain the balance of energy metabolism. Excessive energy stress can lead to mitochondrial dysfunction, which promotes metabolic inflammation. The inflammatory environment further impairs mitochondrial function. Accordingly, excellent organism design keeps the body metabolically healthy in the context of mitochondrial dysfunction, and moderate mitochondrial stress can have a beneficial effect. This review summarises the research progress on the multifaceted characterisation of mitochondrial dysfunction and its role in metabolic inflammation.
Collapse
Affiliation(s)
- Qing Yuan
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Z. L. Zeng
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Shiqi Yang
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Anqi Li
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xuyu Zu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
- Diabetes Clinical Medical Research Center of Hunan Province, Department of Metabolism and Endocrinology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
22
|
Díaz-Pozo P, Canet F, Grirrane A, Lopez-Domenech S, Herance JR, Apostolova N, Luna-Marco C, Rovira-Llopis S, Marti M, Morillas C, Rocha M, Garcia H, Victor VM. Gold Nanoparticles Supported on Ceria Nanoparticles Modulate Leukocyte-Endothelium Cell Interactions and Inflammation in Type 2 Diabetes. Antioxidants (Basel) 2022; 11:2297. [PMID: 36421483 PMCID: PMC9686981 DOI: 10.3390/antiox11112297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 10/07/2024] Open
Abstract
Gold-ceria nanoparticles (Au/CeO2) are known to have antioxidant properties. However, whether these nanoparticles can provide benefits in type 2 diabetes mellitus (T2D) remains unknown. This work aimed to study the effects of Au/CeO2 nanoparticles at different rates of gold purity (10, 4.4, 1.79 and 0.82) on leukocyte-endothelium interactions and inflammation in T2D patients. Anthropometric and metabolic parameters, leukocyte-endothelium interactions, ROS production and NF-κB expression were assessed in 57 T2D patients and 51 healthy subjects. T2D patients displayed higher Body Mass Index (BMI) and characteristic alterations in carbohydrate and lipid metabolism. ROS production was increased in leukocytes of T2D patients and decreased by Au/CeO2 at 0.82% gold. Interestingly, Au/CeO2 0.82% modulated leukocyte-endothelium interactions (the first step in the atherosclerotic process) by increasing leukocyte rolling velocity and decreasing rolling flux and adhesion in T2D. A static adhesion assay also revealed diminished leukocyte-endothelium interactions by Au/CeO2 0.82% treatment. NF-κB (p65) levels increased in T2D patients and were reduced by Au/CeO2 treatment. Cell proliferation, viability, and apoptosis assays demonstrated no toxicity produced by Au/CeO2 nanoparticles. These results demonstrate that Au/CeO2 nanoparticles at 0.82% exert antioxidant and anti-inflammatory actions in the leukocyte-endothelium interaction of T2D patients, suggesting a protective role against the appearance of atherosclerosis and cardiovascular diseases when this condition exists.
Collapse
Affiliation(s)
- Pedro Díaz-Pozo
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
| | - Francisco Canet
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
| | - Abdessamad Grirrane
- Instituto Universitario de Tecnología Química CSIC-UPV, Universidad Politécnica de Valencia, Avenida de los Naranjos s/n, 46022 Valencia, Spain
| | - Sandra Lopez-Domenech
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
- Department of Pharmacology, University of Valencia, 46003 Valencia, Spain
| | - José Raul Herance
- Medical Molecular Imaging Research Group, Vall d’Hebron Research Institute, CIBBIM-Nanomedicine, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina CIBERBBN, 28029 Madrid, Spain
| | - Nadezda Apostolova
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
- Department of Pharmacology, University of Valencia, 46003 Valencia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 46010 Valencia, Spain
| | - Clara Luna-Marco
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
- Department of Physiology, INCLIVA (Biomedical Research Institute Valencia), University of Valencia, 46010 Valencia, Spain
| | - Susana Rovira-Llopis
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
- Department of Physiology, INCLIVA (Biomedical Research Institute Valencia), University of Valencia, 46010 Valencia, Spain
| | - Miguel Marti
- Department of Pharmacology, University of Valencia, 46003 Valencia, Spain
| | - Carlos Morillas
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
| | - Milagros Rocha
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 46010 Valencia, Spain
| | - Hermenegildo Garcia
- Instituto Universitario de Tecnología Química CSIC-UPV, Universidad Politécnica de Valencia, Avenida de los Naranjos s/n, 46022 Valencia, Spain
| | - Victor M. Victor
- Service of Endocrinology and Nutrition, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), University Hospital Doctor Peset, 46017 Valencia, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), 46010 Valencia, Spain
- Department of Physiology, INCLIVA (Biomedical Research Institute Valencia), University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
23
|
Mitochondria-Targeted Antioxidant Mitoquinone Maintains Mitochondrial Homeostasis through the Sirt3-Dependent Pathway to Mitigate Oxidative Damage Caused by Renal Ischemia/Reperfusion. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2213503. [PMID: 36193071 PMCID: PMC9526615 DOI: 10.1155/2022/2213503] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022]
Abstract
Mitochondrial dysfunction is a critical factor contributing to oxidative stress and apoptosis in ischemia-reperfusion (I/R) diseases. Mitoquinone (MitoQ) is a mitochondria-targeted antioxidant whose potent anti-I/R injury capacity has been demonstrated in organs such as the heart and the intestine. In the present study, we explored the role of MitoQ in maintaining mitochondrial homeostasis and attenuating oxidative damage in renal I/R injury. We discovered that the decreased renal function and pathological damage caused by renal I/R injury were significantly ameliorated by MitoQ. MitoQ markedly reversed mitochondrial damage after I/R injury and inhibited renal reactive oxygen species production. In vitro, hypoxia/reoxygenation resulted in increased mitochondrial fission and decreased mitochondrial fusion in human renal tubular epithelial cells (HK-2), which were partially prevented by MitoQ. MitoQ treatment inhibited oxidative stress and reduced apoptosis in HK-2 cells by restoring mitochondrial membrane potential, promoting ATP production, and facilitating mitochondrial fusion. Deeply, renal I/R injury led to a decreased expression of sirtuin-3 (Sirt3), which was recovered by MitoQ. Moreover, the inhibition of Sirt3 partially eliminated the protective effect of MitoQ on mitochondria and increased oxidative damage. Overall, our data demonstrate a mitochondrial protective effect of MitoQ, which raises the possibility of MitoQ as a novel therapy for renal I/R.
Collapse
|
24
|
Satta S, Hugo C, Sharma M, Rezek V, Kossyvakis A, Roy SS, Kitchen S, Kelesidis T. Mitoquinone mesylate attenuates brain inflammation in humanized mouse model of chronic HIV infection. AIDS 2022; 36:1609-1611. [PMID: 35979834 PMCID: PMC10924803 DOI: 10.1097/qad.0000000000003291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Affiliation(s)
- Sandro Satta
- Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, USA
| | - Cristelle Hugo
- Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, USA
| | - Madhav Sharma
- Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, USA
| | - Valerie Rezek
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Athanassios Kossyvakis
- Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, USA
| | - Shubhendu Sen Roy
- Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, USA
| | - Scott Kitchen
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Theodoros Kelesidis
- Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
25
|
Ji Y, Leng Y, Lei S, Qiu Z, Ming H, Zhang Y, Zhang A, Wu Y, Xia Z. The mitochondria-targeted antioxidant MitoQ ameliorates myocardial ischemia-reperfusion injury by enhancing PINK1/Parkin-mediated mitophagy in type 2 diabetic rats. Cell Stress Chaperones 2022; 27:353-367. [PMID: 35426609 PMCID: PMC9346044 DOI: 10.1007/s12192-022-01273-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/14/2022] [Accepted: 03/30/2022] [Indexed: 01/03/2023] Open
Abstract
Type 2 diabetic hearts are more vulnerable to myocardial ischemia reperfusion (MIR) injury, which involves decreased mitophagy status with unknown mechanisms. MitoQ, a mitochondria-targeted antioxidant, has been shown to have protection against ischemia reperfusion injury through upregulating mitophagy. The aim of this study was to investigate the effects of MitoQ on myocardium during MIR injury in type 2 diabetes (T2D). Herein, this study discovered that type 2 diabetic hearts with PINK1/Parkin downregulation suffered more MIR injury accompanied by reduced mitophagy. Treatment with MitoQ significantly decreased the levels of CK-MB, LDH, myocardial infarction, myocardial pathological damage, and cardiomyocytes apoptosis, while it improved cardiac function, mitophagy status, and PINK1/Parkin pathway in vivo study. Furthermore, MitoQ significantly reduced high glucose/high fat and hypoxia/reoxygenation induced injury in H9C2 cells as evidenced by reduced cardiomyocytes apoptosis and ROS production, and increased cell viability, the level of mitochondrial membrane potential, PINK1/Parkin expression. However, mitochondrial division inhibitor (mdivi-1), an inhibitor of mitophagy, reversed the improvement and protein expression levels of PINK1/Parkin pathway in vitro models. In conclusion, MIR induced more severe damage in T2D by reduction of mitophagy. MitoQ can confer cardioprotection following MIR in T2D by mitophagy up-regulation via PINK1/Parkin pathway.
Collapse
Affiliation(s)
- Yelong Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yan Leng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Hao Ming
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yi Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Aining Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China
| | - Yang Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China.
| | - Zhongyaun Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
26
|
Adisa RA, Sulaimon LA, Okeke EG, Ariyo OC, Abdulkareem FB. Mitoquinol mesylate (MITOQ) attenuates diethyl nitrosamine-induced hepatocellular carcinoma through modulation of mitochondrial antioxidant defense systems. Toxicol Res 2022; 38:275-291. [PMID: 35874502 PMCID: PMC9247134 DOI: 10.1007/s43188-021-00105-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/05/2021] [Accepted: 09/07/2021] [Indexed: 12/24/2022] Open
Abstract
Diethyl nitrosamine (DEN) induced cirrhosis-hepatocellular carcinoma (HCC) model associates cancer progression with oxidative stress and mitochondrial dysfunction. This study investigated the effects of mitoquinol mesylate (MitoQ), a mitochondrial-targeted antioxidant on DEN-induced oxidative damage in HCC Wistar rats. Fifty male Wistar rats were randomly divided into five groups. Healthy control, DEN, and MitoQ groups were orally administered exactly 10 mg/kg of distilled water, DEN, and MitoQ, respectively for 16 weeks. Animals in the MitoQ + DEN group were pre-treated with MitoQ for a week followed by co-administration of 10 mg/kg each of MitoQ and DEN. DEN + MitoQ group received DEN for 8 weeks, then co-administration of 10 mg/kg each of DEN and MitoQ till the end of 16th week. Survival index, tumour incidence, hematological profile, liver function indices, lipid profile, mitochondrial membrane composition, mitochondrial respiratory enzymes, and antioxidant defense status in both mitochondrial and post-mitochondrial fractions plus expression of antioxidant genes were assessed. In MitoQ + DEN and DEN + MitoQ groups, 80% survival occurred while tumour incidence decreased by 60% and 40% respectively, compared to the DEN-only treated group. Similarly, MitoQ-administered groups showed a significant (p < 0.05) decrease in the activities of liver function enzymes while hemoglobin concentration, red blood cell count, and packed cell volume were significantly elevated compared to the DEN-only treated group. Administration of MitoQ to the DEN-intoxicated groups successfully enhanced the activities of mitochondrial F1F0-ATPase and succinate dehydrogenase; and up-regulated the expression and activities of SOD2, CAT, and GPx1. Macroscopic and microscopic features indicated a reversal of DEN-induced hepatocellular degeneration in the MitoQ + DEN and DEN + MitoQ groups. These data revealed that MitoQ intervention attenuated DEN-induced oxidative stress through modulation of mitochondrial antioxidant defense systems and alleviated the burden of HCC as a chemotherapeutic agent.
Collapse
Affiliation(s)
- Rahmat Adetutu Adisa
- Laboratories for Bio-membranes and Cancer Research, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of University of Lagos, Idi-araba, Lagos, P.M.B. 12003 Nigeria
| | - Lateef Adegboyega Sulaimon
- Laboratories for Bio-membranes and Cancer Research, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of University of Lagos, Idi-araba, Lagos, P.M.B. 12003 Nigeria
| | - Ebele Geraldine Okeke
- Laboratories for Bio-membranes and Cancer Research, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of University of Lagos, Idi-araba, Lagos, P.M.B. 12003 Nigeria
| | - Olubukola Christianah Ariyo
- Laboratories for Bio-membranes and Cancer Research, Department of Biochemistry, Faculty of Basic Medical Sciences, College of Medicine of University of Lagos, Idi-araba, Lagos, P.M.B. 12003 Nigeria
| | - Fatimah B. Abdulkareem
- Department of Anatomic and Molecular Pathology, Faculty of Basic Medical Sciences,, College of Medicine of University of Lagos, Idi-araba, P.M.B. 12003 Lagos, Nigeria
| |
Collapse
|
27
|
Liu C, Sun Z, Wang M, Yang Z, Zhang W, Ren Y, Han X, Zhang B, Yao M, Nie S. Mitoquinone mitigates paraquat-induced A549 lung epithelial cell injury by promoting MFN1/MFN2-mediated mitochondrial fusion. J Biochem Mol Toxicol 2022; 36:e23127. [PMID: 35686354 DOI: 10.1002/jbt.23127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 04/16/2022] [Accepted: 05/29/2022] [Indexed: 12/21/2022]
Abstract
Paraquat (PQ) poisoning often leads to severe lung injuries, in which the mitochondria damage plays a critical role. Mitoquinone (MitoQ), a newly designed mitochondria-targeted antioxidant, has been proved for its benefit in mitochondria protection. However, the role of MitoQ in PQ-induced lung injury remains unclear. Thus, this study was performed to investigate the effect of MitoQ on PQ-induced lung injury and its underlying mechanisms. Our work showed that PQ caused the inhibition of A549 lung epithelial cell viability in a dose-dependent manner, while MitoQ remarkably mitigated the PQ-induced cell viability suppression. Besides this, PQ-mediated apoptosis of A549 cells was significantly attenuated by MitoQ, as indicated by the TUNEL assay and mitochondria membrane potential assay. Moreover, the intracellular reactive oxygen species (ROS) production was also dramatically suppressed when cotreated MitoQ with PQ. This could be ascribed to enhanced mitochondrial fusion mediated by Mitofusin 1 (MFN1)/Mitofusin 2 (MFN2), because MitoQ preserved mitochondrial network integrity, as reflected by MitoTracker staining, and MitoQ also increased the expression of MFN1/MFN2 in A549 cells after PQ treatment. Our data suggested MitoQ mitigated PQ-induced lung epithelial cell injury by promoting MFN1/MFN2-mediated mitochondrial fusion, and MitoQ might be a potential candidate drug for the treatment of PQ-induced lung injury.
Collapse
Affiliation(s)
- Chao Liu
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Zhaorui Sun
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Mengmeng Wang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Zhizhou Yang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Wei Zhang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Yi Ren
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Xiaoqin Han
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Bo Zhang
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Mengya Yao
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| | - Shinan Nie
- Department of Emergency Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, PR China
| |
Collapse
|
28
|
Xu Z, Liu Y, Ma R, Chen J, Qiu J, Du S, Li C, Wu Z, Yang X, Chen Z, Chen T. Thermosensitive Hydrogel Incorporating Prussian Blue Nanoparticles Promotes Diabetic Wound Healing via ROS Scavenging and Mitochondrial Function Restoration. ACS APPLIED MATERIALS & INTERFACES 2022; 14:14059-14071. [PMID: 35298140 DOI: 10.1021/acsami.1c24569] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Diabetic foot ulcer is a serious complication in diabetes patients, imposing a serious physical and economic burden to patients and to the healthcare system as a whole. Oxidative stress is thought to be a key driver of the pathogenesis of such ulcers. However, no antioxidant drugs have received clinical approval to date, underscoring the need for the further development of such medications. Hydrogels can be applied directly to the wound site, wherein they function to prevent infection and maintain local moisture concentrations, in addition to serving as a reservoir for the delivery of a range of therapeutic compounds with the potential to expedite wound healing in a synergistic manner. Herein, we synthesized Prussian blue nanoparticles (PBNPs) capable of efficiently scavenging reactive oxygen species (ROS) owing to their ability to mimic the activity of catalase (CAT), peroxidase (POD), and superoxide dismutase (SOD). In the context of in vitro oxidative stress, these PBNPs were able to protect against cytotoxicity, protect mitochondria from oxidative stress-related damage, and restore nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase-1 (HO-1) pathway activity. To expand on these results in an in vivo context, we prepared a thermosensitive poly (d,l-lactide)-poly(ethylene glycol)-poly(d,l-lactide) (PDLLA-PEG-PDLLA) hydrogel (PLEL)-based wound dressing in which PBNPs had been homogenously incorporated, and we then used this dressing as a platform for controlled PBNP release. The resultant PBNPs@PLEL wound dressing was able to improve diabetic wound healing, decrease ROS production, promote angiogenesis, and reduce pro-inflammatory interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) levels within diabetic wounds. Overall, our results suggest that this PBNPs@PLEL platform holds great promise as a treatment for diabetic foot ulcers.
Collapse
Affiliation(s)
- Zhao Xu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yujing Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Rui Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinmei Qiu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shuang Du
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Chengcheng Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zihan Wu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
29
|
The Effect of Antioxidant Added to Preservation Solution on the Protection of Kidneys before Transplantation. Int J Mol Sci 2022; 23:ijms23063141. [PMID: 35328560 PMCID: PMC8954097 DOI: 10.3390/ijms23063141] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/07/2022] [Accepted: 03/13/2022] [Indexed: 02/04/2023] Open
Abstract
Ischemia–reperfusion injury is a key clinical problem of transplantology. Current achievements in optimizing organ rinse solutions and storage techniques have significantly influenced the degree of graft damage and its survival after transplantation. In recent years, intensive research has been carried out to maintain the viability of tissues and organs outside the integral environment of the body. Innovative solutions for improving the biochemical functions of the stored organ have been developed. The article discusses directions for modifying preservation solutions with antioxidants. Clinical and experimental studies aimed at optimizing these fluids, as well as perfusion and organ preservation techniques, are presented.
Collapse
|
30
|
Trinity JD, Drummond MJ, Fermoyle CC, McKenzie AI, Supiano MA, Richardson RS. Cardiovasomobility: an integrative understanding of how disuse impacts cardiovascular and skeletal muscle health. J Appl Physiol (1985) 2022; 132:835-861. [PMID: 35112929 PMCID: PMC8934676 DOI: 10.1152/japplphysiol.00607.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cardiovasomobility is a novel concept that encompasses the integration of cardiovascular and skeletal muscle function in health and disease with critical modification by physical activity, or lack thereof. Compelling evidence indicates that physical activity improves health while a sedentary, or inactive, lifestyle accelerates cardiovascular and skeletal muscle dysfunction and hastens disease progression. Identifying causative factors for vascular and skeletal muscle dysfunction, especially in humans, has proven difficult due to the limitations associated with cross-sectional investigations. Therefore, experimental models of physical inactivity and disuse, which mimic hospitalization, injury, and illness, provide important insight into the mechanisms and consequences of vascular and skeletal muscle dysfunction. This review provides an overview of the experimental models of disuse and inactivity and focuses on the integrated responses of the vasculature and skeletal muscle in response to disuse/inactivity. The time course and magnitude of dysfunction evoked by various models of disuse/inactivity are discussed in detail, and evidence in support of the critical roles of mitochondrial function and oxidative stress are presented. Lastly, strategies aimed at preserving vascular and skeletal muscle dysfunction during disuse/inactivity are reviewed. Within the context of cardiovasomobility, experimental manipulation of physical activity provides valuable insight into the mechanisms responsible for vascular and skeletal muscle dysfunction that limit mobility, degrade quality of life, and hasten the onset of disease.
Collapse
Affiliation(s)
- Joel D Trinity
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| | - Micah J Drummond
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah.,Department of Physical Therapy, University of Utah, Salt Lake City, Utah
| | - Caitlin C Fermoyle
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Alec I McKenzie
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Mark A Supiano
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah
| | - Russell S Richardson
- Salt Lake City Veteran Affairs Medical Center Geriatric Research, Education, and Clinical Center, Salt Lake City, Utah.,Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, Utah
| |
Collapse
|
31
|
Zhang W, Zhang D, Cheng Y, Liang X, Wang J. Runx1 regulates Tff1 expression to expedite viability of retinal microvascular endothelial cells in mice with diabetic retinopathy. Exp Eye Res 2022; 217:108969. [PMID: 35114215 DOI: 10.1016/j.exer.2022.108969] [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: 09/14/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/04/2022]
Abstract
Diabetic retinopathy (DR) represents a major complication of diabetes, and molecular mechanisms related to vascular dysfunction, particularly endothelial dysfunction, in DR remains unclear. In the present work, we generated a DR animal model using mice and a cell model in mouse retinal microvascular endothelial cells (mRMECs) to examine the role of Trefoil factor family 1 (Tff1) in DR. Tff1 was poorly expressed in DR mice and high glucose (HG)-treated mRMECs. Overexpression of Tff1 significantly attenuated streptozotocin-induced retinal proliferation and angiogenesis in DR mice and reduced the secretion of inflammatory factors. In HG-treated mRMECs, overexpression of Tff1 remarkably reduced the proliferation and angiogenesis of mRMECs. In further experiments, we found that Tff1 was transcriptionally repressed by Runt-related transcription factor 1 (Runx1) directly, and Tff1 expression was indirectly modulated by Runx1 via the core-binding factor subunit beta (CBF-β)/nuclear factor, erythroid 2/microRNA-423-5p axis and the CBF-β/estrogen receptor 1 (ESR1) axis. Moreover, Tff1 could inhibit the activation of NF-κB signaling pathway, which in turn attenuated retinal endothelial cell proliferation and angiogenesis. It was thus proposed that Runx1/Tff1/NF-κB axis may be a potential target for the treatment strategy of DR, and further studies are needed.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Ophthalmology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030012, Shanxi, PR China; Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, PR China.
| | - Dingguo Zhang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, PR China
| | - Yan Cheng
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Xing Liang
- Department of Ophthalmology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030012, Shanxi, PR China
| | - Jingjing Wang
- Department of Ophthalmology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030012, Shanxi, PR China
| |
Collapse
|
32
|
Salahi E, Amidi F, Zahiri Z, Aghahosseini M, Mashayekhi F, Amani Abkenari S, Hosseinishenatal S, Sobhani A. The effect of mitochondria-targeted antioxidant MitoQ10 on redox signaling pathway components in PCOS mouse model. Arch Gynecol Obstet 2021; 305:985-994. [PMID: 34633506 DOI: 10.1007/s00404-021-06230-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/01/2021] [Indexed: 12/01/2022]
Abstract
PURPOSE Considerable evidence suggests that mitochondrial dysfunction and oxidative stress contribute to the pathogenesis of Polycystic ovary syndrome (PCOS). We aimed to evaluate the effectiveness of mitochondria-targeted antioxidant, MitoQ10, on the redox signaling pathway's component in PCOS. METHOD We assessed TXNIP, TRX, and ASK1 expression in granulosa cells (GCs) of the DHEA-induced PCOS mouse model. Female BALB/c mice in five groups of Control, DHEA, and DHEA + MitoQ10 in three doses of 250, 500, and 750 μmol/L MitoQ10 were treated for 21 days. RESULTS Histological investigation showed a probable improvement in folliculogenesis; besides, ASK1 and TXNIP expression were significantly increased in GCs of the PCOS mouse F4Fmodel as compared to the control groups and decreased steadily in groups treated by MitoQ10. However, TRX expression showed a drop that was restored by MitoQ10 meaningfully (P ≤ 0.05). CONCLUSION The work presented herein suggests mitochondria-targeted antioxidant, MitoQ10, have modulating effects on folliculogenesis in the ovary and also on the redox signaling pathway in GCs of PCOS mouse model which may have potential to attenuate oxidative stress and its relative damages.
Collapse
Affiliation(s)
- Elnaz Salahi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina ST, Tehran, Iran
| | - Fardin Amidi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina ST, Tehran, Iran
| | - Ziba Zahiri
- Reproductive Health Research Center, Department of Obstetrics and Gynecology, Alzahra Hospital, School of Medicine, Guilan University of Medical Science, Rasht, Iran
| | - Marziye Aghahosseini
- Department of Obstetrics and Gynecology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Mashayekhi
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Showra Amani Abkenari
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina ST, Tehran, Iran
| | - Shirzad Hosseinishenatal
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina ST, Tehran, Iran
| | - Aligholi Sobhani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Poursina ST, Tehran, Iran.
| |
Collapse
|
33
|
Effect of Endogenic and Exogenic Oxidative Stress Triggers on Adverse Pregnancy Outcomes: Preeclampsia, Fetal Growth Restriction, Gestational Diabetes Mellitus and Preterm Birth. Int J Mol Sci 2021; 22:ijms221810122. [PMID: 34576285 PMCID: PMC8468091 DOI: 10.3390/ijms221810122] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/10/2021] [Accepted: 09/16/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is caused by an imbalance between the production of reactive oxygen species (ROS) in cells and tissues and the ability of a biological system to detoxify them. During a normal pregnancy, oxidative stress increases the normal systemic inflammatory response and is usually well-controlled by the balanced body mechanism of the detoxification of anti-oxidative products. However, pregnancy is also a condition in which this adaptation and balance can be easily disrupted. Excessive ROS is detrimental and associated with many pregnancy complications, such as preeclampsia (PE), fetal growth restriction (FGR), gestational diabetes mellitus (GDM), and preterm birth (PTB), by damaging placentation. The placenta is a tissue rich in mitochondria that produces the majority of ROS, so it is important to maintain normal placental function and properly develop its vascular network to ensure a safe and healthy pregnancy. Antioxidants may ameliorate these diseases, and related research is progressing. This review aimed to determine the association between oxidative stress and adverse pregnancy outcomes, especially PE, FGR, GDM, and PTB, and explore how to overcome this oxidative stress in these unfavorable conditions.
Collapse
|
34
|
Zhong L, Deng J, Gu C, Shen L, Ren Z, Ma X, Yan Q, Deng J, Zuo Z, Wang Y, Cao S, Yu S. Protective effect of MitoQ on oxidative stress-mediated senescence of canine bone marrow mesenchymal stem cells via activation of the Nrf2/ARE pathway. In Vitro Cell Dev Biol Anim 2021; 57:685-694. [PMID: 34518994 DOI: 10.1007/s11626-021-00605-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/26/2021] [Indexed: 10/20/2022]
Abstract
The destruction of biological activity such as senescence and apoptosis caused by oxidative stress could play a pivotal role in the poor therapeutic efficiency of bone marrow mesenchymal stem cells (BMSCs) transplantation. Mitoquinone (MitoQ) has a highly effective mitochondrial antioxidant effect, and has been widely used in many oxidative damage models. This study aimed to investigate the protective effect of MitoQ on the oxidative stress-mediated senescence of canine BMSCs and the underlying mechanism. The senescence of BMSCs was determined by senescence-associated β-galactosidase staining and quantitative real-time PCR. The expression of p-Nrf2 protein was detected by Western blotting. The results demonstrated that, as BMSCs were expanded in vitro, the senescent phenotype appeared. And the senescence of BMSCs may be caused by oxidative stress, manifested by increasing the level of ROS and decreasing the activity of antioxidant enzymes. Treatment of MitoQ down-regulated the mRNA levels of senescence-related and apoptosis-related genes, but up-regulated the mRNA levels of proliferation-related genes. Meanwhile, ROS generation and senescent activity were reduced in MitoQ-treated BMSCs. Further mechanism studies showed that MitoQ obviously promoted Nrf2 phosphorylation, and also facilitated the translocation of Nrf2 into the nucleus. Moreover, treatment of MitoQ increased the mRNA levels of downstream antioxidant genes and enhanced the activities of superoxide dismutase, catalase, and glutathione peroxidase. Thus, our study revealed that MitoQ, via the Nrf2/ARE signaling pathway, exerts an antioxidant effect as well as potentially delays OS-mediated senescence during BMSCs that were expanded in vitro, which may serve as a novel strategy to optimize the clinical application of BMSCs.
Collapse
Affiliation(s)
- Lijun Zhong
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Jiaqiang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Congwei Gu
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China.,Laboratory Animal Centre, Southwest Medical University, Luzhou, Sichuan, China
| | - Liuhong Shen
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Zhihua Ren
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Xiaoping Ma
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Qigui Yan
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Junliang Deng
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Zhicai Zuo
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Ya Wang
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Suizhong Cao
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China
| | - Shumin Yu
- College of Veterinary Medicine, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
35
|
Shyam R, Ogando DG, Choi M, Liton PB, Bonanno JA. Mitochondrial ROS Induced Lysosomal Dysfunction and Autophagy Impairment in an Animal Model of Congenital Hereditary Endothelial Dystrophy. Invest Ophthalmol Vis Sci 2021; 62:15. [PMID: 34533563 PMCID: PMC8458782 DOI: 10.1167/iovs.62.12.15] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/18/2021] [Indexed: 01/04/2023] Open
Abstract
Purpose The Slc4a11 knock out (KO) mouse model recapitulates the human disease phenotype associated with congenital hereditary endothelial dystrophy (CHED). Increased mitochondrial reactive oxygen species (ROS) in the Slc4a11 KO mouse model is a major cause of edema and endothelial cell loss. Here, we asked if autophagy was activated by ROS in the KO mice. Methods Immortalized cell lines and mouse corneal endothelia were used to measure autophagy and lysosome associated protein expressions using Protein Simple Wes immunoassay. Autophagy and lysosome functions were examined in wild type (WT) and KO cells as well as animals treated with the mitochondrial ROS quencher MitoQ. Results Even though autophagy activation was evident, autophagy flux was aberrant in Slc4a11 KO cells and corneal endothelium. Expression of lysosomal proteins and lysosomal mass were decreased along with reduced nuclear translocation of lysosomal master regulator, transcription factor EB (TFEB). MitoQ reversed aberrant lysosomal functions and TFEB nuclear localization in KO cells. MitoQ injections in KO animals reduced corneal edema and decreased the rate of endothelial cell loss. Conclusions Mitochondrial ROS disrupts TFEB signaling causing lysosomal dysfunction with impairment of autophagy in Slc4a11 KO corneal endothelium. Our study is the first to identify the presence as well as cause of lysosomal dysfunction in an animal model of CHED, and to identify a potential therapeutic approach.
Collapse
MESH Headings
- Animals
- Anion Transport Proteins/genetics
- Autophagy/physiology
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism
- Blotting, Western
- Cathepsin L/metabolism
- Cells, Cultured
- Corneal Dystrophies, Hereditary/genetics
- Corneal Dystrophies, Hereditary/metabolism
- Corneal Dystrophies, Hereditary/pathology
- Disease Models, Animal
- Endothelium, Corneal/drug effects
- Endothelium, Corneal/metabolism
- Gene Expression Regulation
- Immunohistochemistry
- Injections, Intraperitoneal
- Lysosomes/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Fluorescence
- Mitochondria/metabolism
- Organophosphorus Compounds/pharmacology
- Reactive Oxygen Species/metabolism
- Real-Time Polymerase Chain Reaction
- Symporters/genetics
- Transfection
- Ubiquinone/analogs & derivatives
- Ubiquinone/pharmacology
Collapse
Affiliation(s)
- Rajalekshmy Shyam
- Vision Science Program, School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Diego G. Ogando
- Vision Science Program, School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Moonjung Choi
- Vision Science Program, School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Paloma B. Liton
- Department of Ophthalmology, Duke University, Durham, North Carolina, United States
| | - Joseph A. Bonanno
- Vision Science Program, School of Optometry, Indiana University, Bloomington, Indiana, United States
| |
Collapse
|
36
|
Lu PH, Yu MC, Wei MJ, Kuo KL. The Therapeutic Strategies for Uremic Toxins Control in Chronic Kidney Disease. Toxins (Basel) 2021; 13:573. [PMID: 34437444 PMCID: PMC8402511 DOI: 10.3390/toxins13080573] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 12/27/2022] Open
Abstract
Uremic toxins (UTs) are mainly produced by protein metabolized by the intestinal microbiota and converted in the liver or by mitochondria or other enzymes. The accumulation of UTs can damage the intestinal barrier integrity and cause vascular damage and progressive kidney damage. Together, these factors lead to metabolic imbalances, which in turn increase oxidative stress and inflammation and then produce uremia that affects many organs and causes diseases including renal fibrosis, vascular disease, and renal osteodystrophy. This article is based on the theory of the intestinal-renal axis, from bench to bedside, and it discusses nonextracorporeal therapies for UTs, which are classified into three categories: medication, diet and supplement therapy, and complementary and alternative medicine (CAM) and other therapies. The effects of medications such as AST-120 and meclofenamate are described. Diet and supplement therapies include plant-based diet, very low-protein diet, probiotics, prebiotics, synbiotics, and nutraceuticals. The research status of Chinese herbal medicine is discussed for CAM and other therapies. This review can provide some treatment recommendations for the reduction of UTs in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Ping-Hsun Lu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan; (P.-H.L.); (M.-C.Y.); (M.-J.W.)
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97048, Taiwan
| | - Min-Chien Yu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan; (P.-H.L.); (M.-C.Y.); (M.-J.W.)
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien 97048, Taiwan
| | - Meng-Jiun Wei
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan; (P.-H.L.); (M.-C.Y.); (M.-J.W.)
| | - Ko-Lin Kuo
- Division of Nephrology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 23142, Taiwan
- School of Medicine, Buddhist Tzu Chi University, Hualien 97048, Taiwan
| |
Collapse
|
37
|
Donia T, Khamis A. Management of oxidative stress and inflammation in cardiovascular diseases: mechanisms and challenges. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:34121-34153. [PMID: 33963999 DOI: 10.1007/s11356-021-14109-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/21/2021] [Indexed: 06/12/2023]
Abstract
Cardiovascular diseases (CVDs) have diverse physiopathological mechanisms with interconnected oxidative stress and inflammation as one of the common etiologies which result in the onset and development of atherosclerotic plaques. In this review, we illustrate this strong crosstalk between oxidative stress, inflammation, and CVD. Also, mitochondrial functions underlying this crosstalk, and various approaches for the prevention of redox/inflammatory biological impacts will be illustrated. In part, we focus on the laboratory biomarkers and physiological tests for the evaluation of oxidative stress status and inflammatory processes. The impact of a healthy lifestyle on CVD onset and development is displayed as well. Furthermore, the differences in oxidative stress and inflammation are related to genetic susceptibility to cardiovascular diseases and the variability in the assessment of CVDs risk between individuals; Omics technologies for measuring oxidative stress and inflammation will be explored. Finally, we display the oxidative stress-related microRNA and the functions of the redox basis of epigenetic modifications.
Collapse
Affiliation(s)
- Thoria Donia
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Abeer Khamis
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt.
| |
Collapse
|
38
|
Hamed M, Logan A, Gruszczyk AV, Beach TE, James AM, Dare AJ, Barlow A, Martin J, Georgakopoulos N, Gane AM, Crick K, Fouto D, Fear C, Thiru S, Dolezalova N, Ferdinand JR, Clatworthy MR, Hosgood SA, Nicholson ML, Murphy MP, Saeb-Parsy K. Mitochondria-targeted antioxidant MitoQ ameliorates ischaemia-reperfusion injury in kidney transplantation models. Br J Surg 2021; 108:1072-1081. [PMID: 33963377 DOI: 10.1093/bjs/znab108] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 02/28/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND Ischaemia-reperfusion (IR) injury makes a major contribution to graft damage during kidney transplantation. Oxidative damage to mitochondria is an early event in IR injury. Therefore, the uptake, safety, and efficacy of the mitochondria-targeted antioxidant MitoQ were investigated in models of transplant IR injury. METHODS MitoQ uptake by warm and cooled pairs of pig and declined human kidneys was measured when preserved in cold static storage or by hypothermic machine perfusion. Pairs of pigs' kidneys were exposed to defined periods of warm and cold ischaemia, flushed and stored at 4°C with or without MitoQ (50 nmol/l to 250 µmol/l), followed by reperfusion with oxygenated autologous blood in an ex vivo normothermic perfusion (EVNP). Pairs of declined human kidneys were flushed and stored with or without MitoQ (5-100 µmol/l) at 4°C for 6 h and underwent EVNP with ABO group-matched blood. RESULTS Stable and concentration-dependent uptake of MitoQ was demonstrated for up to 24 h in pig and human kidneys. Total blood flow and urine output were significantly greater in pig kidneys treated with 50 µmol/l MitoQ compared with controls (P = 0.006 and P = 0.007 respectively). In proof-of-concept experiments, blood flow after 1 h of EVNP was significantly greater in human kidneys treated with 50 µmol/l MitoQ than in controls (P ≤ 0.001). Total urine output was numerically higher in the 50-µmol/l MitoQ group compared with the control, but the difference did not reach statistical significance (P = 0.054). CONCLUSION Mitochondria-targeted antioxidant MitoQ can be administered to ischaemic kidneys simply and effectively during cold storage, and may improve outcomes after transplantation.
Collapse
Affiliation(s)
- M Hamed
- Department of Surgery, University of Cambridge, Cambridge, UK.,MRC Mitochondrial Biology Unit, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - A Logan
- MRC Mitochondrial Biology Unit, Cambridge, UK
| | - A V Gruszczyk
- Department of Surgery, University of Cambridge, Cambridge, UK.,MRC Mitochondrial Biology Unit, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - T E Beach
- Department of Surgery, University of Cambridge, Cambridge, UK.,MRC Mitochondrial Biology Unit, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - A M James
- MRC Mitochondrial Biology Unit, Cambridge, UK
| | - A J Dare
- Department of Surgery, University of Cambridge, Cambridge, UK.,MRC Mitochondrial Biology Unit, Cambridge, UK
| | - A Barlow
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - J Martin
- Department of Surgery, University of Cambridge, Cambridge, UK.,MRC Mitochondrial Biology Unit, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - N Georgakopoulos
- Department of Surgery, University of Cambridge, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - A M Gane
- Department of Surgery, University of Cambridge, Cambridge, UK.,MRC Mitochondrial Biology Unit, Cambridge, UK
| | - K Crick
- Department of Surgery, University of Cambridge, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - D Fouto
- Department of Surgery, University of Cambridge, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - C Fear
- Department of Surgery, University of Cambridge, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - S Thiru
- Department of Pathology, Cambridge University Hospitals NHS Trust, Addenbrooke's Hospital, Cambridge, UK
| | - N Dolezalova
- Department of Surgery, University of Cambridge, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - J R Ferdinand
- Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - M R Clatworthy
- Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - S A Hosgood
- Department of Surgery, University of Cambridge, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - M L Nicholson
- Department of Surgery, University of Cambridge, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| | - M P Murphy
- MRC Mitochondrial Biology Unit, Cambridge, UK.,Department of Medicine, University of Cambridge, Cambridge, UK
| | - K Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK.,Cambridge National Institute for Health Research (NIHR) Biomedical Research Centre and NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, Cambridge, UK
| |
Collapse
|
39
|
Anastasiou IA, Eleftheriadou I, Tentolouris A, Koliaki C, Kosta OA, Tentolouris N. CDATA[The Effect of Oxidative Stress and Antioxidant Therapies on Pancreatic β-cell Dysfunction: Results from in Vitro and in Vivo Studies. Curr Med Chem 2021; 28:1328-1346. [PMID: 32452321 DOI: 10.2174/0929867327666200526135642] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/07/2020] [Accepted: 04/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Oxidative stress is a hallmark of many diseases. A growing body of evidence suggests that hyperglycemia-induced oxidative stress plays an important role in pancreatic β-cells dysfunction and apoptosis, as well as in the development and progression of diabetic complications. Considering the vulnerability of pancreatic β-cells to oxidative damage, the induction of endogenous antioxidant enzymes or exogenous antioxidant administration has been proposed to protect pancreatic β-cells from damage. OBJECTIVES The present review aims to provide evidence of the effect of oxidative stress and antioxidant therapies on pancreatic β-cell function, based on in vitro and in vivo studies. METHODS The MEDLINE and EMBASE databases were searched to retrieve available data. RESULTS Due to poor endogenous antioxidant mechanisms, pancreatic β-cells are extremely sensitive to Reactive Oxygen Species (ROS). Many natural extracts have been tested in vitro in pancreatic β-cell lines in terms of their antioxidant and diabetes mellitus ameliorating effects, and the majority of them have shown a dose-dependent protective role. On the other hand, there is relatively limited evidence regarding the in vitro antioxidant effects of antidiabetic drugs on pancreatic β -cells. Concerning in vivo studies, several natural extracts have shown beneficial effects in the setting of diabetes by decreasing blood glucose and lipid levels, increasing insulin sensitivity, and by up-regulating intrinsic antioxidant enzyme activity. However, there is limited evidence obtained from in vivo studies regarding antidiabetic drugs. CONCLUSION Antioxidants hold promise for developing strategies aimed at the prevention or treatment of diabetes mellitus associated with pancreatic β-cells dysfunction, as supported by in vitro and in vivo studies. However, more in vitro studies are required for drugs.
Collapse
Affiliation(s)
- Ioanna A Anastasiou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 17 AgiouThoma St., 11527 Athens, Greece
| | - Ioanna Eleftheriadou
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 17 AgiouThoma St., 11527 Athens, Greece
| | - Anastasios Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 17 AgiouThoma St., 11527 Athens, Greece
| | - Chrysi Koliaki
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 17 AgiouThoma St., 11527 Athens, Greece
| | - Ourania A Kosta
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 17 AgiouThoma St., 11527 Athens, Greece
| | - Nikolaos Tentolouris
- Diabetes Center, First Department of Propaedeutic Internal Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, 17 AgiouThoma St., 11527 Athens, Greece
| |
Collapse
|
40
|
Cen M, Ouyang W, Zhang W, Yang L, Lin X, Dai M, Hu H, Tang H, Liu H, Xia J, Xu F. MitoQ protects against hyperpermeability of endothelium barrier in acute lung injury via a Nrf2-dependent mechanism. Redox Biol 2021; 41:101936. [PMID: 33752110 PMCID: PMC8005834 DOI: 10.1016/j.redox.2021.101936] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 01/28/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
Recently, numerous evidence has revealed that excessive reactive oxygen species (ROS) production and mitochondrial disruption during acute lung injury (ALI) and its most severe form, acute respiratory distress syndrome (ARDS) will aggravate the inflammatory process. To identify whether antioxidation can be one of the treatment strategies during this progress, we chose mitoQ, a mitochondria-targeted antioxidant that was proved to be effective in reducing ROS generated in mitochondria, as a ROS scavenger to investigate the role of antioxidation in ALI. We demonstrated that overoxidation occurred during the process of ALI, which could be reduced by mitoQ. In the meantime, apoptosis of endothelial cells of ALI mice, accompanied by hyperpermeability of pulmonary vascular and impaired pulmonary function, was partially reversed following an intraperitoneal injection of mitoQ. Moreover, in in vitro study, lipopolysaccharides (LPS) induced excessive ROS production, mitochondrial dysfunction and apoptosis in human pulmonary microvascular endothelial cells (HPMECs), which were rectified by mitoQ. To explore underlying mechanisms, we proceeded RNA-sequencing and found significantly upregulated expression of musculoaponeurotic fibrosarcoma F (MafF) in mitoQ treated group. Additionally, mitoQ inhibited the degradation and increased nuclear translocation of NF-E2-related factor 2 (Nrf2) and upregulated its downstream antioxidant response elements (AREs), such as heme oxygenase (HO)-1 and NAD(P)H:quinone oxidoreductase (NQO)-1. This effect was abolished by transfecting HPMECs with Nrf2 or Maff siRNA. In Nrf2 deficient mice, the protective effects of mitoQ on LPS model of ALI were largely vanished. Taken together, these results provide insights into how antioxidation exerts beneficial effects on ALI via maintaining mitochondrial hemostasis, inhibiting endothelial cells apoptosis, attenuating the endothelial disruption and regulating lung inflammation via Nrf2-MafF/ARE pathway.
Collapse
Affiliation(s)
- Mengyuan Cen
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Wei Ouyang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Wanying Zhang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Liping Yang
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xiuhui Lin
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Min Dai
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Huiqun Hu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Huifang Tang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Hongyun Liu
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Jingyan Xia
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
41
|
Bai J, Liu C, Zhu P, Li Y. Novel Insights Into Molecular Mechanism of Mitochondria in Diabetic Cardiomyopathy. Front Physiol 2021; 11:609157. [PMID: 33536936 PMCID: PMC7849834 DOI: 10.3389/fphys.2020.609157] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/21/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular complication is one of the significant causes of death in diabetic mellitus (DM) in which diabetic cardiomyopathy, independent of hypertension, cardiac valvular disease, and coronary atherosclerosis, occupies an important position. Although the detailed pathogenesis of diabetic cardiomyopathy remains unclear currently, mitochondrial morphological abnormality and dysfunction were observed in diabetic cardiomyopathy animal models according to much research, suggesting that mitochondrial structural and functional impairment played an integral role in the formation of diabetic cardiomyopathy. Thus, we have summarized the effect of mitochondria on the process of diabetic cardiomyopathy, including abnormal mitochondrial morphology, mitochondrial energy metabolism disorder, enhanced mitochondrial oxidative stress, mitochondrial unbalanced calcium homeostasis, and mitochondrial autophagy. Based on the above mechanisms and the related evidence, more therapeutic strategies targeting mitochondria in diabetic cardiomyopathy have been and will be proposed to delay the progression of the disease.
Collapse
Affiliation(s)
- Jing Bai
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Chuanbin Liu
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Pingjun Zhu
- Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yang Li
- Medical School of Chinese PLA, Beijing, China.,Department of Cardiology, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
42
|
Yang D, Xu D, Wang T, Yuan Z, Liu L, Shen Y, Wen F. Mitoquinone ameliorates cigarette smoke-induced airway inflammation and mucus hypersecretion in mice. Int Immunopharmacol 2021; 90:107149. [PMID: 33191175 DOI: 10.1016/j.intimp.2020.107149] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/19/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cigarette smoking, which induces airway inflammation and mucus hypersecretion, is a major risk factor for the development of cigarette smoke (CS)-induced airway disorders. In this study, we investigated the effects and mechanisms of mitoquinone (MitoQ), a mitochondria-targeted antioxidant, on CS-induced airway inflammation and mucus hypersecretion in mice. METHODS C57BL/6J mice were exposed to CS for 75 min twice daily, 5 days per week for 4 weeks. MitoQ (2.5, 5 mg/kg/day) was administered intraperitoneally 1 h before CS exposure. Bronchoalveolar lavage fluid (BALF) was obtained for cell counting and determination of pro-inflammatory cytokine levels. Lung tissue was collected for histological examination; Western blotting was used to measure levels of Mfn2, Drp1, cytochrome c, NF-κB p65, and IκBα. RESULTS Pretreatment with MitoQ significantly attenuated CS-induced thickening of the airway epithelium, peribronchial inflammatory cell infiltration, goblet cell hyperplasia and Muc5ac staining. The numbers of total cells, neutrophils and macrophages, as well as levels of TNF-α and IL-6 in BALF were remarkably decreased by MitoQ in a dose-dependent manner. MitoQ attenuated oxidative stress by preventing the CS-induced increase in malondialdehyde level and decrease in superoxide dismutase activity and GSH/GSSG ratio. MitoQ decreased the expression of mitochondrial fission protein Drp1 and increased that of mitochondrial fusion protein Mfn2, as well as reduced cytochrome c release into the cytosol. Furthermore, MitoQ suppressed IκBα degradation and NF-κB p65 nuclear translocation. CONCLUSIONS MitoQ attenuates inflammation, mucus hypersecretion, and oxidative stress induced by CS. It may exert these effects in part by modulating mitochondrial function and the NF-κB signal pathway.
Collapse
Affiliation(s)
- Deqing Yang
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, and Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Dan Xu
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, and Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Tao Wang
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, and Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Zhicheng Yuan
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, and Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Lian Liu
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, and Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yongchun Shen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, and Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China.
| | - Fuqiang Wen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, and Department of Respiratory and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China.
| |
Collapse
|
43
|
Chernyak BV, Popova EN, Zinovkina LA, Lyamzaev KG, Zinovkin RA. Mitochondria as Targets for Endothelial Protection in COVID-19. Front Physiol 2020; 11:606170. [PMID: 33329059 PMCID: PMC7710659 DOI: 10.3389/fphys.2020.606170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 10/27/2020] [Indexed: 12/27/2022] Open
Affiliation(s)
- Boris V Chernyak
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina N Popova
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ludmila A Zinovkina
- Faculty of Bioengineering and Bioinformatics, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Konstantin G Lyamzaev
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Roman A Zinovkin
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Moscow, Russia.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
44
|
Brand MD. Riding the tiger - physiological and pathological effects of superoxide and hydrogen peroxide generated in the mitochondrial matrix. Crit Rev Biochem Mol Biol 2020; 55:592-661. [PMID: 33148057 DOI: 10.1080/10409238.2020.1828258] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Elevated mitochondrial matrix superoxide and/or hydrogen peroxide concentrations drive a wide range of physiological responses and pathologies. Concentrations of superoxide and hydrogen peroxide in the mitochondrial matrix are set mainly by rates of production, the activities of superoxide dismutase-2 (SOD2) and peroxiredoxin-3 (PRDX3), and by diffusion of hydrogen peroxide to the cytosol. These considerations can be used to generate criteria for assessing whether changes in matrix superoxide or hydrogen peroxide are both necessary and sufficient to drive redox signaling and pathology: is a phenotype affected by suppressing superoxide and hydrogen peroxide production; by manipulating the levels of SOD2, PRDX3 or mitochondria-targeted catalase; and by adding mitochondria-targeted SOD/catalase mimetics or mitochondria-targeted antioxidants? Is the pathology associated with variants in SOD2 and PRDX3 genes? Filtering the large literature on mitochondrial redox signaling using these criteria highlights considerable evidence that mitochondrial superoxide and hydrogen peroxide drive physiological responses involved in cellular stress management, including apoptosis, autophagy, propagation of endoplasmic reticulum stress, cellular senescence, HIF1α signaling, and immune responses. They also affect cell proliferation, migration, differentiation, and the cell cycle. Filtering the huge literature on pathologies highlights strong experimental evidence that 30-40 pathologies may be driven by mitochondrial matrix superoxide or hydrogen peroxide. These can be grouped into overlapping and interacting categories: metabolic, cardiovascular, inflammatory, and neurological diseases; cancer; ischemia/reperfusion injury; aging and its diseases; external insults, and genetic diseases. Understanding the involvement of mitochondrial matrix superoxide and hydrogen peroxide concentrations in these diseases can facilitate the rational development of appropriate therapies.
Collapse
|
45
|
Ouyang L, Gong J. Mitochondrial-targeted ubiquinone: A potential treatment for COVID-19. Med Hypotheses 2020; 144:110161. [PMID: 32795832 PMCID: PMC7403158 DOI: 10.1016/j.mehy.2020.110161] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/16/2020] [Accepted: 08/01/2020] [Indexed: 12/16/2022]
Abstract
Immune dysregulation characterized by T cell exhaustion and high level of inflammatory cytokines is associated with severe COVID-19. Figuring out the early event of immune dysregulation would provide a potential treatment for COVID-19. Recent evidence indicate that mitochondrial dysfunction participates in the development of COVID-19 and may be responsible for the dysregulated immune response. Mitochondrial-targeted ubiquinone (MitoQ), a mitochondrial-targeted antioxidant, shows beneficial effects on various diseases through improving mitochondrial dysfunction. We hypothesize that MitoQ could act as a potential treatment in COVID-19. MitoQ may alleviate cytokine storm and restore the function of exhausted T cells in COVID-19 patients through improving mitochondrial dysfunction. In this article, we provide evidence to support the use of MitoQ as a potential treatment or adjunct therapy in the context of COVID-19.
Collapse
Affiliation(s)
- Lichen Ouyang
- Department of Immunology, School of Medicine, Jianghan University, Wuhan, China.
| | - Jie Gong
- The First Clinical College, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Anesthesiology and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
46
|
Marei WFA, Van den Bosch L, Pintelon I, Mohey-Elsaeed O, Bols PEJ, Leroy JLMR. Mitochondria-targeted therapy rescues development and quality of embryos derived from oocytes matured under oxidative stress conditions: a bovine in vitro model. Hum Reprod 2020; 34:1984-1998. [PMID: 31625574 DOI: 10.1093/humrep/dez161] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/05/2019] [Indexed: 12/20/2022] Open
Abstract
STUDY QUESTION Can we use a mitochondrial-targeted antioxidant (Mitoquinone) during in vitro embryo culture to rescue developmental competence of oocytes matured under lipotoxic conditions, exhibiting mitochondrial dysfunction and oxidative stress? SUMMARY ANSWER Supplementation of embryo culture media with Mitoquinone reduced oxidative stress and prevented mitochondrial uncoupling in embryos derived from metabolically compromised oocytes in vitro, leading to higher blastocyst rates and lower blastomeric apoptosis. WHAT IS KNOWN ALREADY Maternal metabolic disorders, such as obesity and type-II diabetes are associated with hyperlipidemia and elevated free fatty acid (FFA) concentrations in the ovarian follicular fluid (FF). Oocyte maturation under these lipotoxic conditions results in increased oxidative stress levels, mitochondrial dysfunction, reduced developmental competence and disappointing IVF results. STUDY DESIGN, SIZE, DURATION A well-described bovine oocyte IVM model was used, where a pathophysiologically relevant elevated FF concentrations of palmitic acid (PA; 150 μM or 300 μM) were added to induce oxidative stress. After fertilization (Day 0, D0), zygotes were in vitro cultured (IVC, from D1 to D8) in standard fatty acid-free media in the presence or absence of Mitoquinone or its carrier triphenyl-phosphonium. PARTICIPANTS/MATERIALS, SETTING, METHODS Embryo cleavage and fragmentation (D2) and blastocyst rates (D8) were recorded. Mitochondrial activity and oxidative stress in cleaved embryos at D2 were determined using specific fluorogenic probes and confocal microscopy. D8 blastocysts were used to (i) examine the expression of marker genes related to mitochondrial unfolded protein responses (UPRmt; HSPD1 and HSPE1), mitochondrial biogenesis (TFAM), endoplasmic reticulum (ER) UPR (ATF4, ATF6 and BiP) and oxidative stress (CAT, GPX1 and SOD2) using real time RT-PCR; (ii) determine cell differentiation and apoptosis using CDX-2 and cleaved caspase-3 immunostaining; and (iii) measure mtDNA copy numbers. This was tested in a series of experiments with at least three independent replicates for each, using a total of 2525 oocytes. Differences were considered significant if a P value was <0.05 after Bonferroni correction. MAIN RESULTS AND THE ROLE OF CHANCE Exposure to PA during IVM followed by culture under control conditions resulted in a significant increase in oxidative stress in embryos at D2. This was associated with a significant reduction in mitochondrial inner membrane potential (uncoupling) compared with solvent control (P < 0.05). The magnitude of these effects was PA-concentration dependent. Consequently, development to the blastocysts stage was significantly hampered. Surviving blastocysts exhibited high apoptotic cell indices and upregulated mRNA expression indicating persistent oxidative stress, mitochondrial and ER UPRs. In contrast, supplementation of PA-derived zygotes with Mitoquinone during IVC (i) prevented mitochondrial uncoupling and alleviated oxidative stress at D2; and (ii) rescued blastocyst quality; normalized oxidative stress and UPR related genes and apoptotic cell indices (P > 0.01 compared with solvent control). Mitoquinone also improved blastocyst rate in PA-exposed groups, an effect that was dependent on PA concentration. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION This is a fundamental study performed using a bovine in vitro model using PA-induced lipotoxicity during oocyte maturation. PA is the most predominant FFA in the FF that is known to induce lipotoxicity; however, in vivo maturation in patients suffering from maternal metabolic disorders involve more factors that cannot be represented in one model. Nevertheless, focusing on the carryover oxidative stress as a known key factor affecting developmental competence, and considering the novel beneficial rescuing effects of Mitoquinone shown here, we believe this model is of high biological relevance. WIDER IMPLICATIONS OF THE FINDINGS Human oocytes collected for IVF treatments from patients with maternal metabolic disorders are vulnerable to lipotoxicity and oxidative stress during in vivo maturation. The results shown here suggest that mitochondrial targeted therapy, such as using Mitoquinone, during IVC may rescue the developmental competence and quality of these compromised oocytes. After further clinical trials, this may be a valuable approach to increase IVF success rates for infertile patients experiencing metabolic disorders. STUDY FUNDING/COMPETING INTEREST(S) This study was financially supported by a BOF/KP grant number 34399, from the University of Antwerp, Belgium. W.F.A.M. was supported by a postdoctoral fellowship from the Research Foundation-Flanders (FWO), grant number 12I1417N, Antwerp, Belgium. The Leica SP 8 confocal microscope used in this study was funded by the Hercules Foundation of the Flemish Government (Hercules grant AUHA.15.12). All authors have no financial or non-financial competing interests to declare.
Collapse
Affiliation(s)
- Waleed F A Marei
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium.,Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
| | - Lotte Van den Bosch
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Isabel Pintelon
- Laboratory of Cell Biology and Histology, University of Antwerp, 2610 Wilrijk, Belgium
| | - Omnia Mohey-Elsaeed
- Laboratory of Cell Biology and Histology, University of Antwerp, 2610 Wilrijk, Belgium.,Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
| | - Peter E J Bols
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Jo L M R Leroy
- Gamete Research Centre, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
47
|
Vezza T, Abad-Jiménez Z, Marti-Cabrera M, Rocha M, Víctor VM. Microbiota-Mitochondria Inter-Talk: A Potential Therapeutic Strategy in Obesity and Type 2 Diabetes. Antioxidants (Basel) 2020; 9:antiox9090848. [PMID: 32927712 PMCID: PMC7554719 DOI: 10.3390/antiox9090848] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
The rising prevalence of obesity and type 2 diabetes (T2D) is a growing concern worldwide. New discoveries in the field of metagenomics and clinical research have revealed that the gut microbiota plays a key role in these metabolic disorders. The mechanisms regulating microbiota composition are multifactorial and include resistance to stress, presence of pathogens, diet, cultural habits and general health conditions. Recent evidence has shed light on the influence of microbiota quality and diversity on mitochondrial functions. Of note, the gut microbiota has been shown to regulate crucial transcription factors, coactivators, as well as enzymes implicated in mitochondrial biogenesis and metabolism. Moreover, microbiota metabolites seem to interfere with mitochondrial oxidative/nitrosative stress and autophagosome formation, thus regulating the activation of the inflammasome and the production of inflammatory cytokines, key players in chronic metabolic disorders. This review focuses on the association between intestinal microbiota and mitochondrial function and examines the mechanisms that may be the key to their use as potential therapeutic strategies in obesity and T2D management.
Collapse
Affiliation(s)
- Teresa Vezza
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (T.V.); (Z.A.-J.)
| | - Zaida Abad-Jiménez
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (T.V.); (Z.A.-J.)
| | | | - Milagros Rocha
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (T.V.); (Z.A.-J.)
- CIBERehd—Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
- Correspondence: (M.R.); (V.M.V.); Tel.: +34-963-189-132 (M.R. & V.M.V.); Fax: +34-961-622-492 (M.R. & V.M.V.)
| | - Víctor Manuel Víctor
- Service of Endocrinology and Nutrition, University Hospital Doctor Peset, Foundation for the Promotion of Health and Biomedical Research in the Valencian Region (FISABIO), 46017 Valencia, Spain; (T.V.); (Z.A.-J.)
- CIBERehd—Department of Pharmacology, University of Valencia, 46010 Valencia, Spain
- Department of Physiology, University of Valencia, 46010 Valencia, Spain
- Correspondence: (M.R.); (V.M.V.); Tel.: +34-963-189-132 (M.R. & V.M.V.); Fax: +34-961-622-492 (M.R. & V.M.V.)
| |
Collapse
|
48
|
Oxidative Stress in Reproduction: A Mitochondrial Perspective. BIOLOGY 2020; 9:biology9090269. [PMID: 32899860 PMCID: PMC7564700 DOI: 10.3390/biology9090269] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/31/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022]
Abstract
Mitochondria are fundamental organelles in eukaryotic cells that provide ATP through oxidative phosphorylation. During this process, reactive oxygen species (ROS) are produced, and an imbalance in their concentrations can induce oxidative stress (OS), causing cellular damage. However, mitochondria and ROS play also an important role in cellular homeostasis through a variety of other signaling pathways not related to metabolic rates, highlighting the physiological relevance of mitochondria–ROS interactions. In reproduction, mitochondria follow a peculiar pattern of activation, especially in gametes, where they are relatively inactive during the initial phases of development, and become more active towards the final maturation stages. The reasons for the lower metabolic rates are attributed to the evolutionary advantage of keeping ROS levels low, thus avoiding cellular damage and apoptosis. In this review, we provide an overview on the interplay between mitochondrial metabolism and ROS during gametogenesis and embryogenesis, and how OS can influence these physiological processes. We also present the possible effects of assisted reproduction procedures on the levels of OS, and the latest techniques developed to select gametes and embryos based on their redox state. Finally, we evaluate the treatments developed to manage OS in assisted reproduction to improve the chances of pregnancy.
Collapse
|
49
|
Yang J, Suo H, Song J. Protective role of mitoquinone against impaired mitochondrial homeostasis in metabolic syndrome. Crit Rev Food Sci Nutr 2020; 61:3857-3875. [PMID: 32815398 DOI: 10.1080/10408398.2020.1809344] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mitochondria control various processes in cellular metabolic homeostasis, such as adenosine triphosphate production, generation and clearance of reactive oxygen species, control of intracellular Ca2+ and apoptosis, and are thus a critical therapeutic target for metabolic syndrome (MetS). The mitochondrial targeted antioxidant mitoquinone (MitoQ) reduces mitochondrial oxidative stress, prevents impaired mitochondrial dynamics, and increases mitochondrial turnover by promoting autophagy (mitophagy) and mitochondrial biogenesis, which ultimately contribute to the attenuation of MetS conditions, including obesity, insulin resistance, hypertension and cardiovascular disease. The regulatory effect of MitoQ on mitochondrial homeostasis is mediated through AMPK and its downstream signaling pathways, including MTOR, SIRT1, Nrf2 and NF-κB. However, there are few reviews focusing on the critical role of MitoQ as a therapeutic agent in the treatment of MetS. The purpose of this review is to summarize the mitochondrial role in the pathogenesis of MetS, especially in obesity and type 2 diabetes, and discuss the effect and underlying mechanism of MitoQ on mitochondrial homeostasis in MetS.
Collapse
Affiliation(s)
- Jing Yang
- Chongqing Engineering Research Center for Processing & Storage of Distinct Agricultural Products, Chongqing Technology and Business University, Chongqing, China.,Graduate School, Chongqing Technology and Business University, Chongqing, China
| | - Huayi Suo
- College of Food Science, Southwest University, Chongqing, China
| | - Jiajia Song
- College of Food Science, Southwest University, Chongqing, China
| |
Collapse
|
50
|
Bordoni L, Gabbianelli R. Mitochondrial DNA and Neurodegeneration: Any Role for Dietary Antioxidants? Antioxidants (Basel) 2020; 9:E764. [PMID: 32824558 PMCID: PMC7466149 DOI: 10.3390/antiox9080764] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
The maintenance of the mitochondrial function is essential in preventing and counteracting neurodegeneration. In particular, mitochondria of neuronal cells play a pivotal role in sustaining the high energetic metabolism of these cells and are especially prone to oxidative damage. Since overproduction of reactive oxygen species (ROS) is involved in the pathogenesis of neurodegeneration, dietary antioxidants have been suggested to counteract the detrimental effects of ROS and to preserve the mitochondrial function, thus slowing the progression and limiting the extent of neuronal cell loss in neurodegenerative disorders. In addition to their role in the redox-system homeostasis, mitochondria are unique organelles in that they contain their own genome (mtDNA), which acts at the interface between environmental exposures and the molecular triggers of neurodegeneration. Indeed, it has been demonstrated that mtDNA (including both genetics and, from recent evidence, epigenetics) might play relevant roles in modulating the risk for neurodegenerative disorders. This mini-review describes the link between the mitochondrial genome and cellular oxidative status, with a particular focus on neurodegeneration; moreover, it provides an overview on potential beneficial effects of antioxidants in preserving mitochondrial functions through the protection of mtDNA.
Collapse
Affiliation(s)
- Laura Bordoni
- Unit of Molecular Biology, School of Pharmacy, University of Camerino, 62032 Camerino, Italy;
| | | |
Collapse
|