1
|
Samanta S, Akhter F, Roy A, Chen D, Turner B, Wang Y, Clemente N, Wang C, Swerdlow RH, Battaile KP, Lovell S, Yan SF, Yan SS. New cyclophilin D inhibitor rescues mitochondrial and cognitive function in Alzheimer's disease. Brain 2024; 147:1710-1725. [PMID: 38146639 PMCID: PMC11484516 DOI: 10.1093/brain/awad432] [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: 11/25/2022] [Revised: 11/16/2023] [Accepted: 12/05/2023] [Indexed: 12/27/2023] Open
Abstract
Mitochondrial dysfunction is an early pathological feature of Alzheimer disease and plays a crucial role in the development and progression of Alzheimer's disease. Strategies to rescue mitochondrial function and cognition remain to be explored. Cyclophilin D (CypD), the peptidylprolyl isomerase F (PPIase), is a key component in opening the mitochondrial membrane permeability transition pore, leading to mitochondrial dysfunction and cell death. Blocking membrane permeability transition pore opening by inhibiting CypD activity is a promising therapeutic approach for Alzheimer's disease. However, there is currently no effective CypD inhibitor for Alzheimer's disease, with previous candidates demonstrating high toxicity, poor ability to cross the blood-brain barrier, compromised biocompatibility and low selectivity. Here, we report a new class of non-toxic and biocompatible CypD inhibitor, ebselen, using a conventional PPIase assay to screen a library of ∼2000 FDA-approved drugs with crystallographic analysis of the CypD-ebselen crystal structure (PDB code: 8EJX). More importantly, we assessed the effects of genetic and pharmacological blockade of CypD on Alzheimer's disease mitochondrial and glycolytic bioenergetics in Alzheimer's disease-derived mitochondrial cybrid cells, an ex vivo human sporadic Alzheimer's disease mitochondrial model, and on synaptic function, inflammatory response and learning and memory in Alzheimer's disease mouse models. Inhibition of CypD by ebselen protects against sporadic Alzheimer's disease- and amyloid-β-induced mitochondrial and glycolytic perturbation, synaptic and cognitive dysfunction, together with suppressing neuroinflammation in the brain of Alzheimer's disease mouse models, which is linked to CypD-related membrane permeability transition pore formation. Thus, CypD inhibitors have the potential to slow the progression of neurodegenerative diseases, including Alzheimer's disease, by boosting mitochondrial bioenergetics and improving synaptic and cognitive function.
Collapse
Affiliation(s)
- Sourav Samanta
- Division of Surgical Science of Department of Surgery, Columbia University in New York, New York, NY 10032, USA
| | - Firoz Akhter
- Division of Surgical Science of Department of Surgery, Columbia University in New York, New York, NY 10032, USA
| | - Anuradha Roy
- High Throughput Screening Laboratory, Del M. Shankel Structural Biology Center, University of Kansas, Lawrence, KS 66047, USA
| | - Doris Chen
- Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA
| | - Benjamin Turner
- High Throughput Screening Laboratory, Del M. Shankel Structural Biology Center, University of Kansas, Lawrence, KS 66047, USA
| | - Yongfu Wang
- Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA
| | - Nicolina Clemente
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, New York, NY 12180-3590, USA
| | - Chunyu Wang
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, New York, NY 12180-3590, USA
| | | | - Kevin P Battaile
- New York Structural Biology Center, NSLS-II, Upton, NY 11973, USA
| | - Scott Lovell
- Protein Structure and X-Ray Crystallography Laboratory, The University of Kansas, Lawrence, KS 66047, USA
| | - Shi Fang Yan
- Division of Surgical Science of Department of Surgery, Columbia University in New York, New York, NY 10032, USA
| | - Shirley ShiDu Yan
- Division of Surgical Science of Department of Surgery, Columbia University in New York, New York, NY 10032, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, NY 10032, USA
| |
Collapse
|
2
|
Baev AY, Vinokurov AY, Potapova EV, Dunaev AV, Angelova PR, Abramov AY. Mitochondrial Permeability Transition, Cell Death and Neurodegeneration. Cells 2024; 13:648. [PMID: 38607087 PMCID: PMC11011324 DOI: 10.3390/cells13070648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 03/27/2024] [Accepted: 04/06/2024] [Indexed: 04/13/2024] Open
Abstract
Neurodegenerative diseases are chronic conditions occurring when neurons die in specific brain regions that lead to loss of movement or cognitive functions. Despite the progress in understanding the mechanisms of this pathology, currently no cure exists to treat these types of diseases: for some of them the only help is alleviating the associated symptoms. Mitochondrial dysfunction has been shown to be involved in the pathogenesis of most the neurodegenerative disorders. The fast and transient permeability of mitochondria (the mitochondrial permeability transition, mPT) has been shown to be an initial step in the mechanism of apoptotic and necrotic cell death, which acts as a regulator of tissue regeneration for postmitotic neurons as it leads to the irreparable loss of cells and cell function. In this study, we review the role of the mitochondrial permeability transition in neuronal death in major neurodegenerative diseases, covering the inductors of mPTP opening in neurons, including the major ones-free radicals and calcium-and we discuss perspectives and difficulties in the development of a neuroprotective strategy based on the inhibition of mPTP in neurodegenerative disorders.
Collapse
Affiliation(s)
- Artyom Y. Baev
- Laboratory of Experimental Biophysics, Centre for Advanced Technologies, Tashkent 100174, Uzbekistan;
- Department of Biophysics, Faculty of Biology, National University of Uzbekistan, Tashkent 100174, Uzbekistan
| | - Andrey Y. Vinokurov
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (A.Y.V.); (E.V.P.); (A.V.D.)
| | - Elena V. Potapova
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (A.Y.V.); (E.V.P.); (A.V.D.)
| | - Andrey V. Dunaev
- Cell Physiology and Pathology Laboratory, Orel State University, Orel 302026, Russia; (A.Y.V.); (E.V.P.); (A.V.D.)
| | - Plamena R. Angelova
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK;
| | - Andrey Y. Abramov
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, UK;
| |
Collapse
|
3
|
Rahman MM, Tumpa MAA, Rahaman MS, Islam F, Sutradhar PR, Ahmed M, Alghamdi BS, Hafeez A, Alexiou A, Perveen A, Ashraf GM. Emerging Promise of Therapeutic Approaches Targeting Mitochondria in Neurodegenerative Disorders. Curr Neuropharmacol 2023; 21:1081-1099. [PMID: 36927428 PMCID: PMC10286587 DOI: 10.2174/1570159x21666230316150559] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 12/05/2022] [Accepted: 12/06/2022] [Indexed: 03/18/2023] Open
Abstract
Mitochondria are critical for homeostasis and metabolism in all cellular eukaryotes. Brain mitochondria are the primary source of fuel that supports many brain functions, including intracellular energy supply, cellular calcium regulation, regulation of limited cellular oxidative capacity, and control of cell death. Much evidence suggests that mitochondria play a central role in neurodegenerative disorders (NDDs) such as Parkinson's disease, Alzheimer's disease, Huntington's disease, and amyotrophic lateral sclerosis. Ongoing studies of NDDs have revealed that mitochondrial pathology is mainly found in inherited or irregular NDDs and is thought to be associated with the pathophysiological cycle of these disorders. Typical mitochondrial disturbances in NDDs include increased free radical production, decreased ATP synthesis, alterations in mitochondrial permeability, and mitochondrial DNA damage. The main objective of this review is to highlight the basic mitochondrial problems that occur in NDDs and discuss the use mitochondrial drugs, especially mitochondrial antioxidants, mitochondrial permeability transition blockade, and mitochondrial gene therapy, for the treatment and control of NDDs.
Collapse
Affiliation(s)
- Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Mst. Afroza Alam Tumpa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. Saidur Rahaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Popy Rani Sutradhar
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Muniruddin Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Badrah S. Alghamdi
- Department of Physiology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- The Neuroscience Research Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdul Hafeez
- Glocal School of Pharmacy, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
- AFNP Med Austria, Wien, Austria
| | - Asma Perveen
- Glocal School of Life Sciences, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh, India
| | - Ghulam Md. Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates
| |
Collapse
|
4
|
Uchino H, Ogihara Y, Fukui H, Chijiiwa M, Sekine S, Hara N, Elmér E. Brain injury following cardiac arrest: pathophysiology for neurocritical care. J Intensive Care 2016; 4:31. [PMID: 27123307 PMCID: PMC4847238 DOI: 10.1186/s40560-016-0140-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 02/04/2016] [Indexed: 11/27/2022] Open
Abstract
Cardiac arrest induces the cessation of cerebral blood flow, which can result in brain damage. The primary intervention to salvage the brain under such a pathological condition is to restore the cerebral blood flow to the ischemic region. Ischemia is defined as a reduction in blood flow to a level that is sufficient to alter normal cellular function. Brain tissue is highly sensitive to ischemia, such that even brief ischemic periods in neurons can initiate a complex sequence of events that may ultimately culminate in cell death. However, paradoxically, restoration of blood flow can cause additional damage and exacerbate the neurocognitive deficits in patients who suffered a brain ischemic event, which is a phenomenon referred to as “reperfusion injury.” Transient brain ischemia following cardiac arrest results from the complex interplay of multiple pathways including excitotoxicity, acidotoxicity, ionic imbalance, peri-infarct depolarization, oxidative and nitrative stress, inflammation, and apoptosis. The pathophysiology of post-cardiac arrest brain injury involves a complex cascade of molecular events, most of which remain unknown. Many lines of evidence have shown that mitochondria suffer severe damage in response to ischemic injury. Mitochondrial dysfunction based on the mitochondrial permeability transition after reperfusion, particularly involving the calcineurin/immunophilin signal transduction pathway, appears to play a pivotal role in the induction of neuronal cell death. The aim of this article is to discuss the underlying pathophysiology of brain damage, which is a devastating pathological condition, and highlight the central signal transduction pathway involved in brain damage, which reveals potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Hiroyuki Uchino
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023 Japan
| | - Yukihiko Ogihara
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023 Japan
| | - Hidekimi Fukui
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023 Japan
| | - Miyuki Chijiiwa
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023 Japan
| | - Shusuke Sekine
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023 Japan
| | - Naomi Hara
- Department of Anesthesiology, Tokyo Medical University, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023 Japan
| | - Eskil Elmér
- Mitochondrial Pathophysiology Unit, Department of Clinical Sciences, Lund University, Box 117, 221 00 Lund, Sweden
| |
Collapse
|
5
|
Chen Q, Wang K, Jiang D, Wang Y, Xiao X, Zhu N, Li M, Jia S, Wang Y. Blocking mPTP on Neural Stem Cells and Activating the Nicotinic Acetylcholine Receptor α7 Subunit on Microglia Attenuate Aβ-Induced Neurotoxicity on Neural Stem Cells. Neurochem Res 2016; 41:1483-95. [PMID: 26875732 DOI: 10.1007/s11064-016-1862-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 02/01/2016] [Accepted: 02/03/2016] [Indexed: 12/31/2022]
Abstract
β-Amyloid (Aβ) can stimulate microglia to release a variety of proinflammatory cytokines and induce neurotoxicity. Nicotine has been reported to inhibit TNF-α, IL-1, and ROS production in microglia. Mitochondrial permeability transition pore (mPTP) plays an important role in neurotoxicity as well. Here, we investigated whether activating the microglial α7-nAChR has a neuroprotective role on neural stem cells (NSCs) and the function of mPTP in NSCs in this process. The expression of α7-nAChR in rat NSCs was detected by immunocytochemistry and RT-PCR. The viability of microglia and NSCs was examined by MTT assay. The mitochondrial membrane potential (ΔΨm) and morphological characteristics of NSCs was measured by JC-1 staining and transmission electron microscopy respectively. The distribution of cytochrome c in the subcellular regions of NSCs was visualized by confocal laser scanning microscopy, and the expression levels of cyclophilin D and cleaved caspase-3 were assayed by western blot. The apoptotic rate of NSCs was measured by flow cytometry. The expression of α7-nAChR was detected in microglial cells, but no expression was found in NSCs. The viability of rat microglial cells and NSCs was not affected by reagents or coculture itself. Aβ1-42-mediated microglial activation impaired the morphology and the ΔΨm of mitochondria of NSCs as well as increased cell apoptosis. However, the damage was attenuated when the α7-nAChRs on microglial cells were activated or the mPTPs on NSCs were blocked. Blockade of mPTPs on NSCs and activation of α7-nAChRs on microglia exhibit neuroprotective roles in Aβ-induced neurotoxicity of NSCs.
Collapse
Affiliation(s)
- Qingzhuang Chen
- Department of Pharmacy, Southern Medical University, Zhujiang Hospital, Guangzhou, China.,Department of Clinical Pharmacy, Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou, China
| | - Kewan Wang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Deqi Jiang
- Department of Pharmacy, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Yan Wang
- Department of Pharmacy, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Xiaodan Xiao
- Department of Pharmacy, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Ning Zhu
- Department of Pharmacy, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Mingxing Li
- Department of Pharmacy, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Siyuan Jia
- Department of Pharmacy, Southern Medical University, Zhujiang Hospital, Guangzhou, China
| | - Yong Wang
- Department of Pharmacy, Southern Medical University, Zhujiang Hospital, Guangzhou, China.
| |
Collapse
|
6
|
Kim DY, Simeone KA, Simeone TA, Pandya JD, Wilke JC, Ahn Y, Geddes JW, Sullivan PG, Rho JM. Ketone bodies mediate antiseizure effects through mitochondrial permeability transition. Ann Neurol 2015; 78:77-87. [PMID: 25899847 DOI: 10.1002/ana.24424] [Citation(s) in RCA: 128] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 04/03/2015] [Accepted: 04/03/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Ketone bodies (KB) are products of fatty acid oxidation and serve as essential fuels during fasting or treatment with the high-fat antiseizure ketogenic diet (KD). Despite growing evidence that KB exert broad neuroprotective effects, their role in seizure control has not been firmly demonstrated. The major goal of this study was to demonstrate the direct antiseizure effects of KB and to identify an underlying target mechanism. METHODS We studied the effects of both the KD and KB in spontaneously epileptic Kcna1-null mice using a combination of behavioral, planar multielectrode, and standard cellular electrophysiological techniques. Thresholds for mitochondrial permeability transition (mPT) were determined in acutely isolated brain mitochondria. RESULTS KB alone were sufficient to: (1) exert antiseizure effects in Kcna1-null mice, (2) restore intrinsic impairment of hippocampal long-term potentiation and spatial learning-memory defects in Kcna1-null mutants, and (3) raise the threshold for calcium-induced mPT in acutely prepared mitochondria from hippocampi of Kcna1-null animals. Targeted deletion of the cyclophilin D subunit of the mPT complex abrogated the effects of KB on mPT, and in vivo pharmacological inhibition and activation of mPT were found to mirror and reverse, respectively, the antiseizure effects of the KD in Kcna1-null mice. INTERPRETATION The present data reveal the first direct link between mPT and seizure control, and provide a potential mechanistic explanation for the KD. Given that mPT is increasingly being implicated in diverse neurological disorders, our results suggest that metabolism-based treatments and/or metabolic substrates might represent a worthy paradigm for therapeutic development.
Collapse
Affiliation(s)
- Do Young Kim
- Barrow Neurological Institute, St Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Kristina A Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE
| | - Timothy A Simeone
- Department of Pharmacology, Creighton University School of Medicine, Omaha, NE
| | - Jignesh D Pandya
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY
| | - Julianne C Wilke
- Barrow Neurological Institute, St Joseph's Hospital and Medical Center, Phoenix, AZ
| | - Younghee Ahn
- Departments of Pediatrics and Clinical Neurosciences, Alberta Children's Hospital Research Institute for Child and Maternal Health, University of Calgary Faculty of Medicine, Calgary, Alberta, Canada
| | - James W Geddes
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY
| | - Patrick G Sullivan
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY
| | - Jong M Rho
- Departments of Pediatrics and Clinical Neurosciences, Alberta Children's Hospital Research Institute for Child and Maternal Health, University of Calgary Faculty of Medicine, Calgary, Alberta, Canada
| |
Collapse
|
7
|
Bramlett HM, Dietrich WD. Long-Term Consequences of Traumatic Brain Injury: Current Status of Potential Mechanisms of Injury and Neurological Outcomes. J Neurotrauma 2014; 32:1834-48. [PMID: 25158206 DOI: 10.1089/neu.2014.3352] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Traumatic brain injury (TBI) is a significant clinical problem with few therapeutic interventions successfully translated to the clinic. Increased importance on the progressive, long-term consequences of TBI have been emphasized, both in the experimental and clinical literature. Thus, there is a need for a better understanding of the chronic consequences of TBI, with the ultimate goal of developing novel therapeutic interventions to treat the devastating consequences of brain injury. In models of mild, moderate, and severe TBI, histopathological and behavioral studies have emphasized the progressive nature of the initial traumatic insult and the involvement of multiple pathophysiological mechanisms, including sustained injury cascades leading to prolonged motor and cognitive deficits. Recently, the increased incidence in age-dependent neurodegenerative diseases in this patient population has also been emphasized. Pathomechanisms felt to be active in the acute and long-term consequences of TBI include excitotoxicity, apoptosis, inflammatory events, seizures, demyelination, white matter pathology, as well as decreased neurogenesis. The current article will review many of these pathophysiological mechanisms that may be important targets for limiting the chronic consequences of TBI.
Collapse
Affiliation(s)
- Helen M Bramlett
- The Miami Project to Cure Paralysis/Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida
| | - W Dalton Dietrich
- The Miami Project to Cure Paralysis/Department of Neurological Surgery, University of Miami Miller School of Medicine , Miami, Florida
| |
Collapse
|
8
|
Kim SY, Shim MS, Kim KY, Weinreb RN, Wheeler LA, Ju WK. Inhibition of cyclophilin D by cyclosporin A promotes retinal ganglion cell survival by preventing mitochondrial alteration in ischemic injury. Cell Death Dis 2014; 5:e1105. [PMID: 24603333 PMCID: PMC3973219 DOI: 10.1038/cddis.2014.80] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 01/22/2014] [Accepted: 01/30/2014] [Indexed: 01/10/2023]
Abstract
Cyclosporin A (CsA) inhibits the opening of the mitochondrial permeability transition pore (MPTP) by interacting with cyclophilin D (CypD) and ameliorates neuronal cell death in the central nervous system against ischemic injury. However, the molecular mechanisms underlying CypD/MPTP opening-mediated cell death in ischemic retinal injury induced by acute intraocular pressure (IOP) elevation remain unknown. We observed the first direct evidence that acute IOP elevation significantly upregulated CypD protein expression in ischemic retina at 12 h. However, CsA prevented the upregulation of CypD protein expression and promoted retinal ganglion cell (RGC) survival against ischemic injury. Moreover, CsA blocked apoptotic cell death by decreasing cleaved caspase-3 protein expression in ischemic retina. Of interest, although the expression level of Bcl-xL protein did not show a significant change in ischemic retina treated with vehicle or CsA at 12 h, ischemic damage induced the reduction of Bcl-xL immunoreactivity in RGCs. More importantly, CsA preserved Bcl-xL immunoreactivity in RGCs of ischemic retina. In parallel, acute IOP elevation significantly increased phosphorylated Bad (pBad) at Ser112 protein expression in ischemic retina at 12 h. However, CsA significantly preserved pBad protein expression in ischemic retina. Finally, acute IOP elevation significantly increased mitochondrial transcription factor A (Tfam) protein expression in ischemic retina at 12 h. However, CsA significantly preserved Tfam protein expression in ischemic retina. Studies on mitochondrial DNA (mtDNA) content in ischemic retina showed that there were no statistically significant differences in mtDNA content among control and ischemic groups treated with vehicle or CsA. Therefore, these results provide evidence that the activation of CypD-mediated MPTP opening is associated with the apoptotic pathway and the mitochondrial alteration in RGC death of ischemic retinal injury. On the basis of these observations, our findings suggest that CsA-mediated CypD inhibition may provide a promising therapeutic potential for protecting RGCs against ischemic injury-mediated mitochondrial dysfunction.
Collapse
Affiliation(s)
- S Y Kim
- Laboratory for Optic Nerve Biology, Department of Ophthalmology, Hamilton Glaucoma Center, University of California San Diego, La Jolla, CA, USA
| | - M S Shim
- Laboratory for Optic Nerve Biology, Department of Ophthalmology, Hamilton Glaucoma Center, University of California San Diego, La Jolla, CA, USA
| | - K-Y Kim
- Center for Research on Biological Systems, National Center for Microscopy and Imaging Research and Department of Neuroscience, University of California San Diego, La Jolla, CA, USA
| | - R N Weinreb
- Laboratory for Optic Nerve Biology, Department of Ophthalmology, Hamilton Glaucoma Center, University of California San Diego, La Jolla, CA, USA
| | - L A Wheeler
- Department of Biological Sciences, Allergan Inc., Irvine, CA, USA
| | - W-K Ju
- Laboratory for Optic Nerve Biology, Department of Ophthalmology, Hamilton Glaucoma Center, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|