1
|
Dmitriev AV, Linsenmeier RA. pH in the vertebrate retina and its naturally occurring and pathological changes. Prog Retin Eye Res 2025; 104:101321. [PMID: 39608565 PMCID: PMC11711014 DOI: 10.1016/j.preteyeres.2024.101321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
This review summarizes the existing information on the concentration of H+ (pH) in vertebrate retinae and its changes due to various reasons. Special features of H+ homeostasis that make it different from other ions will be discussed, particularly metabolic production of H+ and buffering. The transretinal distribution of extracellular H+ concentration ([H+]o) and its changes under illumination and other conditions will be described in detail, since [H+]o is more intensively investigated than intracellular pH. In vertebrate retinae, the highest [H+]o occurs in the inner part of the outer nuclear layer, and decreases toward the RPE, reaching the blood level on the apical side of the RPE. [H+]o falls toward the vitreous as well, but less, so that the inner retina is acidic to the vitreous. Light leads to complex changes with both electrogenic and metabolic origins, culminating in alkalinization. There is a rhythm of [H+]o with H+ being higher during circadian night. Extracellular pH can potentially be used as a signal in intercellular volume transmission, but evidence is against pH as a normal controller of fluid transport across the RPE or as a horizontal cell feedback signal. Pathological and experimentally created conditions (systemic metabolic acidosis, hypoxia and ischemia, vascular occlusion, excess glucose and diabetes, genetic disorders, and blockade of carbonic anhydrase) disturb H+ homeostasis, mostly producing retinal acidosis, with consequences for retinal blood flow, metabolism and function.
Collapse
Affiliation(s)
- Andrey V Dmitriev
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA.
| | - Robert A Linsenmeier
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Department of Neurobiology, Northwestern University, Evanston, IL, USA; Department of Ophthalmology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
2
|
Grosche A, Grosche J, Verkhratsky A. Physiology and pathophysiology of the retinal neuroglia. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:239-265. [PMID: 40148047 DOI: 10.1016/b978-0-443-19102-2.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Neuroglia of the retina are represented by Müller glia, parenchymal astrocytes, microglia and oligodendrocytes mainly associated with the optic nerve. Müller glia are the most numerous glia, endowed with multiple homeostatic functions and indispensable for the retinal morphofunctional organization. Müller cells integrate retinal neurons into individual functional units (known as retinal columns) and act as a living light guide, transmitting photons to photoreceptors. In pathology, retinal neuroglia undergo complex changes, which include upregulation of neuroprotection, reactive gliosis, and functional asthenia. The balance between all these changes defines the progression and outcome of retinal disorders.
Collapse
Affiliation(s)
- Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, München, Germany.
| | | | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Department of Neurosciences, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Bizkaia, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
3
|
Spitz MP, Anderson DR, Vrabec TR. An unusual pAIR: Anti-PKM2 antibody and occult pancreatic adenocarcinoma. Am J Ophthalmol Case Rep 2024; 36:102166. [PMID: 39351584 PMCID: PMC11440255 DOI: 10.1016/j.ajoc.2024.102166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/10/2024] [Accepted: 09/01/2024] [Indexed: 10/04/2024] Open
Abstract
Purpose To describe the clinical, laboratory and multimodal imaging findings in paraneoplastic autoimmune retinopathy (p-AIR) associated with anti-pyruvate kinase M2 antibody (anti-PKM2) and occult pancreatic adenocarcinoma. Observations A 70 year old male with blurred vision, nyctalopia and concurrent difficulty with glucose control had retinal vascular attenuation and diffuse punctate pigment clumping in both eyes. Multimodal imaging demonstrated corresponding stippled hypofluorescence on fluorescein angiography, stippled hyperautofluorescence and a hyperautoflourescent macular ring with fundus autofluorescence, and focal hyperreflectivity at the level of the RPE-Bruch's membrane complex with diffuse loss of outer retinal layers on ocular coherence tomography. In addition, diffuse ganglion cell loss and severe visual field constriction were present. Genetic testing for retinitis pigmentosa was normal. Screening for anti-retinal antibodies was positive for only anti-PKM2. Systemic evaluation revealed previously undiagnosed adenocarcinoma of the pancreas. Conclusions and importance Anti-PKM2 in the setting of autoimmune retinopathy may be associated with occult pancreatic cancer. The diagnosis of pAIR should be considered and systemic investigation for occult malignancy initiated even in the absence of more commonly associated anti-retinal antibodies.
Collapse
|
4
|
Kapoor S, Kalmegh V, Kumar H, Mandoli A, Shard A. Rare diseases and pyruvate kinase M2: a promising therapeutic connection. Drug Discov Today 2024; 29:103949. [PMID: 38492882 DOI: 10.1016/j.drudis.2024.103949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Pyruvate kinase M2 (PKM2) is a key glycolytic enzyme that regulates proliferating cell metabolism. The role of PKM2 in common diseases has been well established, but its role in rare diseases (RDs) is less understood. Over the past few years, PKM2 has emerged as a crucial player in RDs, including, neoplastic, respiratory, metabolic, and neurological disorders. Herein, we summarize recent findings and developments highlighting PKM2 as an emerging key player in RDs. We also discuss the current status of PKM2 modulation in RDs with particular emphasis on preclinical and clinical studies in addition to current challenges in the field.
Collapse
Affiliation(s)
- Saumya Kapoor
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Vaishnavi Kalmegh
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, NIPER-A, Gandhinagar, Gujarat, India.
| | - Amit Mandoli
- Department of Biotechnology, NIPER-A, Gandhinagar, Gujarat, India.
| | - Amit Shard
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Ahmedabad (NIPER-A), Gandhinagar, Gujarat, India.
| |
Collapse
|
5
|
Calbiague-Garcia V, Chen Y, Cádiz B, Tapia F, Paquet-Durand F, Schmachtenberg O. Extracellular lactate as an alternative energy source for retinal bipolar cells. J Biol Chem 2024; 300:106794. [PMID: 38403245 PMCID: PMC10966802 DOI: 10.1016/j.jbc.2024.106794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 02/27/2024] Open
Abstract
Retinal bipolar and amacrine cells receive visual information from photoreceptors and participate in the first steps of image processing in the retina. Several studies have suggested the operation of aerobic glycolysis and a lactate shuttle system in the retina due to the high production of this metabolite under aerobic conditions. However, whether bipolar cells form part of this metabolic circuit remains unclear. Here, we show that the monocarboxylate transporter 2 is expressed and functional in inner retinal neurons. Additionally, we used genetically encoded FRET nanosensors to demonstrate the ability of inner retinal neurons to consume extracellular lactate as an alternative to glucose. In rod bipolar cells, lactate consumption allowed cells to maintain the homeostasis of ions and electrical responses. We also found that lactate synthesis and transporter inhibition caused functional alterations and an increased rate of cell death. Overall, our data shed light on a notable but still poorly understood aspect of retinal metabolism.
Collapse
Affiliation(s)
- Victor Calbiague-Garcia
- PhD Program in Neuroscience, Universidad de Valparaíso, Valparaíso, Chile; CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile.
| | - Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, Tübingen, Germany
| | - Bárbara Cádiz
- CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile
| | - Felipe Tapia
- CINV, Instituto de Biología, Universidad de Valparaíso, Valparaíso, Chile
| | | | | |
Collapse
|
6
|
Subramanya S, Goswami MT, Miller N, Weh E, Chaudhury S, Zhang L, Andren A, Hager H, Weh KM, Lyssiotis CA, Besirli CG, Wubben TJ. Rod photoreceptor-specific deletion of cytosolic aspartate aminotransferase, GOT1, causes retinal degeneration. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1306019. [PMID: 38725581 PMCID: PMC11081273 DOI: 10.3389/fopht.2023.1306019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/15/2023] [Indexed: 05/12/2024]
Abstract
Photoreceptor cell death is the cause of vision loss in many forms of retinal disease. Metabolic dysfunction within the outer retina has been shown to be an underlying factor contributing to photoreceptor loss. Therefore, a comprehensive understanding of the metabolic pathways essential to photoreceptor health and function is key to identifying novel neuroprotective strategies. Glutamic-oxaloacetic transaminase 1 (Got1) encodes for a cytosolic aspartate aminotransferase that reversibly catalyzes the transfer of an amino group between glutamate and aspartate and is an important aspect of the malate-aspartate shuttle (MAS), which transfers reducing equivalents from the cytosol to the mitochondrial matrix. Previous work has demonstrated that the activity of this enzyme is highest in photoreceptor inner segments. Furthermore, ex vivo studies have demonstrated that the retina relies on aspartate aminotransferase for amino acid metabolism. Importantly, aspartate aminotransferase has been suggested to be an early biomarker of retinal degeneration in retinitis pigmentosa and a possible target for neuroprotection. In the present study, we characterized the effect of Got1 deletion on photoreceptor metabolism, function, and survival in vivo by using a rod photoreceptor-specific, Got1 knockout mouse model. Loss of the GOT1 enzyme from rod photoreceptors resulted in age-related photoreceptor degeneration with an accumulation of retinal aspartate and NADH and alterations in the expression of genes involved in the MAS, the tricarboxylic acid (TCA) cycle, and redox balance. Hence, GOT1 is critical to in vivo photoreceptor metabolism, function, and survival.
Collapse
Affiliation(s)
- Shubha Subramanya
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Moloy T. Goswami
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Nicholas Miller
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Eric Weh
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Sraboni Chaudhury
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Anthony Andren
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
| | - Heather Hager
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Katherine M. Weh
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Costas A. Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, United States
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Cagri G. Besirli
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| | - Thomas J. Wubben
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
7
|
Nsiah NY, Morgan AB, Donkor N, Inman DM. Long-term HIF-1α stabilization reduces respiration, promotes mitophagy, and results in retinal cell death. Sci Rep 2023; 13:20541. [PMID: 37996657 PMCID: PMC10667534 DOI: 10.1038/s41598-023-47942-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/20/2023] [Indexed: 11/25/2023] Open
Abstract
Ocular hypertension during glaucoma can lead to hypoxia, activation of the HIF transcription factors, and a metabolic shift toward glycolysis. This study aims to test whether chronic HIF activation and the attendant metabolic reprogramming can initiate glaucoma-associated pathology independently of ocular hypertension. HIF-1α stabilization was induced in mice for 2 and 4 weeks by inhibiting prolyl hydroxylases using the small molecule Roxadustat. HIF-1α stabilization and the expression of its downstream bioenergetic targets were investigated in the retina by immunofluorescence, capillary electrophoresis, and biochemical enzyme activity assays. Roxadustat dosing resulted in significant stabilization of HIF-1α in the retina by 4 weeks, and upregulation in glycolysis-associated proteins (GLUT3, PDK-1) and enzyme activity in both neurons and glia. Accordingly, succinate dehydrogenase, mitochondrial marker MTCO1, and citrate synthase activity were significantly decreased at 4 weeks, while mitophagy was significantly increased. TUNEL assay showed significant apoptosis of cells in the retina, and PERG amplitude was significantly decreased with 4 weeks of HIF-1α stabilization. A significant increase in AMPK activation and glial hypertrophy, concomitant with decreases in retinal ganglion cell function and inner retina cell death suggests that chronic HIF-1α stabilization alone is detrimental to retina metabolic homeostasis and cellular survival.
Collapse
Affiliation(s)
- Nana Yaa Nsiah
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
- Genentech, South San Francisco, CA, USA
| | - Autumn B Morgan
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Nina Donkor
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Denise M Inman
- Department of Pharmaceutical Sciences, North Texas Eye Research Institute, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
8
|
Rajala RVS, Rajala A. Unlocking the role of lactate: metabolic pathways, signaling, and gene regulation in postmitotic retinal cells. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1296624. [PMID: 38983010 PMCID: PMC11182115 DOI: 10.3389/fopht.2023.1296624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 10/19/2023] [Indexed: 07/11/2024]
Abstract
The Warburg effect, which was first described a century ago, asserts that mitotic tumor cells generate higher quantities of lactate. Intriguingly, even in typical physiological circumstances, postmitotic retinal photoreceptor cells also produce elevated levels of lactate. Initially classified as metabolic waste, lactate has since gained recognition as a significant intracellular signaling mediator and extracellular ligand. This current review endeavors to provide a concise overview and discourse on the following topics: the localization of lactate-producing enzymes, the functional significance of these enzymes, the signaling functions of lactate, and its impact on the gene expression of photoreceptors in retinal cells.
Collapse
Affiliation(s)
- Raju V. S. Rajala
- Departments of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Departments of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Departments of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Dean McGee Eye Institute, Oklahoma City, OK, United States
| | - Ammaji Rajala
- Departments of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Dean McGee Eye Institute, Oklahoma City, OK, United States
| |
Collapse
|
9
|
Okoye CN, Koren SA, Wojtovich AP. Mitochondrial complex I ROS production and redox signaling in hypoxia. Redox Biol 2023; 67:102926. [PMID: 37871533 PMCID: PMC10598411 DOI: 10.1016/j.redox.2023.102926] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/25/2023] Open
Abstract
Mitochondria are a main source of cellular energy. Oxidative phosphorylation (OXPHOS) is the major process of aerobic respiration. Enzyme complexes of the electron transport chain (ETC) pump protons to generate a protonmotive force (Δp) that drives OXPHOS. Complex I is an electron entry point into the ETC. Complex I oxidizes nicotinamide adenine dinucleotide (NADH) and transfers electrons to ubiquinone in a reaction coupled with proton pumping. Complex I also produces reactive oxygen species (ROS) under various conditions. The enzymatic activities of complex I can be regulated by metabolic conditions and serves as a regulatory node of the ETC. Complex I ROS plays diverse roles in cell metabolism ranging from physiologic to pathologic conditions. Progress in our understanding indicates that ROS release from complex I serves important signaling functions. Increasing evidence suggests that complex I ROS is important in signaling a mismatch in energy production and demand. In this article, we review the role of ROS from complex I in sensing acute hypoxia.
Collapse
Affiliation(s)
- Chidozie N Okoye
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Shon A Koren
- Department of Neurobiology, Harvard Medical School, Boston, MA, 02115, USA
| | - Andrew P Wojtovich
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA; Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
10
|
Holeček M. Aspartic Acid in Health and Disease. Nutrients 2023; 15:4023. [PMID: 37764806 PMCID: PMC10536334 DOI: 10.3390/nu15184023] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Aspartic acid exists in L- and D-isoforms (L-Asp and D-Asp). Most L-Asp is synthesized by mitochondrial aspartate aminotransferase from oxaloacetate and glutamate acquired by glutamine deamidation, particularly in the liver and tumor cells, and transamination of branched-chain amino acids (BCAAs), particularly in muscles. The main source of D-Asp is the racemization of L-Asp. L-Asp transported via aspartate-glutamate carrier to the cytosol is used in protein and nucleotide synthesis, gluconeogenesis, urea, and purine-nucleotide cycles, and neurotransmission and via the malate-aspartate shuttle maintains NADH delivery to mitochondria and redox balance. L-Asp released from neurons connects with the glutamate-glutamine cycle and ensures glycolysis and ammonia detoxification in astrocytes. D-Asp has a role in brain development and hypothalamus regulation. The hereditary disorders in L-Asp metabolism include citrullinemia, asparagine synthetase deficiency, Canavan disease, and dicarboxylic aminoaciduria. L-Asp plays a role in the pathogenesis of psychiatric and neurologic disorders and alterations in BCAA levels in diabetes and hyperammonemia. Further research is needed to examine the targeting of L-Asp metabolism as a strategy to fight cancer, the use of L-Asp as a dietary supplement, and the risks of increased L-Asp consumption. The role of D-Asp in the brain warrants studies on its therapeutic potential in psychiatric and neurologic disorders.
Collapse
Affiliation(s)
- Milan Holeček
- Department of Physiology, Faculty of Medicine in Hradec Králové, Charles University, Šimkova 870, 500 03 Hradec Králové, Czech Republic
| |
Collapse
|
11
|
Wubben TJ, Chaudhury S, Watch BT, Stuckey JA, Weh E, Fernando R, Goswami M, Pawar M, Rech JC, Besirli CG. Development of Novel Small-Molecule Activators of Pyruvate Kinase Muscle Isozyme 2, PKM2, to Reduce Photoreceptor Apoptosis. Pharmaceuticals (Basel) 2023; 16:ph16050705. [PMID: 37242488 DOI: 10.3390/ph16050705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Treatment options are lacking to prevent photoreceptor death and subsequent vision loss. Previously, we demonstrated that reprogramming metabolism via the pharmacologic activation of PKM2 is a novel photoreceptor neuroprotective strategy. However, the features of the tool compound used in those studies, ML-265, preclude its advancement as an intraocular, clinical candidate. This study sought to develop the next generation of small-molecule PKM2 activators, aimed specifically for delivery into the eye. Compounds were developed that replaced the thienopyrrolopyridazinone core of ML-265 and modified the aniline and methyl sulfoxide functional groups. Compound 2 demonstrated that structural changes to the ML-265 scaffold are tolerated from a potency and efficacy standpoint, allow for a similar binding mode to the target, and circumvent apoptosis in models of outer retinal stress. To overcome the low solubility and problematic functional groups of ML-265, compound 2's efficacious and versatile core structure for the incorporation of diverse functional groups was then utilized to develop novel PKM2 activators with improved solubility, lack of structural alerts, and retained potency. No other molecules are in the pharmaceutical pipeline for the metabolic reprogramming of photoreceptors. Thus, this study is the first to cultivate the next generation of novel, structurally diverse, small-molecule PKM2 activators for delivery into the eye.
Collapse
Affiliation(s)
- Thomas J Wubben
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Sraboni Chaudhury
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Brennan T Watch
- Department of Internal Medicine, Hematology and Oncology, Michigan Center for Therapeutic Innovation, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jeanne A Stuckey
- Departments of Biological Chemistry and Biophysics, Center for Structural Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Eric Weh
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Roshini Fernando
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Moloy Goswami
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Mercy Pawar
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| | - Jason C Rech
- Department of Internal Medicine, Hematology and Oncology, Michigan Center for Therapeutic Innovation, University of Michigan, Ann Arbor, MI 48109, USA
| | - Cagri G Besirli
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
12
|
Rajala A, Bhat MA, Teel K, Gopinadhan Nair GK, Purcell L, Rajala RVS. The function of lactate dehydrogenase A in retinal neurons: implications to retinal degenerative diseases. PNAS NEXUS 2023; 2:pgad038. [PMID: 36896135 PMCID: PMC9991461 DOI: 10.1093/pnasnexus/pgad038] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
The postmitotic retina is highly metabolic and the photoreceptors depend on aerobic glycolysis for an energy source and cellular anabolic activities. Lactate dehydrogenase A (LDHA) is a key enzyme in aerobic glycolysis, which converts pyruvate to lactate. Here we show that cell-type-specific actively translating mRNA purification by translating ribosome affinity purification shows a predominant expression of LDHA in rods and cones and LDHB in the retinal pigment epithelium and Müller cells. We show that genetic ablation of LDHA in the retina resulted in diminished visual function, loss of structure, and a loss of dorsal-ventral patterning of the cone-opsin gradient. Loss of LDHA in the retina resulted in increased glucose availability, promoted oxidative phosphorylation, and upregulated the expression of glutamine synthetase (GS), a neuron survival factor. However, lacking LDHA in Müller cells does not affect visual function in mice. Glucose shortage is associated with retinal diseases, such as age-related macular degeneration (AMD), and regulating the levels of LDHA may have therapeutic relevance. These data demonstrate the unique and unexplored roles of LDHA in the maintenance of a healthy retina.
Collapse
Affiliation(s)
- Ammaji Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
| | - Mohd A Bhat
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
| | - Kenneth Teel
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
| | - Gopa Kumar Gopinadhan Nair
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
| | - Lindsey Purcell
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
| | - Raju V S Rajala
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
- Dean McGee Eye Institute, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, 608 Stanton L. Young Blvd, Oklahoma City, OK 73104, USA
| |
Collapse
|
13
|
Calbiague García V, Chen Y, Cádiz B, Wang L, Paquet-Durand F, Schmachtenberg O. Imaging of lactate metabolism in retinal Müller cells with a FRET nanosensor. Exp Eye Res 2023; 226:109352. [PMID: 36528083 DOI: 10.1016/j.exer.2022.109352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Müller cells, the glial cells of the retina, provide metabolic support for photoreceptors and inner retinal neurons, and have been proposed as source of the significant lactate production of this tissue. To better understand the role of lactate in retinal metabolism, we expressed a lactate and a glucose nanosensor in organotypic mouse retinal explants cultured for 14 days, and used FRET imaging in acute vibratome sections of the explants to study metabolite flux in real time. Pharmacological manipulation with specific monocarboxylate transporter (MCT) inhibitors and immunohistochemistry revealed the functional expression of MCT1, MCT2 and MCT4 in Müller cells of retinal explants. The introduction of FRET nanosensors to measure key metabolites at the cellular level may contribute to a better understanding of heretofore poorly understood issues in retinal metabolism.
Collapse
Affiliation(s)
- Víctor Calbiague García
- PhD Program in Neuroscience, Universidad de Valparaíso, Valparaíso, Chile; CINV, Instituto de Biología, Universidad de Valparaíso, Chile
| | - Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Bárbara Cádiz
- CINV, Instituto de Biología, Universidad de Valparaíso, Chile
| | - Lan Wang
- Institute for Ophthalmic Research, University of Tübingen, Germany
| | | | | |
Collapse
|
14
|
Hass DT, Bisbach CM, Sadilek M, Sweet IR, Hurley JB. Aerobic Glycolysis in Photoreceptors Supports Energy Demand in the Absence of Mitochondrial Coupling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1415:435-441. [PMID: 37440069 DOI: 10.1007/978-3-031-27681-1_64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Metabolism is adapted to meet energetic needs. Based on the amount of ATP required to maintain plasma membrane potential, photoreceptor energy demands must be high. The available evidence suggests that photoreceptors primarily generate metabolic energy through aerobic glycolysis, though this evidence is based primarily on protein expression and not measurement of metabolic flux. Aerobic glycolysis can be validated by measuring flux of glucose to lactate. Aerobic glycolysis is also inefficient and thus an unexpected adaptation for photoreceptors to make. We measured metabolic rates to determine the energy-generating pathways that support photoreceptor metabolism. We found that photoreceptors indeed perform aerobic glycolysis and this is associated with mitochondrial uncoupling.
Collapse
Affiliation(s)
- Daniel T Hass
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
| | - Celia M Bisbach
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Martin Sadilek
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Ian R Sweet
- UW Medicine - Diabetes Institute, University of Washington, Seattle, WA, USA
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
15
|
Fu Z, Nilsson AK, Hellstrom A, Smith LEH. Retinopathy of prematurity: Metabolic risk factors. eLife 2022; 11:e80550. [PMID: 36420952 PMCID: PMC9691009 DOI: 10.7554/elife.80550] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
At preterm birth, the retina is incompletely vascularized. Retinopathy of prematurity (ROP) is initiated by the postnatal suppression of physiological retinal vascular development that would normally occur in utero. As the neural retina slowly matures, increasing metabolic demand including in the peripheral avascular retina, leads to signals for compensatory but pathological neovascularization. Currently, only late neovascular ROP is treated. ROP could be prevented by promoting normal vascular growth. Early perinatal metabolic dysregulation is a strong but understudied risk factor for ROP and other long-term sequelae of preterm birth. We will discuss the metabolic and oxygen needs of retina, current treatments, and potential interventions to promote normal vessel growth including control of postnatal hyperglycemia, dyslipidemia and hyperoxia-induced retinal metabolic alterations. Early supplementation of missing nutrients and growth factors and control of supplemental oxygen promotes physiological retinal development. We will discuss the current knowledge gap in retinal metabolism after preterm birth.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Anders K Nilsson
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Ann Hellstrom
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Lois EH Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
16
|
Nsiah NY, Inman DM. Destabilizing COXIV in Müller Glia Increases Retinal Glycolysis and Alters Scotopic Electroretinogram. Cells 2022; 11:cells11233756. [PMID: 36497016 PMCID: PMC9737073 DOI: 10.3390/cells11233756] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Müller glia (MG), the principal glial cell of the retina, have a metabolism that defies categorization into glycolytic versus oxidative. We showed that MG mount a strong hypoxia response to ocular hypertension, raising the question of their relative reliance on mitochondria for function. To explore the role of oxidative phosphorylation (OXPHOS) in MG energy production in vivo, we generated and characterized adult mice in which MG have impaired cytochrome c oxidase (COXIV) activity through knockout of the COXIV constituent COX10. Histochemistry and protein analysis showed that COXIV protein levels were significantly lower in knockout mouse retina compared to control. Loss of COXIV activity in MG did not induce structural abnormalities, though oxidative stress was increased. Electroretinography assessment showed that knocking out COX10 significantly impaired scotopic a- and b-wave responses. Inhibiting mitochondrial respiration in MG also altered the retinal glycolytic profile. However, blocking OXPHOS in MG did not significantly exacerbate retinal ganglion cell (RGC) loss or photopic negative response after ocular hypertension (OHT). These results suggest that MG were able to compensate for reduced COXIV stability by maintaining fundamental processes, but changes in retinal physiology and metabolism-associated proteins indicate subtle changes in MG function.
Collapse
|
17
|
Todorova V, Merolla L, Karademir D, Wögenstein GM, Behr J, Ebner LJA, Samardzija M, Grimm C. Retinal Layer Separation (ReLayS) method enables the molecular analysis of photoreceptor segments and cell bodies, as well as the inner retina. Sci Rep 2022; 12:20195. [PMID: 36424523 PMCID: PMC9691741 DOI: 10.1038/s41598-022-24586-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022] Open
Abstract
Understanding the physiology of the retina, and especially of the highly polarized photoreceptors, is essential not only to broaden our knowledge of the processes required for normal vision, but also to develop effective therapies to prevent or slow retinal degenerative diseases. However, the molecular analysis of photoreceptors is a challenge due to the heterogeneity of the retinal tissue and the lack of easy and reliable methods for cell separation. Here we present the ReLayS method-a simple technique for the separation of photoreceptor segments (PS) containing both inner and outer segments, outer nuclear layer (ONL), and inner retina (InR) that contains the remaining retinal layers. The layer-specific material isolated from a mouse half-retina with the ReLayS method was sufficient for protein isolation and Western blotting or RNA isolation and real-time PCR studies. The separation of PS, ONL, and InR was successfully validated by Western blotting and real-time PCR using proteins and genes with known expression profiles within the retina. Furthermore, the separation of the PS from the ONL enabled the detection of light-driven translocation of transducin from the PS to the soma. ReLayS is a simple and useful method to address protein and possibly metabolites distribution in photoreceptor compartments in various situations including development, ageing, and degenerative diseases.
Collapse
Affiliation(s)
- Vyara Todorova
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Zurich, Switzerland
| | - Luca Merolla
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Zurich, Switzerland
| | - Duygu Karademir
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Zurich, Switzerland
| | - Gabriele M Wögenstein
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Zurich, Switzerland
| | - Julian Behr
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Zurich, Switzerland
| | - Lynn J A Ebner
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Zurich, Switzerland
| | - Marijana Samardzija
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Zurich, Switzerland
| | - Christian Grimm
- Laboratory for Retinal Cell Biology, Department of Ophthalmology, University Hospital Zurich, University of Zurich, 8952, Schlieren, Zurich, Switzerland.
| |
Collapse
|
18
|
Chidlow G, Chan WO, Wood JPM, Casson RJ. Investigations into photoreceptor energy metabolism during experimental retinal detachment. Front Cell Neurosci 2022; 16:1036834. [PMID: 36467607 PMCID: PMC9716104 DOI: 10.3389/fncel.2022.1036834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 11/01/2022] [Indexed: 08/27/2023] Open
Abstract
Retinal detachment is a sight-threatening disorder, which occurs when the photoreceptors are separated from their vascular supply. The aim of the present study was to shed light on photoreceptor energy metabolism during experimental detachment in rats. Retinal detachment was induced in the eyes of rats via subretinal injection of sodium hyaluronate. Initially, we investigated whether detachment caused hypoxia within photoreceptors, as evaluated by the exogenous and endogenous biomarkers pimonidazole and HIF-1α, as well as by qPCR analysis of HIF target genes. The results showed no unequivocal staining for pimonidazole or HIF-1α within any detached retina, nor upregulation of HIF target genes, suggesting that any reduction in pO2 is of insufficient magnitude to produce hypoxia-induced covalent protein adducts or HIF-1α stabilisation. Subsequently, we analysed expression of cellular bioenergetic enzymes in photoreceptors during detachment. We documented loss of mitochondrial, and downregulation of glycolytic enzymes during detachment, indicating that photoreceptors have reduced energetic requirements and/or capacity. Given that detachment did not cause widespread hypoxia, but did result in downregulated expression of bioenergetic enzymes, we hypothesised that substrate insufficiency may be critical in terms of pathogenesis, and that boosting metabolic inputs may preserve photoreceptor bioenergetic production and, protect against their degeneration. Thus, we tested whether supplementation with the bioavailable energy substrate pyruvate mitigated rod and cone injury and degeneration. Despite protecting photoreceptors in culture from nutrient deprivation, pyruvate failed to protect against apoptotic death of rods, loss of cone opsins, and loss of inner segment mitochondria, in situ, when evaluated at 3 days after detachment. The regimen was also ineffective against cumulative photoreceptor deconstruction and degeneration when evaluated after 4 weeks. Retinal metabolism, particularly the bioenergetic profiles and pathological responses of the various cellular subtypes still presents a considerable knowledge gap that has important clinical consequences. While our data do not support the use of pyruvate supplementation as a means of protecting detached photoreceptors, they do provide a foundation and motivation for future research in this area.
Collapse
Affiliation(s)
- Glyn Chidlow
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia
| | | | | | | |
Collapse
|
19
|
Ardourel M, Pâris A, Felgerolle C, Lesne F, Ranchon-Cole I, Briault S, Perche O. FMRP-related retinal phenotypes: Evidence of glutamate-glutamine metabolic cycle impairment. Exp Eye Res 2022; 224:109238. [PMID: 36067823 DOI: 10.1016/j.exer.2022.109238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/16/2022] [Accepted: 08/29/2022] [Indexed: 11/18/2022]
Abstract
FMRP, the fragile X mental retardation protein coded by the FMR1 gene, is an RNA-binding protein that assists transport, stabilization and translational regulation of specific synaptic mRNAs. Its expression has been found in multiple cell types of central nervous system (CNS) including glial cells where its involvement in glutamate neurotransmitter homeostasis have been shown. Indeed, glutamate homeostasis deficit has been observed in absence of FMRP in-vivo in cortex and hippocampus structures as well as in vitro on astroglial cell culture. Interestingly, the retina which is an extension of the CNS is presenting electrophysiological alterations in absence of FMRP in both human and murine models suggesting neurotransmitter impairments. Therefore, we investigate the consequences of Fmrp absence on Glutamate-Glutamine cycle in whole retinas and primary retinal Müller cells culture which are the main glial cells of the retina. Using the Fmr1-/y mice, we have shown in vivo and in vitro that the absence of Fmrp in Müller cells is characterized by loss of Glutamate-Glutamine cycle homeostasis due to a lower Glutamine Synthetase protein expression and activity. The lack of Fmrp in the retina induces a reduced flow of glutamine synthesis. Our data established for the first time in literature a direct link between the lack of Fmrp and neurotransmitter homeostasis in the retina.
Collapse
Affiliation(s)
- Maryvonne Ardourel
- UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, F-45071, Orléans Cedex 2, France
| | - Arnaud Pâris
- UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, F-45071, Orléans Cedex 2, France
| | - Chloé Felgerolle
- UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, F-45071, Orléans Cedex 2, France
| | - Fabien Lesne
- Genetic Department, Regional Hospital, 14 Avenue de l'hôpital, 45100, Orléans, France
| | - Isabelle Ranchon-Cole
- Université Clermont Auvergne, CHU Clermont-Ferrand, Inserm, Neuro-Dol, F-63000, Clermont-Ferrand, France
| | - Sylvain Briault
- Genetic Department, Regional Hospital, 14 Avenue de l'hôpital, 45100, Orléans, France; UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, F-45071, Orléans Cedex 2, France
| | - Olivier Perche
- Genetic Department, Regional Hospital, 14 Avenue de l'hôpital, 45100, Orléans, France; UMR7355, CNRS, Orléans, France; Experimental and Molecular Immunology and Neurogenetics, University of Orléans, 3b rue de la Ferollerie, F-45071, Orléans Cedex 2, France.
| |
Collapse
|
20
|
Zhu S, Huang J, Xu R, Wang Y, Wan Y, McNeel R, Parker E, Kolson D, Yam M, Webb B, Zhao C, Sigado J, Du J. Isocitrate dehydrogenase 3b is required for spermiogenesis but dispensable for retinal viability. J Biol Chem 2022; 298:102387. [PMID: 35985423 PMCID: PMC9478456 DOI: 10.1016/j.jbc.2022.102387] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 12/02/2022] Open
Abstract
Isocitrate dehydrogenase 3 (IDH3) is a key enzyme in the mitochondrial tricarboxylic acid (TCA) cycle, which catalyzes the decarboxylation of isocitrate into α-ketoglutarate and concurrently converts NAD+ into NADH. Dysfunction of IDH3B, the β subunit of IDH3, has been previously correlated with retinal degeneration and male infertility in humans, but tissue-specific effects of IDH3 dysfunction are unclear. Here, we generated Idh3b-KO mice and found that IDH3B is essential for IDH3 activity in multiple tissues. We determined that loss of Idh3b in mice causes substantial accumulation of isocitrate and its precursors in the TCA cycle, particularly in the testes, whereas the levels of the downstream metabolites remain unchanged or slightly increased. However, the Idh3b-KO mice did not fully recapitulate the defects observed in humans. Global deletion of Idh3b only causes male infertility but not retinal degeneration in mice. Our investigation showed that loss of Idh3b causes an energetic deficit and disrupts the biogenesis of acrosome and flagellum, resulting in spermiogenesis arrestment in sperm cells. Together, we demonstrate that IDH3B controls its substrate levels in the TCA cycle, and it is required for sperm mitochondrial metabolism and spermiogenesis, highlighting the importance of the tissue-specific function of the ubiquitous TCA cycle.
Collapse
Affiliation(s)
- Siyan Zhu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506; Department of Biochemistry, West Virginia University, Morgantown, WV 26506; Department of Pharmaceutical and Pharmacological Science, West Virginia University, Morgantown, WV 26506
| | - Jiancheng Huang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506; Department of Biochemistry, West Virginia University, Morgantown, WV 26506; Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, China
| | - Rong Xu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506; Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506; Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Yiming Wan
- Department of Biomedical Engineering Department, Stony Brook University, Stony Brook, NY 11794
| | - Rachel McNeel
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506; Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Edward Parker
- Department of Ophthalmology, University of Washington, Seattle, WA 98109
| | - Douglas Kolson
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506
| | - Michelle Yam
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506; Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Bradley Webb
- Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Chen Zhao
- Eye Institute, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China; NHC Key Laboratory of Myopia (Fudan University), Key Laboratory of Myopia, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, China
| | - Jenna Sigado
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506; Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, WV 26506; Department of Biochemistry, West Virginia University, Morgantown, WV 26506.
| |
Collapse
|
21
|
Vohra R, Sanz-Morello B, Tams ALM, Mouhammad ZA, Freude KK, Hannibal J, Aldana BI, Bergersen LH, Kolko M. Prevention of Cell Death by Activation of Hydroxycarboxylic Acid Receptor 1 (GPR81) in Retinal Explants. Cells 2022; 11:cells11132098. [PMID: 35805182 PMCID: PMC9265426 DOI: 10.3390/cells11132098] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 12/22/2022] Open
Abstract
Background: Progressive retinal ganglion cell (RGC) dysfunction and death are common characteristics of retinal neurodegenerative diseases. Recently, hydroxycarboxylic acid receptor 1 (HCA1R, GPR81) was identified as a key modulator of mitochondrial function and cell survival. Thus, we aimed to test whether activation of HCA1R with 3,5-Dihydroxybenzoic acid (DHBA) also promotes RGC survival and improves energy metabolism in mouse retinas. Methods: Retinal explants were treated with 5 mM of the HCA1R agonist, 3,5-DHBA, for 2, 4, 24, and 72 h. Additionally, explants were also treated with 15 mM of L-glutamate to induce toxicity. Tissue survival was assessed through lactate dehydrogenase (LDH) viability assays. RGC survival was measured through immunohistochemical (IHC) staining. Total ATP levels were quantified through bioluminescence assays. Energy metabolism was investigated through stable isotope labeling and gas chromatography-mass spectrometry (GC-MS). Lactate and nitric oxide levels were measured through colorimetric assays. Results: HCA1R activation with 3,5-DHBAincreased retinal explant survival. During glutamate-induced death, 3,5-DHBA treatment also increased survival. IHC analysis revealed that 3,5-DHBA treatment promoted RGC survival in retinal wholemounts. 3,5-DHBA treatment also enhanced ATP levels in retinal explants, whereas lactate levels decreased. No effects on glucose metabolism were observed, but small changes in lactate metabolism were found. Nitric oxide levels remained unaltered in response to 3,5-DHBA treatment. Conclusion: The present study reveals that activation of HCA1R with 3,5-DHBA treatment has a neuroprotective effect specifically on RGCs and on glutamate-induced retinal degeneration. Hence, HCA1R agonist administration may be a potential new strategy for rescuing RGCs, ultimately preventing visual disability.
Collapse
Affiliation(s)
- Rupali Vohra
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (B.S.-M.); (A.L.M.T.); (Z.A.M.); (B.I.A.)
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark;
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
- Correspondence: (R.V.); (M.K.)
| | - Berta Sanz-Morello
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (B.S.-M.); (A.L.M.T.); (Z.A.M.); (B.I.A.)
| | - Anna Luna Mølgaard Tams
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (B.S.-M.); (A.L.M.T.); (Z.A.M.); (B.I.A.)
| | - Zaynab Ahmad Mouhammad
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (B.S.-M.); (A.L.M.T.); (Z.A.M.); (B.I.A.)
| | - Kristine Karla Freude
- Department of Veterinary and Animal Sciences, University of Copenhagen, 1870 Frederiksberg, Denmark;
| | - Jens Hannibal
- Department of Clinical Biochemistry, Bispebjerg Hospital, University of Copenhagen, 2400 Copenhagen, Denmark;
| | - Blanca Irene Aldana
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (B.S.-M.); (A.L.M.T.); (Z.A.M.); (B.I.A.)
| | - Linda Hildegaard Bergersen
- Brain Energy Muscle Group, University of Oslo, NO-0318 Oslo, Norway;
- Center for Healthy Aging, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark; (B.S.-M.); (A.L.M.T.); (Z.A.M.); (B.I.A.)
- Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, 2600 Glostrup, Denmark
- Correspondence: (R.V.); (M.K.)
| |
Collapse
|
22
|
Fort PE, Losiewicz MK, Elghazi L, Kong D, Cras-Méneur C, Fingar DC, Kimball SR, Rajala RVS, Smith AJ, Ali RR, Abcouwer SF, Gardner TW. mTORC1 regulates high levels of protein synthesis in retinal ganglion cells of adult mice. J Biol Chem 2022; 298:101944. [PMID: 35447116 PMCID: PMC9117545 DOI: 10.1016/j.jbc.2022.101944] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/02/2023] Open
Abstract
Mechanistic target of rapamycin (mTOR) and mTOR complex 1 (mTORC1), linchpins of the nutrient sensing and protein synthesis pathways, are present at relatively high levels in the ganglion cell layer (GCL) and retinal ganglion cells (RGCs) of rodent and human retinas. However, the role of mTORCs in the control of protein synthesis in RGC is unknown. Here, we applied the SUrface SEnsing of Translation (SUnSET) method of nascent protein labeling to localize and quantify protein synthesis in the retinas of adult mice. We also used intravitreal injection of an adeno-associated virus 2 vector encoding Cre recombinase in the eyes of mtor- or rptor-floxed mice to conditionally knockout either both mTORCs or only mTORC1, respectively, in cells within the GCL. A novel vector encoding an inactive Cre mutant (CreΔC) served as control. We found that retinal protein synthesis was highest in the GCL, particularly in RGC. Negation of both complexes or only mTORC1 significantly reduced protein synthesis in RGC. In addition, loss of mTORC1 function caused a significant reduction in the pan-RGC marker, RNA-binding protein with multiple splicing, with little decrease of the total number of cells in the RGC layer, even at 25 weeks after adeno-associated virus-Cre injection. These findings reveal that mTORC1 signaling is necessary for maintaining the high rate of protein synthesis in RGCs of adult rodents, but it may not be essential to maintain RGC viability. These findings may also be relevant to understanding the pathophysiology of RGC disorders, including glaucoma, diabetic retinopathy, and optic neuropathies.
Collapse
Affiliation(s)
- Patrice E Fort
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA; Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Mandy K Losiewicz
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Lynda Elghazi
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Dejuan Kong
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Corentin Cras-Méneur
- Internal Medicine (MEND Division), University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Diane C Fingar
- Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Scot R Kimball
- Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Raju V S Rajala
- Departments of Ophthalmology and Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Alexander J Smith
- Centre for Gene Therapy and Regenerative Medicine, King's College London, England, United Kingdom
| | - Robin R Ali
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA; Centre for Gene Therapy and Regenerative Medicine, King's College London, England, United Kingdom
| | - Steven F Abcouwer
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA.
| | - Thomas W Gardner
- Ophthalmology & Visual Sciences, University of Michigan Medical School, Ann Arbor, Michigan, USA; Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, USA; Internal Medicine (MEND Division), University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
23
|
Hanna J, David LA, Touahri Y, Fleming T, Screaton RA, Schuurmans C. Beyond Genetics: The Role of Metabolism in Photoreceptor Survival, Development and Repair. Front Cell Dev Biol 2022; 10:887764. [PMID: 35663397 PMCID: PMC9157592 DOI: 10.3389/fcell.2022.887764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/19/2022] [Indexed: 12/11/2022] Open
Abstract
Vision commences in the retina with rod and cone photoreceptors that detect and convert light to electrical signals. The irreversible loss of photoreceptors due to neurodegenerative disease leads to visual impairment and blindness. Interventions now in development include transplanting photoreceptors, committed photoreceptor precursors, or retinal pigment epithelial (RPE) cells, with the latter protecting photoreceptors from dying. However, introducing exogenous human cells in a clinical setting faces both regulatory and supply chain hurdles. Recent work has shown that abnormalities in central cell metabolism pathways are an underlying feature of most neurodegenerative disorders, including those in the retina. Reversal of key metabolic alterations to drive retinal repair thus represents a novel strategy to treat vision loss based on cell regeneration. Here, we review the connection between photoreceptor degeneration and alterations in cell metabolism, along with new insights into how metabolic reprogramming drives both retinal development and repair following damage. The potential impact of metabolic reprogramming on retinal regeneration is also discussed, specifically in the context of how metabolic switches drive both retinal development and the activation of retinal glial cells known as Müller glia. Müller glia display latent regenerative properties in teleost fish, however, their capacity to regenerate new photoreceptors has been lost in mammals. Thus, re-activating the regenerative properties of Müller glia in mammals represents an exciting new area that integrates research into developmental cues, central metabolism, disease mechanisms, and glial cell biology. In addition, we discuss this work in relation to the latest insights gleaned from other tissues (brain, muscle) and regenerative species (zebrafish).
Collapse
Affiliation(s)
- Joseph Hanna
- Sunnybrook Research Institute, Biological Sciences, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
| | - Luke Ajay David
- Sunnybrook Research Institute, Biological Sciences, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
| | - Yacine Touahri
- Sunnybrook Research Institute, Biological Sciences, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Taylor Fleming
- Sunnybrook Research Institute, Biological Sciences, Toronto, ON, Canada
| | - Robert A. Screaton
- Sunnybrook Research Institute, Biological Sciences, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Carol Schuurmans
- Sunnybrook Research Institute, Biological Sciences, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- *Correspondence: Carol Schuurmans,
| |
Collapse
|
24
|
Ocular Hypertension Results in Hypoxia within Glia and Neurons throughout the Visual Projection. Antioxidants (Basel) 2022; 11:antiox11050888. [PMID: 35624752 PMCID: PMC9137916 DOI: 10.3390/antiox11050888] [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: 03/25/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 12/03/2022] Open
Abstract
The magnitude and duration of hypoxia after ocular hypertension (OHT) has been a matter of debate due to the lack of tools to accurately report hypoxia. In this study, we established a topography of hypoxia in the visual pathway by inducing OHT in mice that express a fusion protein comprised of the oxygen-dependent degradation (ODD) domain of HIF-1α and a tamoxifen-inducible Cre recombinase (CreERT2) driven by a ubiquitous CAG promoter. After tamoxifen administration, tdTomato expression would be driven in cells that contain stabilized HIF-1α. Intraocular pressure (IOP) and visual evoked potential (VEP) were measured after OHT at 3, 14, and 28 days (d) to evaluate hypoxia induction. Immunolabeling of hypoxic cell types in the retina and optic nerve (ON) was performed, as well as retinal ganglion cell (RGC) and axon number quantification at each time point (6 h, 3 d, 14 d, 28 d). IOP elevation and VEP decrease were detected 3 d after OHT, which preceded RGC soma and axon loss at 14 and 28 d after OHT. Hypoxia was detected primarily in Müller glia in the retina, and microglia and astrocytes in the ON and optic nerve head (ONH). Hypoxia-induced factor (HIF-α) regulates the expression of glucose transporters 1 and 3 (GLUT1, 3) to support neuronal metabolic demand. Significant increases in GLUT1 and 3 proteins were observed in the retina and ON after OHT. Interestingly, neurons and endothelial cells within the superior colliculus in the brain also experienced hypoxia after OHT as determined by tdTomato expression. The highest intensity labeling for hypoxia was detected in the ONH. Initiation of OHT resulted in significant hypoxia that did not immediately resolve, with low-level hypoxia apparent out to 14 and 28 d, suggesting that continued hypoxia contributes to glaucoma progression. Restricted hypoxia in retinal neurons after OHT suggests a hypoxia management role for glia.
Collapse
|
25
|
Kanan Y, Hackett SF, Taneja K, Khan M, Campochiaro PA. Oxidative stress-induced alterations in retinal glucose metabolism in Retinitis Pigmentosa. Free Radic Biol Med 2022; 181:143-153. [PMID: 35134532 PMCID: PMC8891093 DOI: 10.1016/j.freeradbiomed.2022.01.032] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 10/19/2022]
Abstract
Retinitis pigmentosa occurs due to mutations that cause rod photoreceptor degeneration. Once most rods are lost, gradual degeneration of cone photoreceptors occurs. Oxidative damage and abnormal glucose metabolism have been implicated as contributors to cone photoreceptor death. Herein, we show increased phosphorylation of key enzymes of glucose metabolism in the retinas of rd10 mice, a model of RP, and retinas of wild type mice with paraquat-induced oxidative stress, thereby inhibiting these key enzymes. Dietary supplementation with glucose and pyruvate failed to overcome the inhibition, but increased reducing equivalents in the retina and improved cone function and survival. Dichloroacetate reversed the increased phosphorylation of pyruvate dehydrogenase in rd10 retina and increased histone acetylation and levels of TP53-induced glycolysis and apoptosis regulator (TIGAR), which redirected glucose metabolism toward the pentose phosphate pathway. These data indicate that oxidative stress induced damage can be reversed by shifting glycolytic intermediates toward the pentose phosphate pathway which increases reducing equivalents and provides photoreceptor protection.
Collapse
Affiliation(s)
- Yogita Kanan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sean F Hackett
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kamil Taneja
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mahmood Khan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A Campochiaro
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
26
|
Zouache MA. Variability in Retinal Neuron Populations and Associated Variations in Mass Transport Systems of the Retina in Health and Aging. Front Aging Neurosci 2022; 14:778404. [PMID: 35283756 PMCID: PMC8914054 DOI: 10.3389/fnagi.2022.778404] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/13/2022] [Indexed: 11/17/2022] Open
Abstract
Aging is associated with a broad range of visual impairments that can have dramatic consequences on the quality of life of those impacted. These changes are driven by a complex series of alterations affecting interactions between multiple cellular and extracellular elements. The resilience of many of these interactions may be key to minimal loss of visual function in aging; yet many of them remain poorly understood. In this review, we focus on the relation between retinal neurons and their respective mass transport systems. These metabolite delivery systems include the retinal vasculature, which lies within the inner portion of the retina, and the choroidal vasculature located externally to the retinal tissue. A framework for investigation is proposed and applied to identify the structures and processes determining retinal mass transport at the cellular and tissue levels. Spatial variability in the structure of the retina and changes observed in aging are then harnessed to explore the relation between variations in neuron populations and those seen among retinal metabolite delivery systems. Existing data demonstrate that the relation between inner retinal neurons and their mass transport systems is different in nature from that observed between the outer retina and choroid. The most prominent structural changes observed across the eye and in aging are seen in Bruch's membrane, which forms a selective barrier to mass transfers at the interface between the choroidal vasculature and the outer retina.
Collapse
Affiliation(s)
- Moussa A. Zouache
- John A. Moran Eye Center, Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
27
|
Goit RK, Taylor AW, Lo ACY. Anti-inflammatory α-Melanocyte-Stimulating Hormone Protects Retina After Ischemia/Reperfusion Injury in Type I Diabetes. Front Neurosci 2022; 16:799739. [PMID: 35281489 PMCID: PMC8914517 DOI: 10.3389/fnins.2022.799739] [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: 10/22/2021] [Accepted: 01/19/2022] [Indexed: 11/13/2022] Open
Abstract
Retinal ischemia/reperfusion (I/R) injury is a major cause of vision loss in many ocular diseases. Retinal I/R injury is common in diabetic retinopathy, which as a result of hyperglycemia damages the retina and can cause blindness if left untreated. Inflammation is a major contributing factor in the pathogenesis of I/R injury. α-Melanocyte-stimulating hormone (α-MSH) is an anti-inflammatory peptide hormone that has displayed protective effects against I/R-induced organ damages. Here, we aimed to investigate the protective role of α-MSH on I/R-induced diabetic retinal damage using hyperglycemic C57BL/6J Ins2Akita/+ mice. Experimental I/R injury was induced by blocking the right middle cerebral artery (MCA) for 2 h followed by 2 h or 22 h of reperfusion using the intraluminal method. Since ophthalmic artery originates proximal to the origin of the MCA, the filament also blocked blood supply to the retina. Upon treatment with α-MSH at 1 h after ischemia and 1 h after reperfusion, animals displayed significant improvement in amplitudes of b-wave and oscillatory potentials during electroretinography. α-MSH also prevented I/R-induced histological alterations and inhibited the development of retinal swelling. Loss of retinal ganglion cells as well as oxidative stress were significantly attenuated in the α-MSH-treated retinae. Level of interleukin 10 was significantly increased after α-MSH treatment. Moreover, gene expression of glutamate aspartate transporter 1, monocarboxylate transporter (MCT) 1 and MCT-2 were significantly higher after α-MSH administration. In conclusion, α-MSH mitigates the severity of I/R-induced retinal damage under hyperglycemic condition. These beneficial effects of α-MSH may have important therapeutic implications against retinal I/R injury under hyperglycemic condition.
Collapse
Affiliation(s)
- Rajesh Kumar Goit
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Andrew W. Taylor
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, United States
| | - Amy C. Y. Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- *Correspondence: Amy C. Y. Lo, , orcid.org/0000-0003-4239-6851
| |
Collapse
|
28
|
Pardo B, Herrada-Soler E, Satrústegui J, Contreras L, del Arco A. AGC1 Deficiency: Pathology and Molecular and Cellular Mechanisms of the Disease. Int J Mol Sci 2022; 23:528. [PMID: 35008954 PMCID: PMC8745132 DOI: 10.3390/ijms23010528] [Citation(s) in RCA: 9] [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: 11/22/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 02/01/2023] Open
Abstract
AGC1/Aralar/Slc25a12 is the mitochondrial carrier of aspartate-glutamate, the regulatory component of the NADH malate-aspartate shuttle (MAS) that transfers cytosolic redox power to neuronal mitochondria. The deficiency in AGC1/Aralar leads to the human rare disease named "early infantile epileptic encephalopathy 39" (EIEE 39, OMIM # 612949) characterized by epilepsy, hypotonia, arrested psychomotor neurodevelopment, hypo myelination and a drastic drop in brain aspartate (Asp) and N-acetylaspartate (NAA). Current evidence suggest that neurons are the main brain cell type expressing Aralar. However, paradoxically, glial functions such as myelin and Glutamine (Gln) synthesis are markedly impaired in AGC1 deficiency. Herein, we discuss the role of the AGC1/Aralar-MAS pathway in neuronal functions such as Asp and NAA synthesis, lactate use, respiration on glucose, glutamate (Glu) oxidation and other neurometabolic aspects. The possible mechanism triggering the pathophysiological findings in AGC1 deficiency, such as epilepsy and postnatal hypomyelination observed in humans and mice, are also included. Many of these mechanisms arise from findings in the aralar-KO mice model that extensively recapitulate the human disease including the astroglial failure to synthesize Gln and the dopamine (DA) mishandling in the nigrostriatal system. Epilepsy and DA mishandling are a direct consequence of the metabolic defect in neurons due to AGC1/Aralar deficiency. However, the deficits in myelin and Gln synthesis may be a consequence of neuronal affectation or a direct effect of AGC1/Aralar deficiency in glial cells. Further research is needed to clarify this question and delineate the transcellular metabolic fluxes that control brain functions. Finally, we discuss therapeutic approaches successfully used in AGC1-deficient patients and mice.
Collapse
Affiliation(s)
- Beatriz Pardo
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (E.H.-S.); (J.S.); (L.C.)
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM)-Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain;
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Eduardo Herrada-Soler
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (E.H.-S.); (J.S.); (L.C.)
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM)-Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain;
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Jorgina Satrústegui
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (E.H.-S.); (J.S.); (L.C.)
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM)-Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain;
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Laura Contreras
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (E.H.-S.); (J.S.); (L.C.)
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM)-Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain;
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Araceli del Arco
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid (UAM)-Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain;
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Centro Regional de Investigaciones Biomédicas, Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla La Mancha, 45071 Toledo, Spain
| |
Collapse
|
29
|
Özgümüs T, Sulaieva O, Jain R, Artner I, Lyssenko V. Starvation to Glucose Reprograms Development of Neurovascular Unit in Embryonic Retinal Cells. Front Cell Dev Biol 2021; 9:726852. [PMID: 34869314 PMCID: PMC8636675 DOI: 10.3389/fcell.2021.726852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022] Open
Abstract
Perinatal exposure to starvation is a risk factor for development of severe retinopathy in adult patients with diabetes. However, the underlying mechanisms are not completely understood. In the present study, we shed light on molecular consequences of exposure to short-time glucose starvation on the transcriptome profile of mouse embryonic retinal cells. We found a profound downregulation of genes regulating development of retinal neurons, which was accompanied by reduced expression of genes encoding for glycolytic enzymes and glutamatergic signaling. At the same time, glial and vascular markers were upregulated, mimicking the diabetes-associated increase of angiogenesis-a hallmark of pathogenic features in diabetic retinopathy. Energy deprivation as a consequence of starvation to glucose seems to be compensated by upregulation of genes involved in fatty acid elongation. Results from the present study demonstrate that short-term glucose deprivation during early fetal life differentially alters expression of metabolism- and function-related genes and could have detrimental and lasting effects on gene expression in the retinal neurons, glial cells, and vascular elements and thus potentially disrupting gene regulatory networks essential for the formation of the retinal neurovascular unit. Abnormal developmental programming during retinogenesis may serve as a trigger of reactive gliosis, accelerated neurodegeneration, and increased vascularization, which may promote development of severe retinopathy in patients with diabetes later in life.
Collapse
Affiliation(s)
- Türküler Özgümüs
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, Bergen, Norway
| | | | - Ruchi Jain
- Department of Clinical Sciences, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Isabella Artner
- Department of Clinical Sciences, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Valeriya Lyssenko
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, Bergen, Norway
- Department of Clinical Sciences, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
30
|
Andreazzoli M, Barravecchia I, De Cesari C, Angeloni D, Demontis GC. Inducible Pluripotent Stem Cells to Model and Treat Inherited Degenerative Diseases of the Outer Retina: 3D-Organoids Limitations and Bioengineering Solutions. Cells 2021; 10:cells10092489. [PMID: 34572137 PMCID: PMC8471616 DOI: 10.3390/cells10092489] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/12/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Inherited retinal degenerations (IRD) affecting either photoreceptors or pigment epithelial cells cause progressive visual loss and severe disability, up to complete blindness. Retinal organoids (ROs) technologies opened up the development of human inducible pluripotent stem cells (hiPSC) for disease modeling and replacement therapies. However, hiPSC-derived ROs applications to IRD presently display limited maturation and functionality, with most photoreceptors lacking well-developed outer segments (OS) and light responsiveness comparable to their adult retinal counterparts. In this review, we address for the first time the microenvironment where OS mature, i.e., the subretinal space (SRS), and discuss SRS role in photoreceptors metabolic reprogramming required for OS generation. We also address bioengineering issues to improve culture systems proficiency to promote OS maturation in hiPSC-derived ROs. This issue is crucial, as satisfying the demanding metabolic needs of photoreceptors may unleash hiPSC-derived ROs full potential for disease modeling, drug development, and replacement therapies.
Collapse
Affiliation(s)
| | - Ivana Barravecchia
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy;
| | | | - Debora Angeloni
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56124 Pisa, Italy;
| | - Gian Carlo Demontis
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy;
- Correspondence: (M.A.); (G.C.D.)
| |
Collapse
|
31
|
Tonade D, Kern TS. Photoreceptor cells and RPE contribute to the development of diabetic retinopathy. Prog Retin Eye Res 2021; 83:100919. [PMID: 33188897 PMCID: PMC8113320 DOI: 10.1016/j.preteyeres.2020.100919] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 10/27/2020] [Accepted: 10/31/2020] [Indexed: 12/26/2022]
Abstract
Diabetic retinopathy (DR) is a leading cause of blindness. It has long been regarded as vascular disease, but work in the past years has shown abnormalities also in the neural retina. Unfortunately, research on the vascular and neural abnormalities have remained largely separate, instead of being integrated into a comprehensive view of DR that includes both the neural and vascular components. Recent evidence suggests that the most predominant neural cell in the retina (photoreceptors) and the adjacent retinal pigment epithelium (RPE) play an important role in the development of vascular lesions characteristic of DR. This review summarizes evidence that the outer retina is altered in diabetes, and that photoreceptors and RPE contribute to retinal vascular alterations in the early stages of the retinopathy. The possible molecular mechanisms by which cells of the outer retina might contribute to retinal vascular damage in diabetes also are discussed. Diabetes-induced alterations in the outer retina represent a novel therapeutic target to inhibit DR.
Collapse
Affiliation(s)
- Deoye Tonade
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Timothy S Kern
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA; Veterans Administration Medical Center Research Service, Cleveland, OH, USA; Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA, USA; Veterans Administration Medical Center Research Service, Long Beach, CA, USA.
| |
Collapse
|
32
|
Absence of retbindin blocks glycolytic flux, disrupts metabolic homeostasis, and leads to photoreceptor degeneration. Proc Natl Acad Sci U S A 2021; 118:2018956118. [PMID: 33526685 DOI: 10.1073/pnas.2018956118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
We previously reported a model of progressive retinal degeneration resulting from the knockout of the retina-specific riboflavin binding protein, retbindin (Rtbdn -/- ). We also demonstrated a reduction in neural retinal flavins as a result of the elimination of RTBDN. Given the role of flavins in metabolism, herein we investigated the underlying mechanism of this retinal degeneration by performing metabolomic analyses on predegeneration at postnatal day (P) 45 and at the onset of functional degeneration in the P120 retinas. Metabolomics of hydrophilic metabolites revealed that individual glycolytic products accumulated in the P45 Rtbdn -/- neural retinas along with the elevation of pentose phosphate pathway, while TCA cycle intermediates remained unchanged. This was confirmed by using 13C-labeled flux measurements and immunoblotting, revealing that the key regulatory step of phosphoenolpyruvate to pyruvate was inhibited via down-regulation of the tetrameric pyruvate kinase M2 (PKM2). Separate metabolite assessments revealed that almost all intermediates of acylcarnitine fatty acid oxidation, ceramides, sphingomyelins, and multiple toxic metabolites were significantly elevated in the predegeneration Rtbdn -/- neural retina. Our data show that lack of RTBDN, and hence reduction in flavins, forced the neural retina into repurposing glucose for free-radical mitigation over ATP production. However, such sustained metabolic reprogramming resulted in an eventual metabolic collapse leading to neurodegeneration.
Collapse
|
33
|
Abstract
The outer retina is nourished from the choroid, a capillary bed just inside the sclera. O2, glucose, and other nutrients diffuse out of the choroid and then filter through a monolayer of retinal pigment epithelium (RPE) cells to fuel the retina. Recent studies of energy metabolism have revealed striking differences between retinas and RPE cells in the ways that they extract energy from fuels. The purpose of this review is to suggest and evaluate the hypothesis that the retina and RPE have complementary metabolic roles that make them depend on each other for survival and for their abilities to perform essential and specialized functions. Expected final online publication date for the Annual Review of Vision Science, Volume 7 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- James B Hurley
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington 98115, USA;
| |
Collapse
|
34
|
Nath M, Shan Y, Myers AM, Fort PE. HspB4/αA-Crystallin Modulates Neuroinflammation in the Retina via the Stress-Specific Inflammatory Pathways. J Clin Med 2021; 10:jcm10112384. [PMID: 34071438 PMCID: PMC8198646 DOI: 10.3390/jcm10112384] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/14/2021] [Accepted: 05/24/2021] [Indexed: 01/28/2023] Open
Abstract
PURPOSE We have previously demonstrated that HspB4/αA-crystallin, a molecular chaperone, plays an important intrinsic neuroprotective role during diabetes, by its phosphorylation on residue 148. We also reported that HspB4/αA-crystallin is highly expressed by glial cells. There is a growing interest in the potential causative role of low-grade inflammation in diabetic retinopathy pathophysiology and retinal Müller glial cells' (MGCs') participation in the inflammatory response. MGCs indeed play a central role in retinal homeostasis via secreting various cytokines and other mediators. Hence, this study was carried out to delineate and understand the regulatory function of HspB4/αA-crystallin in the inflammatory response associated with metabolic stresses. METHODS Primary MGCs were isolated from knockout HspB4/αA-crystallin mice. These primary cells were then transfected with plasmids encoding either wild-type (WT), phosphomimetic (T148D), or non-phosphorylatable mutants (T148A) of HspB4/αA-crystallin. The cells were exposed to multiple metabolic stresses including serum starvation (SS) or high glucose with TNF-alpha (HG + T) before being further evaluated for the expression of inflammatory markers by qPCR. The total protein expression along with subcellular localization of NF-kB and the NLRP3 component was assessed by Western blot. RESULTS Elevated levels of IL-6, IL-1β, MCP-1, and IL-18 in SS were significantly diminished in MGCs overexpressing WT and further in T148D as compared to EV. The HG + T-induced increase in these inflammatory markers was also dampened by WT and even more significantly by T148D overexpression, whereas T148A was ineffective in either stress. Further analysis revealed that overexpression of WT or the T148D, also led to a significant reduction of Nlrp3, Asc, and caspase-1 transcript expression in serum-deprived MGCs and nearly abolished the NF-kB induction in HG + T diabetes-like stress. This mechanistic effect was further evaluated at the protein level and confirmed the stress-dependent regulation of NLRP3 and NF-kB by αA-crystallin. CONCLUSIONS The data gathered in this study demonstrate the central regulatory role of HspB4/αA-crystallin and its modulation by phosphorylation on T148 in retinal MGCs. For the first time, this study demonstrates that HspB4/αA-crystallin can dampen the stress-induced expression of pro-inflammatory cytokines through the modulation of multiple key inflammatory pathways, therefore, suggesting its potential as a therapeutic target for the modulation of chronic neuroinflammation.
Collapse
Affiliation(s)
- Madhu Nath
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (M.N.); (Y.S.); (A.M.M.)
| | - Yang Shan
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (M.N.); (Y.S.); (A.M.M.)
| | - Angela M. Myers
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (M.N.); (Y.S.); (A.M.M.)
| | - Patrice Elie Fort
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA; (M.N.); (Y.S.); (A.M.M.)
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48105, USA
- Correspondence:
| |
Collapse
|
35
|
Shen W, Lee SR, Mathai AE, Zhang R, Du J, Yam MX, Pye V, Barnett NL, Rayner CL, Zhu L, Hurley JB, Seth P, Hirabayashi Y, Furuya S, Gillies MC. Effect of selectively knocking down key metabolic genes in Müller glia on photoreceptor health. Glia 2021; 69:1966-1986. [PMID: 33835598 DOI: 10.1002/glia.24005] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 01/05/2023]
Abstract
The importance of Müller glia for retinal homeostasis suggests that they may have vulnerabilities that lead to retinal disease. Here, we studied the effect of selectively knocking down key metabolic genes in Müller glia on photoreceptor health. Immunostaining indicated that murine Müller glia expressed insulin receptor (IR), hexokinase 2 (HK2) and phosphoglycerate dehydrogenase (PHGDH) but very little pyruvate dehydrogenase E1 alpha 1 (PDH-E1α) and lactate dehydrogenase A (LDH-A). We crossed Müller glial cell-CreER (MC-CreER) mice with transgenic mice carrying a floxed IR, HK2, PDH-E1α, LDH-A, or PHGDH gene to study the effect of selectively knocking down key metabolic genes in Müller glia cells on retinal health. Selectively knocking down IR, HK2, or PHGDH led to photoreceptor degeneration and reduced electroretinographic responses. Supplementing exogenous l-serine prevented photoreceptor degeneration and improved retinal function in MC-PHGDH knockdown mice. We unexpectedly found that the levels of retinal serine and glycine were not reduced but, on the contrary, highly increased in MC-PHGDH knockdown mice. Moreover, dietary serine supplementation, while rescuing the retinal phenotypes caused by genetic deletion of PHGDH in Müller glial cells, restored retinal serine and glycine homeostasis probably through regulation of serine transport. No retinal abnormalities were observed in MC-CreER mice crossed with PDH-E1α- or LDH-A-floxed mice despite Cre expression. Our findings suggest that Müller glia do not complete glycolysis but use glucose to produce serine to support photoreceptors. Supplementation with exogenous serine is effective in preventing photoreceptor degeneration caused by PHGDH deficiency in Müller glia.
Collapse
Affiliation(s)
- Weiyong Shen
- Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - So-Ra Lee
- Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Ashish Easow Mathai
- Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Rui Zhang
- Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Jianhai Du
- Department of Ophthalmology and Biochemistry, West Virginia University, Morgantown, West Virginia, USA
| | - Michelle X Yam
- Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Victoria Pye
- Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - Nigel L Barnett
- Clem Jones Centre for Regenerative Medicine, Faculty of Health Science & Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Cassie L Rayner
- Clem Jones Centre for Regenerative Medicine, Faculty of Health Science & Medicine, Bond University, Gold Coast, Queensland, Australia
| | - Ling Zhu
- Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Pankaj Seth
- Division of Interdisciplinary Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Yoshio Hirabayashi
- Sako Cellular Informatics Laboratory, RIKEN Cluster for Pioneering Research, Wako, Japan
| | - Shigeki Furuya
- Department of Bioscience and Biotechnology, Kyushu University, Fukuoka, Japan
| | - Mark C Gillies
- Discipline of Ophthalmology, Sydney Medical School, The University of Sydney, Save Sight Institute, Sydney, New South Wales, Australia
| |
Collapse
|
36
|
Jaroszynska N, Harding P, Moosajee M. Metabolism in the Zebrafish Retina. J Dev Biol 2021; 9:10. [PMID: 33804189 PMCID: PMC8006245 DOI: 10.3390/jdb9010010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Retinal photoreceptors are amongst the most metabolically active cells in the body, consuming more glucose as a metabolic substrate than even the brain. This ensures that there is sufficient energy to establish and maintain photoreceptor functions during and after their differentiation. Such high dependence on glucose metabolism is conserved across vertebrates, including zebrafish from early larval through to adult retinal stages. As the zebrafish retina develops rapidly, reaching an adult-like structure by 72 hours post fertilisation, zebrafish larvae can be used to study metabolism not only during retinogenesis, but also in functionally mature retinae. The interplay between rod and cone photoreceptors and the neighbouring retinal pigment epithelium (RPE) cells establishes a metabolic ecosystem that provides essential control of their individual functions, overall maintaining healthy vision. The RPE facilitates efficient supply of glucose from the choroidal vasculature to the photoreceptors, which produce metabolic products that in turn fuel RPE metabolism. Many inherited retinal diseases (IRDs) result in photoreceptor degeneration, either directly arising from photoreceptor-specific mutations or secondary to RPE loss, leading to sight loss. Evidence from a number of vertebrate studies suggests that the imbalance of the metabolic ecosystem in the outer retina contributes to metabolic failure and disease pathogenesis. The use of larval zebrafish mutants with disease-specific mutations that mirror those seen in human patients allows us to uncover mechanisms of such dysregulation and disease pathology with progression from embryonic to adult stages, as well as providing a means of testing novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Philippa Harding
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
| | - Mariya Moosajee
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
37
|
Pan WW, Wubben TJ, Besirli CG. Photoreceptor metabolic reprogramming: current understanding and therapeutic implications. Commun Biol 2021; 4:245. [PMID: 33627778 PMCID: PMC7904922 DOI: 10.1038/s42003-021-01765-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 01/28/2021] [Indexed: 02/06/2023] Open
Abstract
Acquired and inherited retinal disorders are responsible for vision loss in an increasing proportion of individuals worldwide. Photoreceptor (PR) death is central to the vision loss individuals experience in these various retinal diseases. Unfortunately, there is a lack of treatment options to prevent PR loss, so an urgent unmet need exists for therapies that improve PR survival and ultimately, vision. The retina is one of the most energy demanding tissues in the body, and this is driven in large part by the metabolic needs of PRs. Recent studies suggest that disruption of nutrient availability and regulation of cell metabolism may be a unifying mechanism in PR death. Understanding retinal cell metabolism and how it is altered in disease has been identified as a priority area of research. The focus of this review is on the recent advances in the understanding of PR metabolism and how it is critical to reduction-oxidation (redox) balance, the outer retinal metabolic ecosystem, and retinal disease. The importance of these metabolic processes is just beginning to be realized and unraveling the metabolic and redox pathways integral to PR health may identify novel targets for neuroprotective strategies that prevent blindness in the heterogenous group of retinal disorders.
Collapse
Affiliation(s)
- Warren W Pan
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | - Thomas J Wubben
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA.
| | - Cagri G Besirli
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
38
|
Fu Z, Kern TS, Hellström A, Smith LEH. Fatty acid oxidation and photoreceptor metabolic needs. J Lipid Res 2021; 62:100035. [PMID: 32094231 PMCID: PMC7905050 DOI: 10.1194/jlr.tr120000618] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/14/2020] [Indexed: 01/31/2023] Open
Abstract
Photoreceptors have high energy demands and a high density of mitochondria that produce ATP through oxidative phosphorylation (OXPHOS) of fuel substrates. Although glucose is the major fuel for CNS brain neurons, in photoreceptors (also CNS), most glucose is not metabolized through OXPHOS but is instead metabolized into lactate by aerobic glycolysis. The major fuel sources for photoreceptor mitochondria remained unclear for almost six decades. Similar to other tissues (like heart and skeletal muscle) with high metabolic rates, photoreceptors were recently found to metabolize fatty acids (palmitate) through OXPHOS. Disruption of lipid entry into photoreceptors leads to extracellular lipid accumulation, suppressed glucose transporter expression, and a duel lipid/glucose fuel shortage. Modulation of lipid metabolism helps restore photoreceptor function. However, further elucidation of the types of lipids used as retinal energy sources, the metabolic interaction with other fuel pathways, as well as the cross-talk among retinal cells to provide energy to photoreceptors is not fully understood. In this review, we will focus on the current understanding of photoreceptor energy demand and sources, and potential future investigations of photoreceptor metabolism.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Manton Center for Orphan Disease, Boston Children's Hospital, Boston, MA, USA.
| | - Timothy S Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, Irvine, CA, USA
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
The innate immune system in diabetic retinopathy. Prog Retin Eye Res 2021; 84:100940. [PMID: 33429059 DOI: 10.1016/j.preteyeres.2021.100940] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/24/2020] [Accepted: 01/03/2021] [Indexed: 12/20/2022]
Abstract
The prevalence of diabetes has been rising steadily in the past half-century, along with the burden of its associated complications, including diabetic retinopathy (DR). DR is currently the most common cause of vision loss in working-age adults in the United States. Historically, DR has been diagnosed and classified clinically based on what is visible by fundoscopy; that is vasculature alterations. However, recent technological advances have confirmed pathology of the neuroretina prior to any detectable vascular changes. These, coupled with molecular studies, and the positive impact of anti-inflammatory therapeutics in DR patients have highlighted the central involvement of the innate immune system. Reminiscent of the systemic impact of diabetes, immune dysregulation has become increasingly identified as a key element of the pathophysiology of DR by interfering with normal homeostatic systems. This review uses the growing body of literature across various model systems to demonstrate the clear involvement of all three pillars of the immune system: immune-competent cells, mediators, and the complement system. It also demonstrates how the relative contribution of each of these requires more extensive analysis, including in human tissues over the continuum of disease progression. Finally, although this review demonstrates how the complex interactions of the immune system pose many more questions than answers, the intimately connected nature of the three pillars of the immune system may also point to possible new targets to reverse or even halt reverse retinopathy.
Collapse
|
40
|
Mitochondria: The Retina's Achilles' Heel in AMD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1256:237-264. [PMID: 33848005 DOI: 10.1007/978-3-030-66014-7_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Strong experimental evidence from studies in human donor retinas and animal models supports the idea that the retinal pathology associated with age-related macular degeneration (AMD) involves mitochondrial dysfunction and consequent altered retinal metabolism. This chapter provides a brief overview of mitochondrial structure and function, summarizes evidence for mitochondrial defects in AMD, and highlights the potential ramifications of these defects on retinal health and function. Discussion of mitochondrial haplogroups and their association with AMD brings to light how mitochondrial genetics can influence disease outcome. As one of the most metabolically active tissues in the human body, there is strong evidence that disruption in key metabolic pathways contributes to AMD pathology. The section on retinal metabolism reviews cell-specific metabolic differences and how the metabolic interdependence of each retinal cell type creates a unique ecosystem that is disrupted in the diseased retina. The final discussion includes strategies for therapeutic interventions that target key mitochondrial pathways as a treatment for AMD.
Collapse
|
41
|
Grotegut P, Perumal N, Kuehn S, Smit A, Dick HB, Grus FH, Joachim SC. Minocycline reduces inflammatory response and cell death in a S100B retina degeneration model. J Neuroinflammation 2020; 17:375. [PMID: 33317557 PMCID: PMC7737388 DOI: 10.1186/s12974-020-02012-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/28/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Previous studies noted that intravitreal injection of S100B triggered a glaucoma-like degeneration of retina and optic nerve as well as microglia activation after 14 days. The precise role of microglia in our intravitreal S100B model is still unclear. Hence, microglia were inhibited through minocycline. The aim is to investigate whether microglia have a significant influence on the degeneration process or whether they are only a side effect in the model studied here. METHODS Minocycline was applied daily in rats by intraperitoneal injection using two different concentrations (13.5 mg/kg body weight, 25 mg/kg body weight). One day after treatment start, S100B or PBS was intravitreally injected in one eye per rat. The naïve groups received no injections. This resulted in a total of five groups (naïve n = 14, PBS n = 14, S100B n = 13, 13.5 mg/kg mino n = 15, 25 mg/kg mino n = 15). At day 14, electroretinogram measurements were performed, followed by immunofluorescence and label-free quantitative proteomics analysis. The focus of these investigations was on the survival of RGCs as well as their axons, the response of the microglia, and the identification of further pathological modes of action of S100B. RESULTS The best signal transmission was detected via ERG in the 13.5 mg/kg mino group. The inhibition of the microglia protected optic nerve neurofilaments and decreased the negative impact of S100B on RGCs. However, the minocycline treatment could not trigger complete protection of RGCs. Furthermore, in retina and optic nerve, the minocycline treatment reduced the number and activity of S100B-triggered microglia in a concentration-dependent manner. Proteomics analysis showed that S100B application led to numerous metabolic functions and cellular stress, mainly an increased inflammatory response, glycolysis, and mitochondrial dysfunction, which caused oxidative stress in the retina. Importantly, the protective capability of lower dose of minocycline was unraveled by suppressing the apoptotic, inflammatory, and the altered metabolic processes caused by S100B insult in the retina. CONCLUSION Intravitreally injected S100B not only led to a pro-inflammatory microglial reaction, but also a mitochondrial and metabolic dysfunction. Also, these results suggest that an excessive microglial response may be a significant degenerative factor, but not the only trigger for increased cell death.
Collapse
Affiliation(s)
- Pia Grotegut
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Natarajan Perumal
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sandra Kuehn
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Andreas Smit
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - H Burkhard Dick
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany
| | - Franz H Grus
- Experimental and Translational Ophthalmology, Department of Ophthalmology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Stephanie C Joachim
- Experimental Eye Research Institute, University Eye Hospital, Ruhr-University Bochum, In der Schornau 23-25, 44892, Bochum, Germany.
| |
Collapse
|
42
|
Yumnamcha T, Guerra M, Singh LP, Ibrahim AS. Metabolic Dysregulation and Neurovascular Dysfunction in Diabetic Retinopathy. Antioxidants (Basel) 2020; 9:E1244. [PMID: 33302369 PMCID: PMC7762582 DOI: 10.3390/antiox9121244] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic retinopathy is a major cause of ocular complications in patients with type 1 and type 2 diabetes in developed countries. Due to the continued increase in the number of people with obesity and diabetes in the United States of America and globally, the incidence of diabetic retinopathy is expected to increase significantly in the coming years. Diabetic retinopathy is widely accepted as a combination of neurodegenerative and microvascular changes; however, which change occurs first is not yet understood. Although the pathogenesis of diabetic retinopathy is very complex, regulated by numerous signaling pathways and cellular processes, maintaining glucose homeostasis is still an essential component for normal physiological functioning of retinal cells. The maintenance of glucose homeostasis is finely regulated by coordinated interplay between glycolysis, Krebs cycle, and oxidative phosphorylation. Glycolysis is the most conserved metabolic pathway in biology and is tightly regulated to maintain a steady-state concentration of glycolytic intermediates; this regulation is called scheduled or regulated glycolysis. However, an abnormal increase in glycolytic flux generates large amounts of intermediate metabolites that can be shunted into different damaging pathways including the polyol pathway, hexosamine pathway, diacylglycerol-dependent activation of the protein kinase C pathway, and Amadori/advanced glycation end products (AGEs) pathway. In addition, disrupting the balance between glycolysis and oxidative phosphorylation leads to other biochemical and molecular changes observed in diabetic retinopathy including endoplasmic reticulum-mitochondria miscommunication and mitophagy dysregulation. This review will focus on how dysregulation of glycolysis contributes to diabetic retinopathy.
Collapse
Affiliation(s)
- Thangal Yumnamcha
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Michael Guerra
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Lalit Pukhrambam Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Ahmed S. Ibrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
43
|
Li B, Zhang T, Liu W, Wang Y, Xu R, Zeng S, Zhang R, Zhu S, Gillies MC, Zhu L, Du J. Metabolic Features of Mouse and Human Retinas: Rods versus Cones, Macula versus Periphery, Retina versus RPE. iScience 2020; 23:101672. [PMID: 33196018 PMCID: PMC7644940 DOI: 10.1016/j.isci.2020.101672] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023] Open
Abstract
Photoreceptors, especially cones, which are enriched in the human macula, have high energy demands, making them vulnerable to metabolic stress. Metabolic dysfunction of photoreceptors and their supporting retinal pigment epithelium (RPE) is an important underlying cause of degenerative retinal diseases. However, how cones and the macula support their exorbitant metabolic demand and communicate with RPE is unclear. By profiling metabolite uptake and release and analyzing metabolic genes, we have found cone-rich retinas and human macula share specific metabolic features with upregulated pathways in pyruvate metabolism, mitochondrial TCA cycle, and lipid synthesis. Human neural retina and RPE have distinct but complementary metabolic features. Retinal metabolism centers on NADH production and neurotransmitter biosynthesis. The retina needs aspartate to sustain its aerobic glycolysis and mitochondrial metabolism. RPE metabolism is directed toward NADPH production and biosynthesis of acetyl-rich metabolites, serine, and others. RPE consumes multiple nutrients, including proline, to produce metabolites for the retina.
Collapse
Affiliation(s)
- Bo Li
- Departments of Ophthalmology and Biochemistry, West Virginia University, WVU Eye Institute, One Medical Center Dr, PO Box 9193, Morgantown, WV 26506, USA.,Department of Ophthalmology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province 225100, China
| | - Ting Zhang
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Wei Liu
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA.,Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yekai Wang
- Departments of Ophthalmology and Biochemistry, West Virginia University, WVU Eye Institute, One Medical Center Dr, PO Box 9193, Morgantown, WV 26506, USA
| | - Rong Xu
- Departments of Ophthalmology and Biochemistry, West Virginia University, WVU Eye Institute, One Medical Center Dr, PO Box 9193, Morgantown, WV 26506, USA
| | - Shaoxue Zeng
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Rui Zhang
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Siyan Zhu
- Departments of Ophthalmology and Biochemistry, West Virginia University, WVU Eye Institute, One Medical Center Dr, PO Box 9193, Morgantown, WV 26506, USA
| | - Mark C Gillies
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Ling Zhu
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Jianhai Du
- Departments of Ophthalmology and Biochemistry, West Virginia University, WVU Eye Institute, One Medical Center Dr, PO Box 9193, Morgantown, WV 26506, USA
| |
Collapse
|
44
|
Haydinger CD, Kittipassorn T, Peet DJ. Power to see-Drivers of aerobic glycolysis in the mammalian retina: A review. Clin Exp Ophthalmol 2020; 48:1057-1071. [PMID: 32710505 DOI: 10.1111/ceo.13833] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/03/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022]
Abstract
The mammalian retina converts most glucose to lactate rather than catabolizing it completely to carbon dioxide via oxidative phosphorylation, despite the availability of oxygen. This unusual metabolism is known as aerobic glycolysis or the Warburg effect. Molecules and pathways that drive aerobic glycolysis have been identified and thoroughly studied in the context of cancer but remain relatively poorly understood in the retina. Here, we review recent research on the molecular mechanisms that underly aerobic glycolysis in the retina, focusing on key glycolytic enzymes including hexokinase 2 (HK2), pyruvate kinase M2 (PKM2) and lactate dehydrogenase A (LDHA). We also discuss the potential involvement of cell signalling and transcriptional pathways including phosphoinositide 3-kinase (PI3K) signalling, fibroblast growth factor receptor (FGFR) signalling, and hypoxia-inducible factor 1 (HIF-1), which have been implicated in driving aerobic glycolysis in the context of cancer.
Collapse
Affiliation(s)
- Cameron D Haydinger
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Thaksaon Kittipassorn
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia.,Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University, Mahidol, Thailand
| | - Daniel J Peet
- School of Biological Sciences, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
45
|
Retinal energy metabolism in health and glaucoma. Prog Retin Eye Res 2020; 81:100881. [PMID: 32712136 DOI: 10.1016/j.preteyeres.2020.100881] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/25/2020] [Accepted: 06/28/2020] [Indexed: 01/17/2023]
Abstract
Energy metabolism refers to the processes by which life transfers energy to do cellular work. The retina's relatively large energy demands make it vulnerable to energy insufficiency. In addition, evolutionary pressures to optimize human vision have been traded against retinal ganglion cell bioenergetic fragility. Details of the metabolic profiles of the different retinal cells remain poorly understood and are challenging to resolve. Detailed immunohistochemical mapping of the energy pathway enzymes and substrate transporters has provided some insights and highlighted interspecies differences. The different spatial metabolic patterns between the vascular and avascular retinas can account for some inconsistent data in the literature. There is a consilience of evidence that at least some individuals with glaucoma have impaired RGC energy metabolism, either due to impaired nutrient supply or intrinsic metabolic perturbations. Bioenergetic-based therapy for glaucoma has a compelling pathophysiological foundation and is supported by recent successes in animal models. Recent demonstrations of visual and electrophysiological neurorecovery in humans with glaucoma is highly encouraging and motivates longer duration trials investigating bioenergetic neuroprotection.
Collapse
|
46
|
Harun-Or-Rashid M, Pappenhagen N, Zubricky R, Coughlin L, Jassim AH, Inman DM. MCT2 overexpression rescues metabolic vulnerability and protects retinal ganglion cells in two models of glaucoma. Neurobiol Dis 2020; 141:104944. [PMID: 32422282 DOI: 10.1016/j.nbd.2020.104944] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 05/07/2020] [Accepted: 05/12/2020] [Indexed: 12/18/2022] Open
Abstract
Improving cellular access to energy substrates is one strategy to overcome observed declines in energy production and utilization in the aged and pathologic central nervous system. Monocarboxylate transporters (MCTs), the movers of lactate, pyruvate, and ketone bodies into or out of a cell, are significantly decreased in the DBA/2 J mouse model of glaucoma. In order to confirm MCT decreases are disease-associated, we decreased MCT2 in the retinas of MCT2fl/+ mice using an injection of AAV2-cre, observing significant decline in ATP production and visual evoked potential. Restoring MCT2 levels in retinal ganglion cells (RGCs) via intraocular injection of AAV2-GFP-MCT2 in two models of glaucoma, the DBA/2 J (D2), and a magnetic bead model of ocular hypertension (OHT), preserved RGCs and their function. Viral-mediated overexpression of MCT2 increased RGC density and axon number, reduced energy imbalance, and increased mitochondrial function as measured by cytochrome c oxidase and succinate dehydrogenase activity in both models of glaucoma. Ocular hypertensive mice injected with AAV2:MCT2 had significantly greater P1 amplitude as measured by pattern electroretinogram than mice with OHT alone. These findings indicate overexpression of MCT2 improves energy homeostasis in the glaucomatous visual system, suggesting that expanding energy input options for cells is a viable option to combat neurodegeneration.
Collapse
Affiliation(s)
| | - Nathaniel Pappenhagen
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States of America; School of Biomedical Sciences, Kent State University, Kent, OH, United States of America
| | - Ryan Zubricky
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Lucy Coughlin
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States of America; School of Biomedical Sciences, Kent State University, Kent, OH, United States of America
| | - Assraa Hassan Jassim
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States of America
| | - Denise M Inman
- Department of Pharmaceutical Sciences, Northeast Ohio Medical University, Rootstown, OH, United States of America.
| |
Collapse
|
47
|
Rajala RVS. Aerobic Glycolysis in the Retina: Functional Roles of Pyruvate Kinase Isoforms. Front Cell Dev Biol 2020; 8:266. [PMID: 32426353 PMCID: PMC7203425 DOI: 10.3389/fcell.2020.00266] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/30/2020] [Indexed: 12/28/2022] Open
Abstract
One hundred years ago, Otto Heinrich Warburg observed that postmitotic retinal cells are the highest oxygen-consuming cells in the body. He compared these cells to actively growing mitotic tumor cells since both cells reprogram glucose for anabolic processes, which include lipid, protein, and RNA/DNA synthesis, and for antioxidant metabolism. To achieve this metabolic reprogramming, cancer cells preferentially express a less active dimeric form, the M2 isoform of pyruvate kinase (PKM2), which shuttles glucose toward the accumulation of glycolytic intermediates that redirect cell activities into anabolic processes. Similar to cancer cells, retinal photoreceptors predominantly express the M2 isoform of PKM2. This isoform performs both metabolic and non-metabolic functions in photoreceptor cells. This review focuses on the metabolic and non-metabolic roles of pyruvate kinases in photoreceptor cell functions.
Collapse
Affiliation(s)
- Raju V S Rajala
- Department of Ophthalmology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Physiology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Cell Biology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Dean McGee Eye Institute, Oklahoma City, OK, United States
| |
Collapse
|
48
|
Sinha T, Naash MI, Al-Ubaidi MR. The Symbiotic Relationship between the Neural Retina and Retinal Pigment Epithelium Is Supported by Utilizing Differential Metabolic Pathways. iScience 2020; 23:101004. [PMID: 32252018 PMCID: PMC7132098 DOI: 10.1016/j.isci.2020.101004] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/09/2020] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
The neural retina and retinal pigment epithelium (RPE) maintain a symbiotic metabolic relationship, disruption of which leads to debilitating vision loss. The current study was undertaken to identify the differences in the steady-state metabolite levels and the pathways functioning between bona fide neural retina and RPE. Global metabolomics and cluster analyses identified 650 metabolites differentially modulated between the murine neural retina and RPE. Of these, 387 and 163 were higher in the RPE and the neural retina, respectively. Further analysis coupled with transcript and protein level investigations revealed that under normal physiological conditions, the RPE utilizes the pentose phosphate (>3-fold in RPE), serine (>10-fold in RPE), and sphingomyelin biosynthesis (>5-fold in RPE) pathways. Conversely, the neural retina relied mostly on glycolysis. These results show how the RPE and the neural retina have acquired an efficient, complementary and metabolically diverse symbiotic niche to support each other's distinct functions.
Collapse
Affiliation(s)
- Tirthankar Sinha
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA.
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204, USA.
| |
Collapse
|
49
|
Singh C, Tran V, McCollum L, Bolok Y, Allan K, Yuan A, Hoppe G, Brunengraber H, Sears JE. Hyperoxia induces glutamine-fuelled anaplerosis in retinal Müller cells. Nat Commun 2020; 11:1277. [PMID: 32152301 PMCID: PMC7062830 DOI: 10.1038/s41467-020-15066-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/17/2020] [Indexed: 01/03/2023] Open
Abstract
Although supplemental oxygen is required to promote survival of severely premature infants, hyperoxia is simultaneously harmful to premature developing tissues such as in the retina. Here we report the effect of hyperoxia on central carbon metabolism in primary mouse Müller glial cells and a human Müller glia cell line (M10-M1 cells). We found decreased flux from glycolysis entering the tricarboxylic acid cycle in Müller cells accompanied by increased glutamine consumption in response to hyperoxia. In hyperoxia, anaplerotic catabolism of glutamine by Müller cells increased ammonium release two-fold. Hyperoxia induces glutamine-fueled anaplerosis that reverses basal Müller cell metabolism from production to consumption of glutamine. Prematurely born babies need extra oxygen to survive, but this can cause damage to the eyes and lead to infant blindness. Here the authors show that this hyperoxia changes the metabolism of Müller cells in the retina such that they use up, rather than produce, glutamine and secrete excess ammonium.
Collapse
Affiliation(s)
- Charandeep Singh
- Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Vincent Tran
- Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Leah McCollum
- Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Youstina Bolok
- Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Kristin Allan
- Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.,Molecular Medicine, Case Western Reserve School of Medicine Cleveland, Cleveland, OH, 44106, USA
| | - Alex Yuan
- Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - George Hoppe
- Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Henri Brunengraber
- Department of Nutrition, Case Western Reserve School of Medicine Cleveland, Cleveland, OH, 44106, USA
| | - Jonathan E Sears
- Ophthalmic Research, Cole Eye Institute, Cleveland Clinic, Cleveland, OH, 44195, USA. .,Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
50
|
Han JYS, Kinoshita J, Bisetto S, Bell BA, Nowak RA, Peachey NS, Philp NJ. Role of monocarboxylate transporters in regulating metabolic homeostasis in the outer retina: Insight gained from cell-specific Bsg deletion. FASEB J 2020; 34:5401-5419. [PMID: 32112484 DOI: 10.1096/fj.201902961r] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/29/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023]
Abstract
The neural retina metabolizes glucose through aerobic glycolysis generating large amounts of lactate. Lactate flux into and out of cells is regulated by proton-coupled monocarboxylate transporters (MCTs), which are encoded by members of the Slc16a family. MCT1, MCT3, and MCT4 are expressed in the retina and require association with the accessory protein basigin, encoded by Bsg, for maturation and trafficking to the plasma membrane. Bsg-/- mice have severely reduced electroretinograms (ERGs) and progressive photoreceptor degeneration, which is presumed to be driven by metabolic dysfunction resulting from loss of MCTs. To understand the basis of the Bsg-/- phenotype, we generated mice with conditional deletion of Bsg in rods (RodΔBsg), cones (Cone∆Bsg), or retinal pigment epithelial cells (RPEΔBsg). RodΔBsg mice showed a progressive loss of photoreceptors, while ConeΔBsg mice did not display a degenerative phenotype. The RPEΔBsg mice developed a distinct phenotype characterized by severely reduced ERG responses as early as 4 weeks of age. The loss of lactate transporters from the RPE most closely resembled the phenotype of the Bsg-/- mouse, suggesting that the regulation of lactate levels in the RPE and the subretinal space is essential for the viability and function of photoreceptors.
Collapse
Affiliation(s)
- John Y S Han
- Department of Pathology, Anatomy, & Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Sara Bisetto
- Department of Pathology, Anatomy, & Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Brent A Bell
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, PA, USA
| | - Romana A Nowak
- Animal Sciences, University of Illinois at Urbana-Champaign, Urbana-Champaign, IL, USA
| | - Neal S Peachey
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA.,Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.,Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Nancy J Philp
- Department of Pathology, Anatomy, & Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|