1
|
Luo H, Jin M, Hu H, Ying Q, Hu P, Sheng W, Huang Y, Xu K, Lu C, Zhang X. SIRT4 Protects Müller Glial Cells Against Apoptosis by Mediating Mitochondrial Dynamics and Oxidative Stress. Mol Neurobiol 2025; 62:6683-6702. [PMID: 39023793 DOI: 10.1007/s12035-024-04349-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
SIRT4 is a member of the sirtuin family, which is related to mitochondrial function and possesses antioxidant and regulatory redox effects. Currently, the roles of SIRT4 in retinal Müller glial cells, oxidative stress, and mitochondrial function are still unclear. We confirmed, by immunofluorescence staining, that SIRT4 is located mainly in the mitochondria of retinal Müller glial cells. Using flow cytometry and Western blotting, we analyzed cell apoptosis, intracellular reactive oxygen species (ROS) levels, apoptotic and proapoptotic proteins, mitochondrial dynamics-related proteins, and mitochondrial morphology and number after the overexpression and downregulation of SIRT4 in rMC-1 cells. Neither the upregulation nor the downregulation of SIRT4 alone affected apoptosis. SIRT4 overexpression reduced intracellular ROS, reduced the BAX/BCL2 protein ratio, and increased the L-OPA/S-OPA1 ratio and the levels of the mitochondrial fusion protein MFN2 and the mitochondrial cleavage protein FIS1, increasing mitochondrial fusion. SIRT4 downregulation had the opposite effect. Mitochondria tend to divide after serum starvation for 24 h, and SIRT4 downregulation increases mitochondrial fragmentation and oxidative stress, leading to aggravated cell damage. The mitochondrial division inhibitor Mdivi-1 reduced oxidative stress levels and thus reduced cell damage caused by serum starvation. The overexpression of SIRT4 in rMC-1 cells reduced mitochondrial fragmentation caused by serum starvation, leading to mitochondrial fusion and reduced expression of cleaved caspase-3, thus alleviating the cellular damage caused by oxidative stress. Thus, we speculate that SIRT4 may protect retinal Müller glial cells against apoptosis by mediating mitochondrial dynamics and oxidative stress.
Collapse
Affiliation(s)
- Hongdou Luo
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Ming Jin
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Haijian Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Qian Ying
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Piaopiao Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Weiwei Sheng
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Yi Huang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Ke Xu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Chuming Lu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Jiangxi Clinical Research Center for Ophthalmic Disease, 463 Bayi Road, Nanchang, 330006, China.
| |
Collapse
|
2
|
Li Z, Liang S, Ke L, Wang M, Gao K, Li D, Xu Z, Li N, Zhang P, Cheng W. Cell life-or-death events in osteoporosis: All roads lead to mitochondrial dynamics. Pharmacol Res 2024; 208:107383. [PMID: 39214266 DOI: 10.1016/j.phrs.2024.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria exhibit heterogeneous shapes and networks within and among cell types and tissues, also in normal or osteoporotic bone tissues with complex cell types. This dynamic characteristic is determined by the high plasticity provided by mitochondrial dynamics and is stemmed from responding to the survival and functional requirements of various bone cells in a specific microenvironments. In contrast, mitochondrial dysfunction, induced by dysregulation of mitochondrial dynamics, may act as a trigger of cell death signals, including common apoptosis and other forms of programmed cell death (PCD). These PCD processes consisting of tightly structured cascade gene expression events, can further influence the bone remodeling by facilitating the death of various bone cells. Mitochondrial dynamics, therefore, drive the bone cells to stand at the crossroads of life and death by integrating external signals and altering metabolism, shape, and signal-response properties of mitochondria. This implies that targeting mitochondrial dynamics displays significant potential in treatment of osteoporosis. Considerable effort has been made in osteoporosis to emphasize the parallel roles of mitochondria in regulating energy metabolism, calcium signal transduction, oxidative stress, inflammation, and cell death. However, the emerging field of mitochondrial dynamics-related PCD is not well understood. Herein, to bridge the gap, we outline the latest knowledge on mitochondrial dynamics regulating bone cell life or death during normal bone remodeling and osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050011, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000, China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300, China.
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Yao BF, Luo XJ, Peng J. A review for the correlation between optic atrophy 1-dependent mitochondrial fusion and cardiovascular disorders. Int J Biol Macromol 2024; 254:127910. [PMID: 37939779 DOI: 10.1016/j.ijbiomac.2023.127910] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/19/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Mitochondrial dynamics homeostasis is sustained by continuous and balanced fission and fusion, which are determinants of morphology, abundance, biogenesis and mitophagy of mitochondria. Optic atrophy 1 (OPA1), as the only inner mitochondrial membrane fusion protein, plays a key role in stabilizing mitochondrial dynamics. The disturbance of mitochondrial dynamics contributes to the pathophysiological progress of cardiovascular disorders, which are the main cause of death worldwide in recent decades and result in tremendous social burden. In this review, we describe the latest findings regarding OPA1 and its role in mitochondrial fusion. We summarize the post-translational modifications (PTMs) for OPA1 and its regulatory role in mitochondrial dynamics. Then the diverse cell fates caused by OPA1 expression during cardiovascular disorders are discussed. Moreover, cardiovascular disorders (such as heart failure, myocardial ischemia/reperfusion injury, cardiomyopathy and cardiac hypertrophy) relevant to OPA1-dependent mitochondrial dynamics imbalance have been detailed. Finally, we highlight the potential that targeting OPA1 to impact mitochondrial fusion may be used as a novel strategy against cardiovascular disorders.
Collapse
Affiliation(s)
- Bi-Feng Yao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| |
Collapse
|
4
|
Yang Z, Liu Y, Chen X, Huang S, Li Y, Ye G, Cao X, Su W, Zhuo Y. Empagliflozin targets Mfn1 and Opa1 to attenuate microglia-mediated neuroinflammation in retinal ischemia and reperfusion injury. J Neuroinflammation 2023; 20:296. [PMID: 38082266 PMCID: PMC10714482 DOI: 10.1186/s12974-023-02982-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Neuroinflammation and mitochondrial dysfunction play crucial roles in retinal ischemia and reperfusion (IR) injury. Recent studies have identified mitochondrial function as a promising target for immunomodulation. Empagliflozin (EMPA), an anti-diabetic drug, has exhibited great potential as both an anti-inflammatory agent and a protector of mitochondrial health. This study aimed to assess the therapeutic efficacy of EMPA in retinal IR injury. METHODS To evaluate the protective effects of EMPA, the drug was injected into the vitreous body of mice post-retinal IR. Single-cell RNA sequencing (scRNA-seq) analysis was conducted to uncover the underlying mechanisms, and the results were further validated through in vivo and in vitro experiments. RESULTS EMPA effectively protected retinal ganglion cells (RGCs) from IR injury by attenuating local retinal inflammation. The scRNA-seq analysis revealed that EMPA downregulated the nucleotide-binding domain and leucine-rich repeat containing protein 3 (NLRP3) signaling pathway and restored mitochondrial dynamics by upregulating the expression of mitochondrial fusion-related genes, Mitofusin 1 (Mfn1) and optic atrophy 1 (Opa1). These findings were further corroborated by Western blotting. In vitro experiments provided additional insights, demonstrating that EMPA suppressed lipopolysaccharide (LPS)-induced cell inflammation and NLRP3 inflammasome activation. Moreover, EMPA enhanced mitochondrial fusion, neutralized mitochondrial reactive oxygen species (mtROS), and restored mitochondrial membrane potential (MMP) in BV2 microglia. Notably, genetic ablation of Mfn1 or Opa1 abolished the anti-inflammatory effects of EMPA. CONCLUSIONS Our findings highlight the positive contribution of Mfn1 and Opa1 to the anti-inflammatory therapeutic effect of EMPA. By restoring mitochondrial dynamics, EMPA effectively mitigates microglia-mediated neuroinflammation and prevents RGC loss in retinal IR injury.
Collapse
Affiliation(s)
- Zhenlan Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yidan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xuhao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Shaofen Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yangyang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Guitong Ye
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xu Cao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
5
|
Yuan Y, Zhang XM. Mechanistic study of optic atrophy 1 in ischemia-reperfusion disease. J Mol Med (Berl) 2023; 101:1-8. [PMID: 36418744 DOI: 10.1007/s00109-022-02271-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/13/2022] [Accepted: 11/09/2022] [Indexed: 11/25/2022]
Abstract
Mitochondria consist of the inner mitochondrial membrane and the outer mitochondrial membrane, which maintain mitochondrial homeostasis through continuous fission and fusion to ensure a healthy mitochondrial network and thus regulate normal cellular function, namely mitochondrial dynamics. The imbalance between mitochondrial fusion and fission results in abnormal mitochondrial structure and eventually mitochondrial dysfunction, which is involved in the pathological process of ischemia-reperfusion injury (IRI). Optic atrophy 1 (OPA1) is a key protein that regulates mitochondrial inner membrane fusion and ensures normal mitochondrial function by balancing mitochondrial dynamics, participating in various processes such as mitochondrial fusion, oxidative stress, and apoptosis. Ischemia-induced changes in mitochondrial dynamics may be a key factor in limiting the recanalization time window and exacerbating reperfusion injury, and the mechanisms of these changes deserve further attention. Therefore, targeting OPA1-related mitochondrial fusions, thereby balancing mitochondrial dynamics and improving mitochondrial dysfunction, is a promising therapeutic strategy for ischemia-reperfusion diseases. This review will elaborate on the structure and function of OPA1 and the role of OPA1 in IRI to provide promising therapeutic targets for the treatment of ischemia-reperfusion diseases.
Collapse
Affiliation(s)
- Ying Yuan
- College of Acupuncture, Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiao-Ming Zhang
- College of Acupuncture, Moxibustion and Orthopedics, Hubei University of Chinese Medicine, Wuhan, 430065, China. .,Sub-health institute Hubei university of Chinese Medicine, Wuhan, 430065, China. .,Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion Wuhan, Wuhan, 430065, China.
| |
Collapse
|
6
|
Chen TW, Wu PY, Wen YT, Desai TD, Huang CT, Liu PK, Tsai RK. Vitamin B3 Provides Neuroprotection via Antioxidative Stress in a Rat Model of Anterior Ischemic Optic Neuropathy. Antioxidants (Basel) 2022; 11:antiox11122422. [PMID: 36552630 PMCID: PMC9774344 DOI: 10.3390/antiox11122422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Supplementing with vitamin B3 has been reported to protect against retinal ganglion cell (RGC) damage events and exhibit multiple neuroprotective properties in a mouse model of optic nerve injury. In this study, a rat model of anterior ischemic optic neuropathy was used to assess the neuroprotective benefits of vitamin B3 (rAION). Vitamin B3 (500 mg/kg/day) or phosphate-buffered saline (PBS) was administered to the rAION-induced rats every day for 28 days. The vitamin B3-treated group had significantly higher first positive and second negative peak (P1-N2) amplitudes of flash visual-evoked potentials and RGC densities than the PBS-treated group (p < 0.05). A terminal deoxynucleotidyl transferase dUTP nick end labeling assay conducted on vitamin B3-treated rats revealed a significant reduction in apoptotic cells (p < 0.05). Superoxide dismutase and thiobarbituric acid reactive substance activity showed that vitamin B3 treatment decreased reactive oxygen species (p < 0.05). Therefore, vitamin B3 supplementation preserves vision in rAION-induced rats by reducing oxidative stress, neuroinflammation, and mitochondrial apoptosis.
Collapse
Affiliation(s)
- Tu-Wen Chen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Po-Ying Wu
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yao-Tseng Wen
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Tushar Dnyaneshwar Desai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
| | - Chin-Te Huang
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Department of Ophthalmology, School of Medicine, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 402, Taiwan
| | - Pei-Kang Liu
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Rong-Kung Tsai
- Institute of Eye Research, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan
- Institute of Medical Sciences, Tzu Chi University, Hualien 970, Taiwan
- Doctoral Degree Program in Translational Medicine, Tzu Chi University and Academia Sinica, Hualien 970, Taiwan
- Correspondence:
| |
Collapse
|
7
|
Soldatov VO, Kubekina MV, Skorkina MY, Belykh AE, Egorova TV, Korokin MV, Pokrovskiy MV, Deykin AV, Angelova PR. Current advances in gene therapy of mitochondrial diseases. J Transl Med 2022; 20:562. [PMID: 36471396 PMCID: PMC9724384 DOI: 10.1186/s12967-022-03685-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/04/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial diseases (MD) are a heterogeneous group of multisystem disorders involving metabolic errors. MD are characterized by extremely heterogeneous symptoms, ranging from organ-specific to multisystem dysfunction with different clinical courses. Most primary MD are autosomal recessive but maternal inheritance (from mtDNA), autosomal dominant, and X-linked inheritance is also known. Mitochondria are unique energy-generating cellular organelles designed to survive and contain their own unique genetic coding material, a circular mtDNA fragment of approximately 16,000 base pairs. The mitochondrial genetic system incorporates closely interacting bi-genomic factors encoded by the nuclear and mitochondrial genomes. Understanding the dynamics of mitochondrial genetics supporting mitochondrial biogenesis is especially important for the development of strategies for the treatment of rare and difficult-to-diagnose diseases. Gene therapy is one of the methods for correcting mitochondrial disorders.
Collapse
Affiliation(s)
- Vladislav O Soldatov
- Core Facility Centre, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia.
- Laboratory of Genome Editing for Biomedicine and Animal Health, Belgorod State National Research University, Belgorod, Russia.
| | - Marina V Kubekina
- Core Facility Centre, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Marina Yu Skorkina
- Department of Biochemistry, Belgorod State National Research University, Belgorod, Russia
- Laboratory of Genome Editing for Biomedicine and Animal Health, Belgorod State National Research University, Belgorod, Russia
| | - Andrei E Belykh
- Dioscuri Centre for Metabolic Diseases, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Tatiana V Egorova
- Laboratory of Modeling and Gene Therapy of Hereditary Diseases, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| | - Mikhail V Korokin
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Mikhail V Pokrovskiy
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Alexey V Deykin
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
- Laboratory of Genome Editing for Biomedicine and Animal Health, Belgorod State National Research University, Belgorod, Russia
| | - Plamena R Angelova
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
| |
Collapse
|
8
|
Grossini E, Venkatesan S, Alkabes M, Toma C, de Cillà S. Membrane Blue Dual Protects Retinal Pigment Epithelium Cells/Ganglion Cells-Like through Modulation of Mitochondria Function. Biomedicines 2022; 10:2854. [PMID: 36359372 PMCID: PMC9687626 DOI: 10.3390/biomedicines10112854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 10/29/2023] Open
Abstract
Although recent data highlight the greater protective effects exerted by Membrane Blue Dual (MBD), a precise analysis of the mechanisms of action is missing. We examined the effects of MBD with/without polyethylene glycol (PEG) on both human retinal pigment epithelial cells (ARPE-19) and retinal ganglion cells-like (RGC-5) cultured in the presence/absence of ultraviolet B (UVB) treatment on mitochondria function, oxidants, and apoptosis. In ARPE-19/RGC-5 cells either treated or not with UVB, the effects of MBD with/without PEG were evaluated by specific assays for viability, mitochondrial membrane potential and mitochondrial reactive oxygen species (mitoROS) release. Annexin V was used to detect apoptosis, whereas trypan blue and the scratch assay were used for proliferation/migration. In both physiologic conditions and in the presence of UVB, MBD with/without PEG increased cell viability, mitochondrial membrane potential, proliferation and migration in both ARPE-19 and RGC-5 cells. In general, the effects of MBD with PEG were greater than those caused by MBD without PEG. Our results suggest that, in particular, MBD with PEG is a safe and effective dye for vitreoretinal surgery through the modulation of mitochondrial function.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, University Eastern Piedmont, 28100 Novara, Italy
- AGING Project Unit, Department of Translational Medicine, University Eastern Piedmont, 28100 Novara, Italy
| | - Sakthipriyan Venkatesan
- Laboratory of Physiology, Department of Translational Medicine, University Eastern Piedmont, 28100 Novara, Italy
- AGING Project Unit, Department of Translational Medicine, University Eastern Piedmont, 28100 Novara, Italy
| | - Micol Alkabes
- Eye Clinic, University Hospital Maggiore della Carità, 28100 Novara, Italy
| | - Caterina Toma
- Eye Clinic, University Hospital Maggiore della Carità, 28100 Novara, Italy
- Department of Health Sciences, University East Piedmont “A. Avogadro”, 28100 Novara, Italy
| | - Stefano de Cillà
- Eye Clinic, University Hospital Maggiore della Carità, 28100 Novara, Italy
- Department of Health Sciences, University East Piedmont “A. Avogadro”, 28100 Novara, Italy
| |
Collapse
|
9
|
Wang L, Yang Z, He X, Pu S, Yang C, Wu Q, Zhou Z, Cen X, Zhao H. Mitochondrial protein dysfunction in pathogenesis of neurological diseases. Front Mol Neurosci 2022; 15:974480. [PMID: 36157077 PMCID: PMC9489860 DOI: 10.3389/fnmol.2022.974480] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Mitochondria are essential organelles for neuronal function and cell survival. Besides the well-known bioenergetics, additional mitochondrial roles in calcium signaling, lipid biogenesis, regulation of reactive oxygen species, and apoptosis are pivotal in diverse cellular processes. The mitochondrial proteome encompasses about 1,500 proteins encoded by both the nuclear DNA and the maternally inherited mitochondrial DNA. Mutations in the nuclear or mitochondrial genome, or combinations of both, can result in mitochondrial protein deficiencies and mitochondrial malfunction. Therefore, mitochondrial quality control by proteins involved in various surveillance mechanisms is critical for neuronal integrity and viability. Abnormal proteins involved in mitochondrial bioenergetics, dynamics, mitophagy, import machinery, ion channels, and mitochondrial DNA maintenance have been linked to the pathogenesis of a number of neurological diseases. The goal of this review is to give an overview of these pathways and to summarize the interconnections between mitochondrial protein dysfunction and neurological diseases.
Collapse
Affiliation(s)
- Liang Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Ziyun Yang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Xiumei He
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Shiming Pu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Cheng Yang
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Qiong Wu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Zuping Zhou
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Hongxia Zhao
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
10
|
Jiang HL, Yang HH, Liu YB, Zhang CY, Zhong WJ, Guan XX, Jin L, Hong JR, Yang JT, Tan XH, Li Q, Zhou Y, Guan CX. L-OPA1 deficiency aggravates necroptosis of alveolar epithelial cells through impairing mitochondrial function during ALI in mice. J Cell Physiol 2022; 237:3030-3043. [PMID: 35478455 DOI: 10.1002/jcp.30766] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 11/11/2022]
Abstract
Necroptosis, a recently described form of programmed cell death, is the main way of alveolar epithelial cells (AECs) death in acute lung injury (ALI). While the mechanism of how to trigger necroptosis in AECs during ALI has been rarely evaluated. Long optic atrophy protein 1 (L-OPA1) is a crucial mitochondrial inner membrane fusion protein, and its deficiency impairs mitochondrial function. This study aimed to investigate the role of L-OPA1 deficiency-mediated mitochondrial dysfunction in AECs necroptosis. We comprehensively investigated the detailed contribution and molecular mechanism of L-OPA1 deficiency in AECs necroptosis by inhibiting or activating L-OPA1. Firstly, our data showed that L-OPA1 expression was down-regulated in the lungs and AECs under the lipopolysaccharide (LPS) challenge. Furthermore, inhibition of L-OPA1 aggravated the pathological injury, inflammatory response, and necroptosis in the lungs of LPS-induced ALI mice. In vitro, inhibition of L-OPA1 induced necroptosis of AECs, while activation of L-OPA1 alleviated necroptosis of AECs under the LPS challenge. Mechanistically, inhibition of L-OPA1 aggravated necroptosis of AECs by inducing mitochondrial fragmentation and reducing mitochondrial membrane potential. While activation of L-OPA1 had the opposite effects. In summary, these findings indicate for the first time that L-OPA1 deficiency mediates mitochondrial fragmentation, induces necroptosis of AECs, and exacerbates ALI in mice. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hui-Ling Jiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Hui-Hui Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Yu-Biao Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Chen-Yu Zhang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Xin-Xin Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Ling Jin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jie-Ru Hong
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Jin-Tong Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Xiao-Hua Tan
- Experimental Center of Medical Morphology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Qing Li
- Department of Physiology, Hunan University of Medicine, Huaihua, Hunan, 418000, China
| | - Yong Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, Hunan, 410078, China
| |
Collapse
|
11
|
Li X, Li H, Xu Z, Ma C, Wang T, You W, Yu Z, Shen H, Chen G. Ischemia-induced cleavage of OPA1 at S1 site aggravates mitochondrial fragmentation and reperfusion injury in neurons. Cell Death Dis 2022; 13:321. [PMID: 35395832 PMCID: PMC8993832 DOI: 10.1038/s41419-022-04782-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 03/07/2022] [Accepted: 03/22/2022] [Indexed: 12/12/2022]
Abstract
Neuronal mitochondrial dynamics are disturbed after ischemic stroke. Optic atrophy 1 (OPA1) and its GTPase activity are involved in maintaining mitochondrial cristae and inner membrane fusion. This study aimed to explore the role of OMA1-mediated OPA1 cleavage (S1-OPA1) in neurons exposed to cerebral ischemia and reperfusion. After oxygen-glucose deprivation (OGD) for 60 min, we found that mitochondrial fragmentation occurred successively in the axon and soma of neurons, accompanied by an increase in S1-OPA1. In addition, S1-OPA1 overexpression significantly aggravated mitochondrial damage in neurons exposed to OGD for 60 min and 24 h after OGD/R, characterized by mitochondrial fragmentation, decreased mitochondrial membrane potential, mitochondrial cristae ultrastructural damage, increased superoxide production, decreased ATP production and increased mitochondrial apoptosis, which was inhibited by the lysine 301 to alanine mutation (K301A). Furthermore, we performed neuron-specific overexpression of S1-OPA1 in the cerebral cortex around ischemia of middle cerebral artery occlusion/reperfusion (MCAO/R) mice. The results further demonstrated in vivo that S1-OPA1 exacerbated neuronal mitochondrial ultrastructural destruction and injury induced by cerebral ischemia-reperfusion, while S1-OPA1-K301 overexpression had no effect. In conclusion, ischemia induced neuronal OMA1-mediated cleavage of OPA1 at the S1 site. S1-OPA1 aggravated neuronal mitochondrial fragmentation and damage in a GTPase-dependent manner, and participated in neuronal ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
- Institute of Stroke Research, Soochow University, Suzhou, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
- Institute of Stroke Research, Soochow University, Suzhou, China
| | - Zhongmou Xu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
- Institute of Stroke Research, Soochow University, Suzhou, China
| | - Cheng Ma
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
- Institute of Stroke Research, Soochow University, Suzhou, China
| | - Tianyi Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
- Institute of Stroke Research, Soochow University, Suzhou, China
| | - Wanchun You
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
- Institute of Stroke Research, Soochow University, Suzhou, China
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
- Institute of Stroke Research, Soochow University, Suzhou, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China
- Institute of Stroke Research, Soochow University, Suzhou, China
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou, Jiangsu Province, 215006, China.
- Institute of Stroke Research, Soochow University, Suzhou, China.
| |
Collapse
|
12
|
Zhou X, Fang G, Zhang L. Optimization of murine retinal mitochondrial injury model. MethodsX 2022; 9:101701. [PMID: 35492209 PMCID: PMC9046943 DOI: 10.1016/j.mex.2022.101701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/08/2022] [Indexed: 11/30/2022] Open
Abstract
The retinal mitochondrial injury model in rat has been developed using the mitochondrial oxidative phosphorylation uncoupler, carbonylcyanide m-chlorophenyl hydrazine (CCCP). However, the CCCP-induced murine retinal mitochondrial injury model has not been reported. Here, the optimized conditions for the murine retinal mitochondrial injury model were established by intravitreal injection of different doses of CCCP (0, 2.5, 5, 7.5, 10, 12.5, 15 μg). Indeed, it has been reported that CCCP induces Opa1 cleavage and phosphorylation of ERK in cultured cells and rat retinas. Thus, we measured phosphorylated (p) -Erk and L/S-Opa1 following CCCP-induced retinal injury. Meanwhile, KW6002 (A2A receptor antagonist) pretreatment inhibited retinal injury induced by CCCP at 10 and 15 μg doses differently. Intravitreal injection of 10 μg doses of CCCP can induce apoptosis of retinal ganglion cells and decrease of retinal thickness, but intravitreal injection of 15 μg doses of CCCP is the appropriate dose to study the protective effect of A2A receptor. (1) Dose dependent effects of intravitreal injection of CCCP on the levels of L/S-Opa1 and p-Erk; (2) A2A receptor antagonist (KW6002) only inhibited the apoptosis of ganglion cells, but did not affect the thickness of retina with 10µg dosage of CCCP intravitreal injection; (3) A2A receptor antagonist (KW6002) inhibited the apoptosis of ganglion cells and increased the thickness of retina with 15µg dosage of CCCP intravitreal injection.
Collapse
|
13
|
Mitochondrial dynamics in the neonatal brain - a potential target following injury? Biosci Rep 2022; 42:231001. [PMID: 35319070 PMCID: PMC8965818 DOI: 10.1042/bsr20211696] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/09/2022] [Accepted: 03/11/2022] [Indexed: 02/08/2023] Open
Abstract
The impact of birth asphyxia and its sequelae, hypoxic–ischaemic (HI) brain injury, is long-lasting and significant, both for the infant and for their family. Treatment options are limited to therapeutic hypothermia, which is not universally successful and is unavailable in low resource settings. The energy deficits that accompany neuronal death following interruption of blood flow to the brain implicate mitochondrial dysfunction. Such HI insults trigger mitochondrial outer membrane permeabilisation leading to release of pro-apoptotic proteins into the cytosol and cell death. More recently, key players in mitochondrial fission and fusion have been identified as targets following HI brain injury. This review aims to provide an introduction to the molecular players and pathways driving mitochondrial dynamics, the regulation of these pathways and how they are altered following HI insult. Finally, we review progress on repurposing or repositioning drugs already approved for other indications, which may target mitochondrial dynamics and provide promising avenues for intervention following brain injury. Such repurposing may provide a mechanism to fast-track, low-cost treatment options to the clinic.
Collapse
|
14
|
Fang G, Zhou Y, Zhou X, Zhou H, Ge YY, Luo S, Chen JF, Zhang L. The adenosine A 2A receptor antagonist protects against retinal mitochondrial injury in association with an altered network of competing endogenous RNAs. Neuropharmacology 2022; 208:108981. [PMID: 35149135 DOI: 10.1016/j.neuropharm.2022.108981] [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: 09/16/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 10/19/2022]
Abstract
Blockade of adenosine A2A receptors (A2ARs) protects against neuronal damage caused by various brain insults including mitochondrial toxicity, but the precise neuroprotective mechanisms are unclear. Here, we studied the effects of the A2AR antagonist KW6002 on retinal injury induced by the mitochondrial oxidative phosphorylation uncoupler, carbonylcyanide m-chlorophenyl hydrazine (CCCP) and alterations in competing endogenous RNA (ceRNA) network. We found that KW6002 treatment partially reversed CCCP-induced reduction in retinal thickness and retinal ganglia cell number by increasing mitochondrial content and reducing retinal ganglia cells apoptosis. Furthermore, we employed whole-transcriptome sequencing to explore ceRNA network changes associated with CCCP-induced retinal injury and its reversal by KW6002. This analysis revealed that A2AR blockade reduced the number of CCCP-induced microRNAs by ∼60%, but increased the number of CCCP-induced circular RNAs by ∼50%. Among CeRNA network changes, CCCP-induced retinal injury was associated with a possible enrichment of the tumor necrosis factor signaling pathway and its related 126 microRNAs, 237 long non-coding RNAs, 58 circular RNAs competing. Moreover, the A2AR antagonist-mediated protection against CCCP-induced retinal injury was possibly associated with the up-regulation of mature brain-derived neurotrophic factor and its related 4 microRNAs competed by 43 long non-coding RNAs and 9 circular RNAs competing. These ceRNA network alterations by CCCP treatment and its reversal by A2AR antagonist may contribute to understanding the transcriptome mechanism for protection against CCCP-induced retinal injury by A2AR antagonists.
Collapse
Affiliation(s)
- Gengjing Fang
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yuling Zhou
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaopeng Zhou
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325000, China
| | - Hui Zhou
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325000, China
| | - Yuan-Yuan Ge
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325000, China
| | - Shengtao Luo
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325000, China
| | - Jiang-Fan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325000, China.
| | - Liping Zhang
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, The State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Optometry and Ophthalmology, Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
15
|
Flores-Toro J, Chun SK, Shin JK, Campbell J, Lichtenberger M, Chapman W, Zendejas I, Behrns K, Leeuwenburgh C, Kim JS. Critical Roles of Calpastatin in Ischemia/Reperfusion Injury in Aged Livers. Cells 2021; 10:1863. [PMID: 34440632 PMCID: PMC8394464 DOI: 10.3390/cells10081863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/02/2022] Open
Abstract
Ischemia/reperfusion (I/R) injury unavoidably occurs during hepatic resection and transplantation. Aged livers poorly tolerate I/R during surgical treatment. Although livers have a powerful endogenous inhibitor of calpains, calpastatin (CAST), I/R activates calpains, leading to impaired autophagy, mitochondrial dysfunction, and hepatocyte death. It is unknown how I/R in aged livers affects CAST. Human and mouse liver biopsies at different ages were collected during in vivo I/R. Hepatocytes were isolated from 3-month- (young) and 26-month-old (aged) mice, and challenged with short in vitro simulated I/R. Cell death, protein expression, autophagy, and mitochondrial permeability transition (MPT) between the two age groups were compared. Adenoviral vector was used to overexpress CAST. Significant cell death was observed only in reperfused aged hepatocytes. Before the commencement of ischemia, CAST expression in aged human and mouse livers and mouse hepatocytes was markedly greater than that in young counterparts. However, reperfusion substantially decreased CAST in aged human and mouse livers. In hepatocytes, reperfusion rapidly depleted aged cells of CAST, cleaved autophagy-related protein 5 (ATG5), and induced defective autophagy and MPT onset, all of which were blocked by CAST overexpression. Furthermore, mitochondrial morphology was shifted toward an elongated shape with CAST overexpression. In conclusion, CAST in aged livers is intrinsically short-lived and lost after short I/R. CAST depletion contributes to age-dependent liver injury after I/R.
Collapse
Affiliation(s)
- Joseph Flores-Toro
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; (J.F.-T.); (S.-K.C.); (I.Z.); (K.B.)
| | - Sung-Kook Chun
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; (J.F.-T.); (S.-K.C.); (I.Z.); (K.B.)
| | - Jun-Kyu Shin
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (J.-K.S.); (J.C.); (M.L.); (W.C.)
| | - Joan Campbell
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (J.-K.S.); (J.C.); (M.L.); (W.C.)
| | - Melissa Lichtenberger
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (J.-K.S.); (J.C.); (M.L.); (W.C.)
| | - William Chapman
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (J.-K.S.); (J.C.); (M.L.); (W.C.)
| | - Ivan Zendejas
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; (J.F.-T.); (S.-K.C.); (I.Z.); (K.B.)
| | - Kevin Behrns
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; (J.F.-T.); (S.-K.C.); (I.Z.); (K.B.)
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL 32610, USA;
| | - Jae-Sung Kim
- Department of Surgery, University of Florida, Gainesville, FL 32610, USA; (J.F.-T.); (S.-K.C.); (I.Z.); (K.B.)
- Department of Surgery, Washington University in St. Louis, St. Louis, MO 63110, USA; (J.-K.S.); (J.C.); (M.L.); (W.C.)
- Department of Cell Biology and Physiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
16
|
Del Dotto V, Carelli V. Dominant Optic Atrophy (DOA): Modeling the Kaleidoscopic Roles of OPA1 in Mitochondrial Homeostasis. Front Neurol 2021; 12:681326. [PMID: 34177786 PMCID: PMC8220150 DOI: 10.3389/fneur.2021.681326] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/22/2021] [Indexed: 02/05/2023] Open
Abstract
In the year 2000, the discovery of OPA1 mutations as causative for dominant optic atrophy (DOA) was pivotal to rapidly expand the field of mitochondrial dynamics and describe the complex machinery governing this pathway, with a multitude of other genes and encoded proteins involved in neurodegenerative disorders of the optic nerve. OPA1 turned out to be a much more complex protein than initially envisaged, connecting multiple pathways beyond its strict role in mitochondrial fusion, such as sensing of OXPHOS needs and mitochondrial DNA maintenance. As a consequence, an increasing need to investigate OPA1 functions at multiple levels has imposed the development of multiple tools and models that are here reviewed. Translational mitochondrial medicine, with the ultimate objective of translating basic science necessary to understand pathogenic mechanisms into therapeutic strategies, requires disease modeling at multiple levels: from the simplest, like in yeast, to cell models, including the increasing use of reprogrammed stem cells (iPSCs) from patients, to animal models. In the present review, we thoroughly examine and provide the state of the art of all these approaches.
Collapse
Affiliation(s)
- Valentina Del Dotto
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Valerio Carelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| |
Collapse
|
17
|
Maloney DM, Chadderton N, Millington-Ward S, Palfi A, Shortall C, O'Byrne JJ, Cassidy L, Keegan D, Humphries P, Kenna P, Farrar GJ. Optimized OPA1 Isoforms 1 and 7 Provide Therapeutic Benefit in Models of Mitochondrial Dysfunction. Front Neurosci 2020; 14:571479. [PMID: 33324145 PMCID: PMC7726421 DOI: 10.3389/fnins.2020.571479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/30/2020] [Indexed: 01/09/2023] Open
Abstract
Optic Atrophy 1 (OPA1) is a mitochondrially targeted GTPase that plays a pivotal role in mitochondrial health, with mutations causing severe mitochondrial dysfunction and typically associated with Dominant Optic Atrophy (DOA), a progressive blinding disease involving retinal ganglion cell loss and optic nerve damage. In the current study, we investigate the use of codon-optimized versions of OPA1 isoform 1 and 7 as potential therapeutic interventions in a range of in vitro and in vivo models of mitochondrial dysfunction. We demonstrate that both isoforms perform equally well in ameliorating mitochondrial dysfunction in OPA1 knockout mouse embryonic fibroblast cells but that OPA1 expression levels require tight regulation for optimal benefit. Of note, we demonstrate for the first time that both OPA1 isoform 1 and 7 can be used independently to protect spatial visual function in a murine model of retinal ganglion cell degeneration caused by mitochondrial dysfunction, as well as providing benefit to mitochondrial bioenergetics in DOA patient derived fibroblast cells. These results highlight the potential value of OPA1-based gene therapy interventions.
Collapse
Affiliation(s)
- Daniel M Maloney
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Naomi Chadderton
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland
| | | | - Arpad Palfi
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Ciara Shortall
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland
| | - James J O'Byrne
- National Centre for Inherited Metabolic Disorders, The Mater Misericordiae University Hospital, Dublin, Ireland.,Clinical Genetics Centre for Ophthalmology, The Mater Misericordiae University Hospital, Dublin, Ireland
| | - Lorraine Cassidy
- The Research Foundation, Royal Victoria Eye and Ear Hospital, Dublin, Ireland
| | - David Keegan
- Clinical Genetics Centre for Ophthalmology, The Mater Misericordiae University Hospital, Dublin, Ireland
| | - Peter Humphries
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Paul Kenna
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland.,The Research Foundation, Royal Victoria Eye and Ear Hospital, Dublin, Ireland
| | - Gwyneth Jane Farrar
- The School of Genetics & Microbiology, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
18
|
Pang Y, Qin M, Hu P, Ji K, Xiao R, Sun N, Pan X, Zhang X. Resveratrol protects retinal ganglion cells against ischemia induced damage by increasing Opa1 expression. Int J Mol Med 2020; 46:1707-1720. [PMID: 32901846 PMCID: PMC7521588 DOI: 10.3892/ijmm.2020.4711] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022] Open
Abstract
Loss of idiopathic retinal ganglion cells (RGCs) leads to irreversible vision defects and is considered the primary characteristic of glaucoma. However, effective treatment strategies in terms of RGC neuroprotection remain elusive. In the present study, the protective effects of resveratrol on RGC apoptosis, and the mechanisms underlying its effects were investigated, with a particular emphasis on the function of optic atrophy 1 (Opa1). In an ischemia/reperfusion (I/R) injury model, the notable thinning of the retina, significant apoptosis of RGCs, reduction in Opa1 expression and long Opa1 isoform to short Opa1 isoform ratios (L-Opa1/S-Opa1 ratio) were observed, all of which were reversed by resveratrol administration. Serum deprivation resulted in reductions in R28 cell viability, superoxide dismutase (SOD) activity, Opa1 expression and induced apoptosis, which were also partially reversed by resveratrol treatment. To conclude, results from the present study suggest that resveratrol treatment significantly reduced retinal damage and RGC apoptosis in I/R injury and serum deprivation models. In addition, resveratrol reversed the downregulated expression of Opa1 and reduced SOD activity. Mechanistically, resveratrol influenced mitochondrial dynamics by regulating the L-Opa1/S-Opa1 ratio. Therefore, these observations suggest that resveratrol may exhibit potential as a therapeutic agent for RGC damage in the future.
Collapse
Affiliation(s)
- Yulian Pang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi 330006, P.R. China
| | - Mengqi Qin
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi 330006, P.R. China
| | - Piaopiao Hu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi 330006, P.R. China
| | - Kaibao Ji
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi 330006, P.R. China
| | - Ruihan Xiao
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi 330006, P.R. China
| | - Nan Sun
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi 330006, P.R. China
| | - Xinghui Pan
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi 330006, P.R. China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Jiangxi Provincial Key Laboratory for Ophthalmology, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
19
|
Kim D, Roy S. Effects of Diabetes on Mitochondrial Morphology and Its Implications in Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2020; 61:10. [PMID: 32756920 PMCID: PMC7441301 DOI: 10.1167/iovs.61.10.10] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 06/15/2020] [Indexed: 12/17/2022] Open
Abstract
Purpose To determine whether high glucose (HG) or diabetes alters mitochondrial morphology and promotes mitochondrial fragmentation in retinal vascular cells and thereby triggers apoptosis associated with diabetic retinopathy. Methods To assess whether diabetes promotes mitochondrial fragmentation and thereby triggers apoptosis, retinas from nondiabetic and diabetic rats were analyzed using electron microscopy (EM) and in parallel, wild-type, diabetic, and OPA1+/- mice were analyzed for optic atrophy gene 1 (OPA1) and cytochrome c levels using Western blot (WB) analysis. To assess the relationship between mitochondrial fragmentation and OPA1 levels, rat retinal endothelial cells (RRECs) were grown in normal (N; 5 mmol/L) medium, HG (30 mmol/L) medium, or in N medium transfected with OPA1 siRNA for seven days. Cells were examined for OPA1 expression and cytochrome c release by WB. In parallel, cells were stained with MitoTracker Red and assessed for mitochondrial fragmentation in live cells using confocal microscopy. Results EM images revealed significant mitochondrial fragmentation in vascular cells of retinal capillaries of diabetic rats compared with that of nondiabetic rats. WB analysis showed significant OPA1 downregulation concomitant with increased levels of proapoptotic cytochrome c levels in cells grown in HG and in cells transfected with OPA1 siRNA alone. Similarly, OPA1 level was significantly reduced in diabetic retinas compared with that of nondiabetic retinas. Interestingly, OPA1+/- animals exhibited elevated cytochrome c release similar to those of diabetic mice. Conclusions Findings indicate that diabetes promotes mitochondrial fragmentation in retinal vascular cells, which are driven, at least in part, by decreased OPA1 levels leading to apoptosis in diabetic retinopathy.
Collapse
MESH Headings
- Animals
- Apoptosis
- Blotting, Western
- Cells, Cultured
- Cytochromes c/metabolism
- Diabetes Mellitus, Experimental/pathology
- Diabetic Retinopathy/metabolism
- Diabetic Retinopathy/pathology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- GTP Phosphohydrolases/genetics
- GTP Phosphohydrolases/metabolism
- Glucose/pharmacology
- Mice
- Mice, Inbred C57BL
- Microscopy, Confocal
- Microscopy, Electron
- Mitochondria/metabolism
- Mitochondria/pathology
- Mitochondrial Diseases/metabolism
- Mitochondrial Diseases/pathology
- RNA, Small Interfering/genetics
- Rats
- Rats, Sprague-Dawley
- Retinal Vessels/pathology
- Transfection
Collapse
Affiliation(s)
- Dongjoon Kim
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| | - Sayon Roy
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States
- Department of Ophthalmology, Boston University School of Medicine, Boston, Massachusetts, United States
| |
Collapse
|
20
|
Yan X, Qu X, Tian R, Xu L, Jin X, Yu S, Zhao Y, Ma J, Liu Y, Sun L, Su J. Hypoxia-induced NAD + interventions promote tumor survival and metastasis by regulating mitochondrial dynamics. Life Sci 2020; 259:118171. [PMID: 32738362 DOI: 10.1016/j.lfs.2020.118171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/16/2020] [Accepted: 07/27/2020] [Indexed: 02/08/2023]
Abstract
Hypoxia, an important feature of the tumor microenvironment, is responsible for the chemo-resistance and metastasis of malignant solid tumors. Recent studies indicated that mitochondria undergo morphological transitions as an adaptive response to maintain self-stability and connectivity under hypoxic conditions. NAD+ may not only provide reducing equivalents for biosynthetic reactions and in determining energy production, but also functions as a signaling molecule in mitochondrial dynamics regulation. In this review, we describe the upregulated KDAC deacetylase expression in the mitochondria and cytoplasm of tumor cells that results from sensing the changes in NAD+ to control mitochondrial dynamics and distribution, which is responsible for survival and metastasis in hypoxia.
Collapse
Affiliation(s)
- Xiaoyu Yan
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xianzhi Qu
- Department of Hepatobiliary & Pancreatic Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin 130021, China
| | - Rui Tian
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Long Xu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Xue Jin
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Sihang Yu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yuanxin Zhao
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jiaoyan Ma
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yanan Liu
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Liankun Sun
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| | - Jing Su
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, China.
| |
Collapse
|
21
|
Lai Y, Lin P, Chen M, Zhang Y, Chen J, Zheng M, Liu J, Du H, Chen R, Pan X, Liu N, Chen H. Restoration of L-OPA1 alleviates acute ischemic stroke injury in rats via inhibiting neuronal apoptosis and preserving mitochondrial function. Redox Biol 2020; 34:101503. [PMID: 32199783 PMCID: PMC7327985 DOI: 10.1016/j.redox.2020.101503] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/24/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ischemic stroke can induce changes in mitochondrial morphology and function. As a regulatory gene in mitochondria, optic atrophy 1 (OPA1) plays a pivotal role in the regulation of mitochondrial dynamics and other related functions. However, its roles in cerebral ischemia-related conditions are barely understood. METHODS Cultured rat primary cortical neurons were respectively transfected with OPA1-v1ΔS1-encoding and OPA1-v1-encoding lentivirus before exposure to 2-h oxygen-glucose deprivation (OGD) and subsequent reoxygenation (OGD/R). Adult male SD rats received an intracranial injection of AAV-OPA1-v1ΔS1 and were subjected to 90 min of transient middle cerebral artery occlusion (tMCAO) followed by reperfusion. OPA1 expression and function were detected by in vitro and in vivo assays. RESULTS OPA1 was excessively cleaved after cerebral ischemia/reperfusion injury, both in vitro and in vivo. Under OGD/R condition, compared with that of the LV-OPA1-v1-treated group, the expression of OPA1-v1ΔS1 efficiently restored L-OPA1 level and alleviated neuronal death and mitochondrial morphological damage. Meanwhile, the expression of OPA1-v1ΔS1 markedly improved cerebral ischemia/reperfusion-induced motor function damage, attenuated brain infarct volume, neuronal apoptosis, mitochondrial bioenergetics deficits, oxidative stress, and restored the morphology of mitochondrial cristae and mitochondrial length. It also preserved the mitochondrial integrity and reinforced the mtDNA content and expression of mitochondrial biogenesis factors in ischemic rats. INTERPRETATION Our results demonstrate that the stabilization of L-OPA1 protects ischemic brains by reducing neuronal apoptosis and preserving mitochondrial function, suggesting its significance as a promising therapeutic target for stroke prevention and treatment.
Collapse
Affiliation(s)
- Yongxing Lai
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Peiqiang Lin
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Manli Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Yixian Zhang
- Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Jianhao Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Mouwei Zheng
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Ji Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Houwei Du
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Ronghua Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Xiaodong Pan
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China
| | - Nan Liu
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China.
| | - Hongbin Chen
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Department of Rehabilitation, Fujian Medical University Union Hospital, Fuzhou, 350001, China; Institute of Cerebral Vascular Disease of Fujian Province, Fuzhou, 350001, China; Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, 350001, China.
| |
Collapse
|
22
|
Lee H, Smith SB, Sheu SS, Yoon Y. The short variant of optic atrophy 1 (OPA1) improves cell survival under oxidative stress. J Biol Chem 2020; 295:6543-6560. [PMID: 32245890 DOI: 10.1074/jbc.ra119.010983] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 03/31/2020] [Indexed: 01/23/2023] Open
Abstract
Optic atrophy 1 (OPA1) is a dynamin protein that mediates mitochondrial fusion at the inner membrane. OPA1 is also necessary for maintaining the cristae and thus essential for supporting cellular energetics. OPA1 exists as membrane-anchored long form (L-OPA1) and short form (S-OPA1) that lacks the transmembrane region and is generated by cleavage of L-OPA1. Mitochondrial dysfunction and cellular stresses activate the inner membrane-associated zinc metallopeptidase OMA1 that cleaves L-OPA1, causing S-OPA1 accumulation. The prevailing notion has been that L-OPA1 is the functional form, whereas S-OPA1 is an inactive cleavage product in mammals, and that stress-induced OPA1 cleavage causes mitochondrial fragmentation and sensitizes cells to death. However, S-OPA1 contains all functional domains of dynamin proteins, suggesting that it has a physiological role. Indeed, we recently demonstrated that S-OPA1 can maintain cristae and energetics through its GTPase activity, despite lacking fusion activity. Here, applying oxidant insult that induces OPA1 cleavage, we show that cells unable to generate S-OPA1 are more sensitive to this stress under obligatory respiratory conditions, leading to necrotic death. These findings indicate that L-OPA1 and S-OPA1 differ in maintaining mitochondrial function. Mechanistically, we found that cells that exclusively express L-OPA1 generate more superoxide and are more sensitive to Ca2+-induced mitochondrial permeability transition, suggesting that S-OPA1, and not L-OPA1, protects against cellular stress. Importantly, silencing of OMA1 expression increased oxidant-induced cell death, indicating that stress-induced OPA1 cleavage supports cell survival. Our findings suggest that S-OPA1 generation by OPA1 cleavage is a survival mechanism in stressed cells.
Collapse
Affiliation(s)
- Hakjoo Lee
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| | - Sylvia B Smith
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia 30912.,Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| | - Shey-Shing Sheu
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| |
Collapse
|
23
|
Wu W, Zhao D, Shah SZA, Zhang X, Lai M, Yang D, Wu X, Guan Z, Li J, Zhao H, Li W, Gao H, Zhou X, Qiao J, Yang L. OPA1 overexpression ameliorates mitochondrial cristae remodeling, mitochondrial dysfunction, and neuronal apoptosis in prion diseases. Cell Death Dis 2019; 10:710. [PMID: 31551424 PMCID: PMC6760175 DOI: 10.1038/s41419-019-1953-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/30/2019] [Accepted: 08/11/2019] [Indexed: 11/29/2022]
Abstract
Prion diseases caused by the cellular prion protein (PrPC) conversion into a misfolded isoform (PrPSc) are associated with multiple mitochondrial damages. We previously reported mitochondrial dynamic abnormalities and cell death in prion diseases via modulation of a variety of factors. Optic atrophy 1 (OPA1) is one of the factors that control mitochondrial fusion, mitochondrial DNA (mtDNA) maintenance, bioenergetics, and cristae integrity. In this study, we observed downregulation of OPA1 in prion disease models in vitro and in vivo, mitochondria structure damage and dysfunction, loss of mtDNA, and neuronal apoptosis. Similar mitochondria findings were seen in OPA1-silenced un-infected primary neurons. Overexpression of OPA1 not only alleviated prion-induced mitochondrial network fragmentation and mtDNA loss, decrease in intracellular ATP, increase in ADP/ATP ratio, and decrease in mitochondrial membrane potential but also protected neurons from apoptosis by suppressing the release of cytochrome c from mitochondria to cytosol and activation of the apoptotic factor, caspase 3. Our results demonstrated that overexpression of OPA1 alleviates prion-associated mitochondrial network fragmentation and cristae remodeling, mitochondrial dysfunction, mtDNA depletion, and neuronal apoptosis, suggesting that OPA1 may be a novel and effective therapeutic target for prion diseases.
Collapse
Affiliation(s)
- Wei Wu
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China.,Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Deming Zhao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Syed Zahid Ali Shah
- Department of Pathology, Faculty of Veterinary Sciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Xixi Zhang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Mengyu Lai
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Dongming Yang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiaoqian Wu
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhiling Guan
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jie Li
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Huafen Zhao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wen Li
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hongli Gao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jian Qiao
- Department of Pathophysiology, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| | - Lifeng Yang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China.
| |
Collapse
|
24
|
The Good and the Bad of Mitochondrial Breakups. Trends Cell Biol 2019; 29:888-900. [PMID: 31495461 DOI: 10.1016/j.tcb.2019.08.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022]
Abstract
Mitochondrial morphology is a crucial determinant of mitochondrial and cellular function. Opposing fusion and fission events shape the tubular mitochondrial reticulum and ensure mitochondrial transport within cells. Cellular stress and pathophysiological conditions can lead to fragmentation of the mitochondrial network, which facilitates mitophagy and is associated with cell death. However, mitochondrial shape changes are also intertwined with the cellular metabolism, and metabolic switches can induce but also result from alterations in mitochondrial morphology. Here, we discuss recent advances in the field of mitochondrial dynamics, demonstrating cell- and tissue-specific effects of mitochondrial fragmentation on cellular metabolism, cell survival, and mitochondrial quality control.
Collapse
|
25
|
Zhao H, Perkins G, Yao H, Callacondo D, Appenzeller O, Ellisman M, La Spada AR, Haddad GG. Mitochondrial dysfunction in iPSC-derived neurons of subjects with chronic mountain sickness. J Appl Physiol (1985) 2018; 125:832-840. [PMID: 29357502 PMCID: PMC6734077 DOI: 10.1152/japplphysiol.00689.2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 02/05/2023] Open
Abstract
Patients with chronic mountain sickness (CMS) suffer from hypoxemia, erythrocytosis, and numerous neurologic deficits. Here we used induced pluripotent stem cell (iPSC)-derived neurons from both CMS and non-CMS subjects to study CMS neuropathology. Using transmission electron microscopy, we report that CMS neurons have a decreased mitochondrial volume density, length, and less cristae membrane surface area. Real-time PCR confirmed a decreased mitochondrial fusion gene optic atrophy 1 (OPA1) expression. Immunoblot analysis showed an accumulation of the short isoform of OPA1 (S-OPA1) in CMS neurons, which have reduced ATP levels under normoxia and increased lactate dehydrogenase (LDH) release and caspase 3 activation after hypoxia. Improving the balance between the long isoform of OPA1 and S-OPA1 in CMS neurons increased the ATP levels and attenuated LDH release under hypoxia. Our data provide initial evidence for altered mitochondrial morphology and function in CMS neurons, and reveal increased cell death under hypoxia due in part to altered mitochondrial dynamics. NEW & NOTEWORTHY Induced pluripotent stem cell-derived neurons from chronic mountain sickness (CMS) subjects have altered mitochondrial morphology and dynamics, and increased sensitivity to hypoxic stress. Modification of OPA1 can attenuate cell death after hypoxic treatment, providing evidence that altered mitochondrial dynamics play an important role in increased vulnerability under stress in CMS neurons.
Collapse
Affiliation(s)
- Helen Zhao
- Department of Pediatrics (Respiratory Medicine), University of California San Diego , La Jolla, California
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, University of California San Diego , La Jolla, California
| | - Hang Yao
- Department of Pediatrics (Respiratory Medicine), University of California San Diego , La Jolla, California
| | - David Callacondo
- School of Medicine, Faculty of Health Sciences, Universidad Privada de Tacna, Tacna, Peru
- Instituto de Evaluación de Tecnologíasen Salud e Investigación (IETSI). EsSalud . Lima , Peru
| | - Otto Appenzeller
- New Mexico Health Enhancement and Marathon Clinics Research Foundation , Albuquerque, New Mexico
| | - Mark Ellisman
- National Center for Microscopy and Imaging Research, University of California San Diego , La Jolla, California
| | - Albert R La Spada
- Department of Pediatrics (Respiratory Medicine), University of California San Diego , La Jolla, California
- Department of Neurosciences, University of California San Diego , La Jolla, California
- Department of Cellular and Molecular Medicine, University of California San Diego , La Jolla, California
- Institute for Genomic Medicine, University of California San Diego , La Jolla, California
- Sanford Consortium for Regenerative Medicine, University of California San Diego , La Jolla, California
- The Rady Children's Hospital , San Diego, California
| | - Gabriel G Haddad
- Department of Pediatrics (Respiratory Medicine), University of California San Diego , La Jolla, California
- Department of Neurosciences, University of California San Diego , La Jolla, California
- The Rady Children's Hospital , San Diego, California
| |
Collapse
|
26
|
Del Dotto V, Mishra P, Vidoni S, Fogazza M, Maresca A, Caporali L, McCaffery JM, Cappelletti M, Baruffini E, Lenaers G, Chan D, Rugolo M, Carelli V, Zanna C. OPA1 Isoforms in the Hierarchical Organization of Mitochondrial Functions. Cell Rep 2018. [PMID: 28636943 DOI: 10.1016/j.celrep.2017.05.073] [Citation(s) in RCA: 169] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
OPA1 is a GTPase that controls mitochondrial fusion, cristae integrity, and mtDNA maintenance. In humans, eight isoforms are expressed as combinations of long and short forms, but it is unclear whether OPA1 functions are associated with specific isoforms and/or domains. To address this, we expressed each of the eight isoforms or different constructs of isoform 1 in Opa1-/- MEFs. We observed that any isoform could restore cristae structure, mtDNA abundance, and energetic efficiency independently of mitochondrial network morphology. Long forms supported mitochondrial fusion; short forms were better able to restore energetic efficiency. The complete rescue of mitochondrial network morphology required a balance of long and short forms of at least two isoforms, as shown by combinatorial isoform silencing and co-expression experiments. Thus, multiple OPA1 isoforms are required for mitochondrial dynamics, while any single isoform can support all other functions. These findings will be useful in designing gene therapies for patients with OPA1 haploinsufficiency.
Collapse
Affiliation(s)
- Valentina Del Dotto
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy; Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA; Unit of Neurology, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40139 Bologna, Italy
| | - Prashant Mishra
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Sara Vidoni
- Medical Research Council, Mitochondrial Biology Unit, Wellcome Trust, MRC Building, Cambridge CB2 0XY, UK
| | - Mario Fogazza
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Alessandra Maresca
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, 40139 Bologna, Italy
| | - Leonardo Caporali
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, 40139 Bologna, Italy
| | - J Michael McCaffery
- Integrated Imaging Center, Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Martina Cappelletti
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Enrico Baruffini
- Department of Chemical Science, Life and Environmental Sustainability, University of Parma, 43124 Parma, Italy
| | - Guy Lenaers
- PREMMi, CNRS UMR6214, INSERM U1083, Université d'Angers, 49933 Angers Cedex 9, France
| | - David Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Michela Rugolo
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy.
| | - Valerio Carelli
- Unit of Neurology, Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, 40139 Bologna, Italy; IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, 40139 Bologna, Italy.
| | - Claudia Zanna
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
27
|
OPA1: How much do we know to approach therapy? Pharmacol Res 2018; 131:199-210. [PMID: 29454676 DOI: 10.1016/j.phrs.2018.02.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 01/01/2023]
Abstract
OPA1 is a GTPase that controls several functions, such as mitochondrial dynamics and energetics, mtDNA maintenance and cristae integrity. In the last years, there have been described other cellular pathways and mechanisms involving OPA1 directly or through its interaction. All this new information, by implementing our knowledge on OPA1 is instrumental to elucidating the pathogenic mechanisms of OPA1 mutations. Indeed, these are associated with dominant optic atrophy (DOA), one of the most common inherited optic neuropathies, and with an increasing number of heterogeneous neurodegenerative disorders. In this review, we overview all recent findings on OPA1 protein functions, on its dysfunction and related clinical phenotypes, focusing on the current therapeutic options and future perspectives to treat DOA and the other associated neurological disorders due to OPA1 mutations.
Collapse
|
28
|
Eight human OPA1 isoforms, long and short: What are they for? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:263-269. [PMID: 29382469 DOI: 10.1016/j.bbabio.2018.01.005] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/31/2022]
Abstract
OPA1 is a dynamin-related GTPase that controls mitochondrial dynamics, cristae integrity, energetics and mtDNA maintenance. The exceptional complexity of this protein is determined by the presence, in humans, of eight different isoforms that, in turn, are proteolytically cleaved into combinations of membrane-anchored long forms and soluble short forms. Recent advances highlight how each OPA1 isoform is able to fulfill "essential" mitochondrial functions, whereas only some variants carry out "specialized" features. Long forms determine fusion, long or short forms alone build cristae, whereas long and short forms together tune mitochondrial morphology. These findings offer novel challenging therapeutic potential to gene therapy.
Collapse
|
29
|
Thornton C, Jones A, Nair S, Aabdien A, Mallard C, Hagberg H. Mitochondrial dynamics, mitophagy and biogenesis in neonatal hypoxic-ischaemic brain injury. FEBS Lett 2017; 592:812-830. [PMID: 29265370 DOI: 10.1002/1873-3468.12943] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/22/2017] [Accepted: 12/11/2017] [Indexed: 12/13/2022]
Abstract
Hypoxic-ischaemic encephalopathy, resulting from asphyxia during birth, affects 2-3 in every 1000 term infants and depending on severity, brings about life-changing neurological consequences or death. This hypoxic-ischaemia (HI) results in a delayed neural energy failure during which the majority of brain injury occurs. Currently, there are limited treatment options and additional therapies are urgently required. Mitochondrial dysfunction acts as a focal point in injury development in the immature brain. Not only do mitochondria become permeabilised, but recent findings implicate perturbations in mitochondrial dynamics (fission, fusion), mitophagy and biogenesis. Mitoprotective therapies may therefore offer a new avenue of intervention for babies who suffer lifelong disabilities due to birth asphyxia.
Collapse
Affiliation(s)
- Claire Thornton
- Perinatal Brain Injury Group, Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Adam Jones
- Perinatal Brain Injury Group, Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Syam Nair
- Perinatal Center, Department of Physiology, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Afra Aabdien
- Perinatal Brain Injury Group, Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Carina Mallard
- Perinatal Center, Department of Physiology, Institute of Physiology and Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Henrik Hagberg
- Perinatal Brain Injury Group, Division of Imaging Sciences and Biomedical Engineering, Centre for the Developing Brain, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK.,Perinatal Center, Department of Clinical Sciences & Physiology and Neuroscience, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
30
|
Lv C, Yuan X, Zeng HW, Liu RH, Zhang WD. Protective effect of cinnamaldehyde against glutamate-induced oxidative stress and apoptosis in PC12 cells. Eur J Pharmacol 2017; 815:487-494. [DOI: 10.1016/j.ejphar.2017.09.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 09/01/2017] [Accepted: 09/08/2017] [Indexed: 02/07/2023]
|
31
|
Seo MJ, Hong JM, Kim SJ, Lee SM. Genipin protects d -galactosamine and lipopolysaccharide-induced hepatic injury through suppression of the necroptosis-mediated inflammasome signaling. Eur J Pharmacol 2017; 812:128-137. [DOI: 10.1016/j.ejphar.2017.07.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/08/2017] [Accepted: 07/10/2017] [Indexed: 01/17/2023]
|
32
|
Kane MS, Alban J, Desquiret‐Dumas V, Gueguen N, Ishak L, Ferre M, Amati‐Bonneau P, Procaccio V, Bonneau D, Lenaers G, Reynier P, Chevrollier A. Autophagy controls the pathogenicity of OPA1 mutations in dominant optic atrophy. J Cell Mol Med 2017; 21:2284-2297. [PMID: 28378518 PMCID: PMC5618673 DOI: 10.1111/jcmm.13149] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 02/02/2017] [Indexed: 12/31/2022] Open
Abstract
Optic Atrophy 1 (OPA1) gene mutations cause diseases ranging from isolated dominant optic atrophy (DOA) to various multisystemic disorders. OPA1, a large GTPase belonging to the dynamin family, is involved in mitochondrial network dynamics. The majority of OPA1 mutations encodes truncated forms of the protein and causes DOA through haploinsufficiency, whereas missense OPA1 mutations are predicted to cause disease through deleterious dominant-negative mechanisms. We used 3D imaging and biochemical analysis to explore autophagy and mitophagy in fibroblasts from seven patients harbouring OPA1 mutations. We report new genotype-phenotype correlations between various types of OPA1 mutation and mitophagy. Fibroblasts bearing dominant-negative OPA1 mutations showed increased autophagy and mitophagy in response to uncoupled oxidative phosphorylation. In contrast, OPA1 haploinsufficiency was correlated with a substantial reduction in mitochondrial turnover and autophagy, unless subjected to experimental mitochondrial injury. Our results indicate distinct alterations of mitochondrial physiology and turnover in cells with OPA1 mutations, suggesting that the level and profile of OPA1 may regulate the rate of mitophagy.
Collapse
Affiliation(s)
- Mariame Selma Kane
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
| | - Jennifer Alban
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
| | - Valérie Desquiret‐Dumas
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
- Département de Biochimie et GénétiqueCentre Hospitalier UniversitaireAngersFrance
| | - Naïg Gueguen
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
- Département de Biochimie et GénétiqueCentre Hospitalier UniversitaireAngersFrance
| | - Layal Ishak
- RGM4645 Université Blaise PascalAubièreFrance
| | - Marc Ferre
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
| | - Patrizia Amati‐Bonneau
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
- Département de Biochimie et GénétiqueCentre Hospitalier UniversitaireAngersFrance
| | - Vincent Procaccio
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
- Département de Biochimie et GénétiqueCentre Hospitalier UniversitaireAngersFrance
| | - Dominique Bonneau
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
- Département de Biochimie et GénétiqueCentre Hospitalier UniversitaireAngersFrance
| | - Guy Lenaers
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
| | - Pascal Reynier
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
- Département de Biochimie et GénétiqueCentre Hospitalier UniversitaireAngersFrance
| | - Arnaud Chevrollier
- PREMMi/Mitochondrial Medicine Research CentreInstitut MITOVASCCNRS UMR 6015INSERM U1083Université d'Angers, CHU d'AngersAngersFrance
| |
Collapse
|
33
|
Shin JK, Lee SM. Genipin protects the liver from ischemia/reperfusion injury by modulating mitochondrial quality control. Toxicol Appl Pharmacol 2017; 328:25-33. [DOI: 10.1016/j.taap.2017.05.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 04/21/2017] [Accepted: 05/02/2017] [Indexed: 12/26/2022]
|
34
|
Chen H, Wang L, Wang W, Cheng C, Zhang Y, Zhou Y, Wang C, Miao X, Wang J, Wang C, Li J, Zheng L, Huang K. ELABELA and an ELABELA Fragment Protect against AKI. J Am Soc Nephrol 2017; 28:2694-2707. [PMID: 28583915 DOI: 10.1681/asn.2016111210] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/09/2017] [Indexed: 12/26/2022] Open
Abstract
Renal ischemia-reperfusion (I/R) injury is the most common cause of AKI, which associates with high mortality and has no effective therapy. ELABELA (ELA) is a newly identified 32-residue hormone peptide highly expressed in adult kidney. To investigate whether ELA has protective effects on renal I/R injury, we administered the mature peptide (ELA32) or the 11-residue furin-cleaved fragment (ELA11) to hypoxia-reperfusion (H/R)-injured or adriamycin-treated renal tubular cells in vitro ELA32 and ELA11 significantly inhibited the elevation of the DNA damage response, apoptosis, and inflammation in H/R-injured renal tubular cells and suppressed adriamycin-induced DNA damage response. Similarly, overexpression of ELA32 or ELA11 significantly inhibited H/R-induced cell death, DNA damage response, and inflammation. Notably, treatment of mice with ELA32 or ELA11 but not an ELA11 mutant with a cysteine to alanine substitution at the N terminus (AE11C) inhibited I/R injury-induced renal fibrosis, inflammation, apoptosis, and the DNA damage response and markedly reduced the renal tubular lesions and renal dysfunction. Together, our results suggest that ELA32 and ELA11 may be therapeutic candidates for treating AKI.
Collapse
Affiliation(s)
| | - Lin Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Wenjun Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; and
| | | | | | - Yu Zhou
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri
| | - Congyi Wang
- The Center for Biomedical Research, Tongji Hospital, and
| | - Xiaoping Miao
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | - Jianshuang Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; and
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China; and
| | | |
Collapse
|
35
|
Wang W, Wang Q, Wan D, Sun Y, Wang L, Chen H, Liu C, Petersen RB, Li J, Xue W, Zheng L, Huang K. Histone HIST1H1C/H1.2 regulates autophagy in the development of diabetic retinopathy. Autophagy 2017; 13:941-954. [PMID: 28409999 DOI: 10.1080/15548627.2017.1293768] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Autophagy plays critical and complex roles in many human diseases, including diabetes and its complications. However, the role of autophagy in the development of diabetic retinopathy remains uncertain. Core histone modifications have been reported involved in the development of diabetic retinopathy, but little is known about the histone variants. Here, we observed increased autophagy and histone HIST1H1C/H1.2, an important variant of the linker histone H1, in the retinas of type 1 diabetic rodents. Overexpression of histone HIST1H1C upregulates SIRT1 and HDAC1 to maintain the deacetylation status of H4K16, leads to upregulation of ATG proteins, then promotes autophagy in cultured retinal cell line. Histone HIST1H1C overexpression also promotes inflammation and cell toxicity in vitro. Knockdown of histone HIST1H1C reduces both the basal and stresses (including high glucose)-induced autophagy, and inhibits high glucose induced inflammation and cell toxicity. Importantly, AAV-mediated histone HIST1H1C overexpression in the retinas leads to increased autophagy, inflammation, glial activation and neuron loss, similar to the pathological changes identified in the early stage of diabetic retinopathy. Furthermore, knockdown of histone Hist1h1c by siRNA in the retinas of diabetic mice significantly attenuated the diabetes-induced autophagy, inflammation, glial activation and neuron loss. These results indicate that histone HIST1H1C may offer a novel therapeutic target for preventing diabetic retinopathy.
Collapse
Affiliation(s)
- Wenjun Wang
- a Hubei Key Laboratory of Cell Homeostasis , College of Life Sciences, Wuhan University , Wuhan , Hubei , China
| | - Qing Wang
- a Hubei Key Laboratory of Cell Homeostasis , College of Life Sciences, Wuhan University , Wuhan , Hubei , China
| | - Danyang Wan
- a Hubei Key Laboratory of Cell Homeostasis , College of Life Sciences, Wuhan University , Wuhan , Hubei , China
| | - Yue Sun
- a Hubei Key Laboratory of Cell Homeostasis , College of Life Sciences, Wuhan University , Wuhan , Hubei , China
| | - Lin Wang
- a Hubei Key Laboratory of Cell Homeostasis , College of Life Sciences, Wuhan University , Wuhan , Hubei , China
| | - Hong Chen
- b Tongji School of Pharmacy , Huazhong University of Science and Technology , Wuhan , Hubei , China
| | - Chengyu Liu
- b Tongji School of Pharmacy , Huazhong University of Science and Technology , Wuhan , Hubei , China
| | - Robert B Petersen
- c Departments of Pathology , Neuroscience, and Neurology, Case Western Reserve University , Cleveland , OH , USA
| | - Jianshuang Li
- a Hubei Key Laboratory of Cell Homeostasis , College of Life Sciences, Wuhan University , Wuhan , Hubei , China
| | - Weili Xue
- a Hubei Key Laboratory of Cell Homeostasis , College of Life Sciences, Wuhan University , Wuhan , Hubei , China
| | - Ling Zheng
- a Hubei Key Laboratory of Cell Homeostasis , College of Life Sciences, Wuhan University , Wuhan , Hubei , China
| | - Kun Huang
- b Tongji School of Pharmacy , Huazhong University of Science and Technology , Wuhan , Hubei , China.,d Centre for Biomedicine Research , Wuhan Institute of Biotechnology , Wuhan , China
| |
Collapse
|
36
|
MacVicar T, Langer T. OPA1 processing in cell death and disease - the long and short of it. J Cell Sci 2016; 129:2297-306. [PMID: 27189080 DOI: 10.1242/jcs.159186] [Citation(s) in RCA: 256] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The regulation of mitochondrial dynamics by the GTPase OPA1, which is located at the inner mitochondrial membrane, is crucial for adapting mitochondrial function and preserving cellular health. OPA1 governs the delicate balance between fusion and fission in the dynamic mitochondrial network. A disturbance of this balance, often observed under stress and pathologic conditions, causes mitochondrial fragmentation and can ultimately result in cell death. As discussed in this Commentary, these morphological changes are regulated by proteolytic processing of OPA1 by the inner-membrane peptidases YME1L (also known as YME1L1) and OMA1. Long, membrane-bound forms of OPA1 are required for mitochondrial fusion, but their processing to short, soluble forms limits fusion and can facilitate mitochondrial fission. Excessive OPA1 processing by the stress-activated protease OMA1 promotes mitochondrial fragmentation and, if persistent, triggers cell death and tissue degeneration in vivo The prevention of OMA1-mediated OPA1 processing and mitochondrial fragmentation might thus offer exciting therapeutic potential for human diseases associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Thomas MacVicar
- Institute of Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| | - Thomas Langer
- Institute of Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| |
Collapse
|
37
|
Millet AMC, Bertholet AM, Daloyau M, Reynier P, Galinier A, Devin A, Wissinguer B, Belenguer P, Davezac N. Loss of functional OPA1 unbalances redox state: implications in dominant optic atrophy pathogenesis. Ann Clin Transl Neurol 2016; 3:408-21. [PMID: 27547769 PMCID: PMC4891995 DOI: 10.1002/acn3.305] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 03/04/2016] [Accepted: 03/05/2016] [Indexed: 12/14/2022] Open
Abstract
Objective OPA1 mutations cause protein haploinsufficiency leading to dominant optic atrophy (DOA), an incurable retinopathy with variable severity. Up to 20% of patients also develop extraocular neurological complications. The mechanisms that cause this optic atrophy or its syndromic forms are still unknown. After identifying oxidative stress in a mouse model of the pathology, we sought to determine the consequences of OPA1 dysfunction on redox homeostasis. Methods Mitochondrial respiration, reactive oxygen species levels, antioxidant defenses, and cell death were characterized by biochemical and in situ approaches in both in vitro and in vivo models of OPA1 haploinsufficiency. Results A decrease in aconitase activity suggesting an increase in reactive oxygene species and an induction of antioxidant defenses was observed in cortices of a murine model as well as in OPA1 downregulated cortical neurons. This increase is associated with a decline in mitochondrial respiration in vitro. Upon exogenous oxidative stress, OPA1‐depleted neurons did not further exhibit upregulated antioxidant defenses but were more sensitive to cell death. Finally, low levels of antioxidant enzymes were found in fibroblasts from patients supporting their role as modifier factors. Interpretation Our study suggests that the pro‐oxidative state induced by OPA1 loss may contribute to DOA pathogenesis and that differences in antioxidant defenses can explain the variability in expressivity. Furthermore, antioxidants may be used as therapy as they could prevent or delay DOA symptoms in patients.
Collapse
Affiliation(s)
- Aurélie M C Millet
- Center of Developmental Biology (CBD)/Research Center on Animal Cognition (CRCA) Center for Integrative Biology (CBI) Toulouse University, CNRS, UPS Toulouse France
| | - Ambre M Bertholet
- Center of Developmental Biology (CBD)/Research Center on Animal Cognition (CRCA) Center for Integrative Biology (CBI) Toulouse University, CNRS, UPS Toulouse France
| | - Marlène Daloyau
- Center of Developmental Biology (CBD)/Research Center on Animal Cognition (CRCA) Center for Integrative Biology (CBI) Toulouse University, CNRS, UPS Toulouse France
| | - Pascal Reynier
- CNRS UMR 6214 Inserm UMR 1083 UFR Sciences médicales Rue Haute de Reculee Angers Cedex 01 49045 France
| | - Anne Galinier
- Laboratoire de Biochimie Nutritionnelle "STROMALab" UMR UPS/CNRS/EFS 5273 Inserm U1031, CHU Rangueil 1 avenue Jean Poulhès Toulouse Cedex 9 31059 France
| | - Anne Devin
- Laboratoire métabolisme énergétique cellulaire IBGC du CNRS 1 rue Camille Saint Saëns Bordeaux Cedex 33077 France
| | - Bernd Wissinguer
- Centre for Ophthalmology University of Tübingen Roentgenweg 11 Tübingen D-72076 Germany
| | - Pascale Belenguer
- Center of Developmental Biology (CBD)/Research Center on Animal Cognition (CRCA) Center for Integrative Biology (CBI) Toulouse University, CNRS, UPS Toulouse France
| | - Noélie Davezac
- Center of Developmental Biology (CBD)/Research Center on Animal Cognition (CRCA) Center for Integrative Biology (CBI) Toulouse University, CNRS, UPS Toulouse France
| |
Collapse
|