1
|
Hass DT, Pandey K, Engel A, Horton N, Haydinger CD, Robbings BM, Lim RR, Sadilek M, Zhang Q, Gulette GA, Li A, Xu L, Miller JML, Chao JR, Hurley JB. Acetyl-CoA carboxylase inhibition increases retinal pigment epithelial cell fatty acid flux and restricts apolipoprotein efflux. J Biol Chem 2024; 300:107772. [PMID: 39276938 PMCID: PMC11490839 DOI: 10.1016/j.jbc.2024.107772] [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: 12/01/2023] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/17/2024] Open
Abstract
Lipid-rich deposits called drusen accumulate under the retinal pigment epithelium (RPE) in the eyes of patients with age-related macular degeneration and Sorsby's fundus dystrophy (SFD). Drusen may contribute to photoreceptor degeneration in these blinding diseases. Stimulating β-oxidation of fatty acids could decrease the availability of lipid with which RPE cells generate drusen. Inhibitors of acetyl-CoA carboxylase (ACC) stimulate β-oxidation and diminish lipid accumulation in fatty liver disease. In this report, we test the hypothesis that an ACC inhibitor, Firsocostat, can diminish lipid deposition by RPE cells. We probed metabolism and cellular function in mouse RPE-choroid tissue and human RPE cells. We used 13C6-glucose, 13C16-palmitate, and gas chromatography-linked mass spectrometry to monitor effects of Firsocostat on glycolytic, Krebs cycle, and fatty acid metabolism. We quantified lipid abundance, apolipoprotein E levels, and vascular endothelial growth factor release using liquid chromatography-mass spectrometry, ELISAs, and immunostaining. RPE barrier function was assessed by trans-epithelial electrical resistance (TEER). Firsocostat-mediated ACC inhibition increases β-oxidation, decreases intracellular lipid levels, diminishes lipoprotein release, and increases TEER. When human serum or outer segments are used to stimulate lipoprotein release, fewer lipoproteins are released in the presence of Firsocostat. In a culture model of SFD, Firsocostat stimulates fatty acid oxidation, increases TEER, and decreases apolipoprotein E release. We conclude that Firsocostat remodels RPE metabolism and can limit lipid deposition. This suggests that ACC inhibition could be an effective strategy for diminishing pathologic drusen in the eyes of patients with age-related macular degeneration or SFD.
Collapse
Affiliation(s)
- Daniel T Hass
- Department of Biochemistry, University of Washington, Seattle, Washington, USA.
| | - Kriti Pandey
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Abbi Engel
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Hospital, Seattle, Washington, USA
| | - Noah Horton
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Cameron D Haydinger
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | - Brian M Robbings
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Rayne R Lim
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA
| | - Martin Sadilek
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Qitao Zhang
- Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Gillian A Gulette
- Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy Li
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - Jason M L Miller
- Kellogg Eye Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer R Chao
- Department of Ophthalmology, University of Washington, Seattle, Washington, USA
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, Washington, USA; Department of Ophthalmology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
2
|
Gulette GA, Hass DT, Pandey K, Zhang Q, Han JYS, Engel A, Chao JR, Philp NJ, Hurley JB, Miller JML. Reassessing retinal pigment epithelial ketogenesis: Enzymatic assays for ketone body levels provide inaccurate results. Exp Eye Res 2024; 245:109966. [PMID: 38857822 DOI: 10.1016/j.exer.2024.109966] [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: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024]
Abstract
The retinal pigment epithelium (RPE) is omnivorous and can utilize a wide range of substrates for oxidative phosphorylation. Certain tissues with high mitochondrial metabolic load are capable of ketogenesis, a biochemical pathway that consolidates acetyl-CoA into ketone bodies. Earlier work demonstrated that the RPE expresses the rate-limiting enzyme for ketogenesis, 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2), and that the RPE indeed produces ketone bodies, including beta-hydroxybutyrate (β-HB). Prior work, based on detecting β-HB via enzymatic assays, suggested that differentiated cultures of primary RPE preferentially export β-HB across the apical membrane. Here, we compare the accuracy of measuring β-HB by enzymatic assay kits to mass spectrometry analysis. We found that commercial kits lack the sensitivity to accurately measure the levels of β-HB in RPE cultures and are prone to artifact. Using mass spectrometry, we found that while RPE cultures secrete β-HB, they do so equally to both apical and basal sides. We also find RPE is capable of consuming β-HB as levels rise. Using isotopically labeled glucose, amino acid, and fatty acid tracers, we found that carbons from both fatty acids and ketogenic amino acids, but not from glucose, produce β-HB. Altogether, we substantiate β-HB secretion in RPE but find that the secretion is equal apically and basally, RPE β-HB can derive from ketogenic amino acids or fatty acids, and accurate β-HB assessment requires mass spectrometric analysis.
Collapse
Affiliation(s)
| | - Daniel T Hass
- Department of Biochemistry, University of Washington, United States
| | - Kriti Pandey
- Department of Biochemistry, University of Washington, United States
| | - Qitao Zhang
- Kellogg Eye Center, University of Michigan, United States
| | - John Y S Han
- Kellogg Eye Center, University of Michigan, United States
| | - Abbi Engel
- Center of Developmental Biology and Regenerative Medicine, Seattle Children's Research Hospital, United States
| | - Jennifer R Chao
- Department of Ophthalmology, University of Washington, United States
| | - Nancy J Philp
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, United States
| | - James B Hurley
- Department of Biochemistry, University of Washington, United States
| | - Jason M L Miller
- Kellogg Eye Center, University of Michigan, United States; Cellular and Molecular Biology Program, University of Michigan, United States.
| |
Collapse
|
3
|
Goswami MT, Weh E, Subramanya S, Weh KM, Durumutla HB, Hager H, Miller N, Chaudhury S, Andren A, Sajjakulnukit P, Zhang L, Besirli CG, Lyssiotis CA, Wubben TJ. Glutamine catabolism supports amino acid biosynthesis and suppresses the integrated stress response to promote photoreceptor survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.26.582525. [PMID: 38586045 PMCID: PMC10996599 DOI: 10.1101/2024.03.26.582525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Photoreceptor loss results in vision loss in many blinding diseases, and metabolic dysfunction underlies photoreceptor degeneration. So, exploiting photoreceptor metabolism is an attractive strategy to prevent vision loss. Yet, the metabolic pathways that maintain photoreceptor health remain largely unknown. Here, we investigated the dependence of photoreceptors on Gln catabolism. Gln is converted to glutamate via glutaminase (GLS), so mice lacking GLS in rod photoreceptors were generated to inhibit Gln catabolism. Loss of GLS produced rapid rod photoreceptor degeneration. In vivo metabolomic methodologies and metabolic supplementation identified Gln catabolism as critical for glutamate and aspartate biosynthesis. Concordant with this amino acid deprivation, the integrated stress response (ISR) was activated with protein synthesis attenuation, and inhibiting the ISR delayed photoreceptor loss. Furthermore, supplementing asparagine, which is synthesized from aspartate, delayed photoreceptor degeneration. Hence, Gln catabolism is integral to photoreceptor health, and these data reveal a novel metabolic axis in these metabolically-demanding neurons.
Collapse
Affiliation(s)
- Moloy T. Goswami
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
- equal contribution
| | - Eric Weh
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
- equal contribution
| | - Shubha Subramanya
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Katherine M. Weh
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Hima Bindu Durumutla
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
- Molecular and Developmental Biology Graduate Program, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Heather Hager
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas Miller
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Sraboni Chaudhury
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Anthony Andren
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Peter Sajjakulnukit
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Li Zhang
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Cagri G. Besirli
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Costas A. Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Thomas J. Wubben
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Hass DT, Pandey K, Engel A, Horton N, Robbings BM, Lim R, Sadilek M, Zhang Q, Autterson GA, Miller JML, Chao JR, Hurley JB. Acetyl-CoA carboxylase Inhibition increases RPE cell fatty acid oxidation and limits apolipoprotein efflux. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566117. [PMID: 37986876 PMCID: PMC10659357 DOI: 10.1101/2023.11.07.566117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Purpose In age-related macular degeneration (AMD) and Sorsby's fundus dystrophy (SFD), lipid-rich deposits known as drusen accumulate under the retinal pigment epithelium (RPE). Drusen may contribute to photoreceptor and RPE degeneration in AMD and SFD. We hypothesize that stimulating β-oxidation in RPE will reduce drusen accumulation. Inhibitors of acetyl-CoA carboxylase (ACC) stimulate β-oxidation and diminish lipid accumulation in fatty liver disease. In this report we test the hypothesis that an ACC inhibitor, Firsocostat, limits the accumulation of lipid deposits in cultured RPE cells. Methods We probed metabolism and cellular function in mouse RPE-choroid, human fetal- derived RPE cells, and induced pluripotent stem cell-derived RPE cells. We used 13 C6-glucose and 13 C16-palmitate to determine the effects of Firsocostat on glycolytic, Krebs cycle, and fatty acid metabolism. 13 C labeling of metabolites in these pathways were analyzed using gas chromatography-linked mass spectrometry. We quantified ApoE and VEGF release using enzyme-linked immunosorbent assays. Immunostaining of sectioned RPE was used to visualize ApoE deposits. RPE function was assessed by measuring the trans-epithelial electrical resistance (TEER). Results ACC inhibition with Firsocostat increases fatty acid oxidation and remodels lipid composition, glycolytic metabolism, lipoprotein release, and enhances TEER. When human serum is used to induce sub-RPE lipoprotein accumulation, fewer lipoproteins accumulate with Firsocostat. In a culture model of Sorsby's fundus dystrophy, Firsocostat also stimulates fatty acid oxidation, improves morphology, and increases TEER. Conclusions Firsocostat remodels intracellular metabolism and improves RPE resilience to serum-induced lipid deposition. This effect of ACC inhibition suggests that it could be an effective strategy for diminishing drusen accumulation in the eyes of patients with AMD.
Collapse
|
5
|
Saravanan M, Xu R, Roby O, Wang Y, Zhu S, Lu A, Du J. Tissue-Specific Sex Difference in Mouse Eye and Brain Metabolome Under Fed and Fasted States. Invest Ophthalmol Vis Sci 2023; 64:18. [PMID: 36892534 PMCID: PMC10010444 DOI: 10.1167/iovs.64.3.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/13/2023] [Indexed: 03/10/2023] Open
Abstract
Purpose Visual physiology and various ocular diseases demonstrate sexual dimorphisms; however, how sex influences metabolism in different eye tissues remains undetermined. This study aims to address common and tissue-specific sex differences in metabolism in the retina, RPE, lens, and brain under fed and fasted conditions. Methods After ad libitum fed or being deprived of food for 18 hours, mouse eye tissues (retina, RPE/choroid, and lens), brain, and plasma were harvested for targeted metabolomics. The data were analyzed with both partial least squares-discriminant analysis and volcano plot analysis. Results Among 133 metabolites that cover major metabolic pathways, we found 9 to 45 metabolites that are sex different in different tissues under the fed state and 6 to 18 metabolites under the fasted state. Among these sex-different metabolites, 33 were changed in 2 or more tissues, and 64 were tissue specific. Pantothenic acid, hypotaurine, and 4-hydroxyproline were the top commonly changed metabolites. The lens and the retina had the most tissue-specific, sex-different metabolites enriched in the metabolism of amino acid, nucleotide, lipids, and tricarboxylic acid cycle. The lens and the brain had more similar sex-different metabolites than other ocular tissues. The female RPE and female brain were more sensitive to fasting with more decreased metabolites in amino acid metabolism, tricarboxylic acid cycles, and glycolysis. The plasma had the fewest sex-different metabolites, with very few overlapping changes with tissues. Conclusions Sex has a strong influence on eye and brain metabolism in tissue-specific and metabolic state-specific manners. Our findings may implicate the sexual dimorphisms in eye physiology and susceptibility to ocular diseases.
Collapse
Affiliation(s)
- Meghashri Saravanan
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Rong Xu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Olivia Roby
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Siyan Zhu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Amy Lu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, West Virginia, United States
- Department of Biochemistry and Molecular Medicine, West Virginia University, Morgantown, West Virginia, United States
| |
Collapse
|
6
|
Cui X, Chang YJ, Jenny LA, Levi SR, Du J, Tsang SH. Metabolite Extraction from RPE Cells and Retinas Related to Retinitis Pigmentosa. Methods Mol Biol 2022; 2560:257-265. [PMID: 36481902 DOI: 10.1007/978-1-0716-2651-1_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The application of metabolomics in ophthalmology helps to identify new biomarkers and elucidate disease mechanisms in different eye diseases, as well as aiding in the development of potential treatment options. Extracting metabolites successfully is essential for potential further analysis using mass spectrometry. In this chapter, we describe how to extract metabolites from a variety of sources including (1) cells on a dish, (2) cell culture medium, and (3) tissues in vivo with and without stable isotope tracers. Samples prepared using this protocol are suitable for a range of downstream mass spectrometry analyses and are stable in solvent for weeks at -80 °C.
Collapse
Affiliation(s)
- Xuan Cui
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York, NY, USA
| | - Ya-Ju Chang
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York, NY, USA
| | - Laura A Jenny
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York, NY, USA
| | - Sarah R Levi
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center, New York, NY, USA.,Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, New York, NY, USA
| | - Jianhai Du
- Department of Ophthalmology, Department of Biochemistry, West Virginia University, Morgantown, WV, USA
| | - Stephen H Tsang
- Departments of Ophthalmology, Pathology & Cell Biology, Graduate Programs in Nutritional & Metabolic Biology and Neurobiology & Behavior, Columbia Stem Cell Initiative, New York, NY, USA.
| |
Collapse
|
7
|
Ross BX, Jia L, Kong D, Wang T, Yao J, Hager HM, Abcouwer SF, Zacks DN. Hypoxia-Inducible Factor-1α in Rods Is Neuroprotective Following Retinal Detachment. Invest Ophthalmol Vis Sci 2022; 63:7. [PMID: 36223101 PMCID: PMC9583748 DOI: 10.1167/iovs.63.11.7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/22/2022] [Indexed: 02/02/2023] Open
Abstract
Purpose Following retinal detachment (RD) photoreceptors (PRs) sustain hypoxic stress and eventually die. Hypoxia-inducible factor-1α (HIF-1α) plays a central role in cellular adaptation to hypoxia. The purpose of this study is to determine the necessity of HIF-1α on PR cell survival after RD. Methods Experimental RD was created in mice by injection of hyaluronic acid (1%) into the subretinal space. Mice with conditional HIF-1α knockout in rods (denoted as HIF-1αΔrod) were used. HIF-1α expression in retinas was measured real-time polymerase chain reaction (RT-PCR) and Western blotting. PR cell death after RD was evaluated using TUNEL assay. Optical coherence tomography (OCT) and histology were used to evaluate retinal layer thicknesses and PR cell densities. A hypoxia signaling pathway PCR array was used to examine the expression of HIF-1α target genes after RD. Results HIF-1α protein levels were significantly increased after RD, and depletion of HIF-1α in rods blunted this increase. A compensatory increase of HIF-2α protein was observed in HIF-1αΔrod mice. Conditional knockout (cKO) of HIF-1α in rods did not lead to any morphologic change in attached retinas but resulted in significantly increased PR cell loss after RD. HIF-1α cKO in rods altered the responses to retinal detachment for 25 out of 83 HIF-1α target genes that were highly enriched for genes involved in glycolysis. Conclusions Rod-derived HIF-1α plays a key role in the PR response to RD, mediating the transcriptional activity of a battery of genes to promote PR cell survival.
Collapse
Affiliation(s)
- Bing X. Ross
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Lin Jia
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Dejuan Kong
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Tiantian Wang
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
| | - Jingyu Yao
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Heather M. Hager
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - Steven F. Abcouwer
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| | - David N. Zacks
- Department of Ophthalmology, University of Michigan Medical School, Kellogg Eye Center, Ann Arbor, Michigan, United States
| |
Collapse
|
8
|
Zhao X, Tebbe L, Naash MI, Al-Ubaidi MR. The Neuroprotective Role of Retbindin, a Metabolic Regulator in the Neural Retina. Front Pharmacol 2022; 13:919667. [PMID: 35873559 PMCID: PMC9298789 DOI: 10.3389/fphar.2022.919667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of retinal metabolism is emerging as one of the major reasons for many inherited retinal diseases (IRDs), a leading cause of blindness worldwide. Thus, the identification of a common regulator that can preserve or revert the metabolic ecosystem to homeostasis is a key step in developing a treatment for different forms of IRDs. Riboflavin (RF) and its derivatives (flavins), flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), are essential cofactors for a wide range of cellular metabolic processes; hence, they are particularly critical in highly metabolically active tissues such as the retina. Patients with RF deficiency (ariboflavinosis) often display poor photosensitivity resulting in impaired low-light vision. We have identified a novel retina-specific RF binding protein called retbindin (Rtbdn), which plays a key role in retaining flavin levels in the neural retina. This role is mediated by its specific localization at the interface between the neural retina and retinal pigment epithelium (RPE), which is essential for metabolite and nutrient exchange. As a consequence of this vital function, Rtbdn's role in flavin utilization and metabolism in retinal degeneration is discussed. The principal findings are that Rtbdn helps maintain high levels of retinal flavins, and its ablation leads to an early-onset retinal metabolic dysregulation, followed by progressive degeneration of rod and cone photoreceptors. Lack of Rtbdn reduces flavin levels, forcing the neural retina to repurpose glucose to reduce the production of free radicals during ATP production. This leads to metabolic breakdown followed by retinal degeneration. Assessment of the role of Rtbdn in several preclinical retinal disease models revealed upregulation of its levels by several folds prior to and during the degenerative process. Ablation of Rtbdn in these models accelerated the rate of retinal degeneration. In agreement with these in vivo studies, we have also demonstrated that Rtbdn protects immortalized cone photoreceptor cells (661W cells) from light damage in vitro. This indicates that Rtbdn plays a neuroprotective role during retinal degeneration. Herein, we discussed the specific function of Rtbdn and its neuroprotective role in retinal metabolic homeostasis and its role in maintaining retinal health.
Collapse
Affiliation(s)
| | | | - Muna I. Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| | - Muayyad R. Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX, United States
| |
Collapse
|
9
|
Hass DT, Bisbach CM, Robbings BM, Sadilek M, Sweet IR, Hurley JB. Succinate metabolism in the retinal pigment epithelium uncouples respiration from ATP synthesis. Cell Rep 2022; 39:110917. [PMID: 35675773 PMCID: PMC9251713 DOI: 10.1016/j.celrep.2022.110917] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/08/2022] [Accepted: 05/12/2022] [Indexed: 11/28/2022] Open
Abstract
Fumarate can be a surrogate for O2 as a terminal electron acceptor in the electron transport chain. Reduction of fumarate produces succinate, which can be exported. It is debated whether intact tissues can import and oxidize succinate produced by other tissues. In a previous report, we showed that mitochondria in retinal pigment epithelium (RPE)-choroid preparations can use succinate to reduce O2 to H2O. However, cells in that preparation could have been disrupted during tissue isolation. We now use multiple strategies to quantify intactness of the isolated RPE-choroid tissue. We find that exogenous 13C4-succinate is oxidized by intact cells then exported as fumarate or malate. Unexpectedly, we also find that oxidation of succinate is different from oxidation of other substrates because it uncouples electron transport from ATP synthesis. Retinas produce and export succinate. Our findings imply that retina succinate may substantially increase O2 consumption by uncoupling adjacent RPE mitochondria. The retina releases succinate, a source of reducing power for mitochondria. Hass et al. outline a pathway by which retina succinate can enter intact RPE-choroid cells and stimulate mitochondrial respiration that is uncoupled from ATP synthesis. Rapid RPE succinate oxidation may limit O2 levels in the retina.
Collapse
Affiliation(s)
- Daniel T Hass
- Biochemistry Department, The University of Washington, Seattle, WA 98195, USA
| | - Celia M Bisbach
- Biochemistry Department, The University of Washington, Seattle, WA 98195, USA; Promega Corporation, 2800 Woods Hollow Road, Fitchburg, WI 53711, USA
| | - Brian M Robbings
- Biochemistry Department, The University of Washington, Seattle, WA 98195, USA; Diabetes Institute, The University of Washington, Seattle, WA 98109, USA
| | - Martin Sadilek
- Chemistry Department, The University of Washington, Seattle, WA 98195, USA
| | - Ian R Sweet
- Diabetes Institute, The University of Washington, Seattle, WA 98109, USA; Division of Metabolism, Endocrinology and Nutrition, The University of Washington, Seattle, WA 98195, USA
| | - James B Hurley
- Biochemistry Department, The University of Washington, Seattle, WA 98195, USA; Opthalmology Department, The University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
10
|
Chen Y, Coorey NJ, Zhang M, Zeng S, Madigan MC, Zhang X, Gillies MC, Zhu L, Zhang T. Metabolism Dysregulation in Retinal Diseases and Related Therapies. Antioxidants (Basel) 2022; 11:antiox11050942. [PMID: 35624805 PMCID: PMC9137684 DOI: 10.3390/antiox11050942] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 02/08/2023] Open
Abstract
The human retina, which is part of the central nervous system, has exceptionally high energy demands that requires an efficient metabolism of glucose, lipids, and amino acids. Dysregulation of retinal metabolism disrupts local energy supply and redox balance, contributing to the pathogenesis of diverse retinal diseases, including age-related macular degeneration, diabetic retinopathy, inherited retinal degenerations, and Macular Telangiectasia. A better understanding of the contribution of dysregulated metabolism to retinal diseases may provide better therapeutic targets than we currently have.
Collapse
Affiliation(s)
- Yingying Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610017, China;
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia; (S.Z.); (M.C.M.); (M.C.G.); (L.Z.)
| | | | - Meixia Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610017, China;
- Macular Disease Research Laboratory, Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu 610017, China
- Correspondence: (M.Z.); (T.Z.)
| | - Shaoxue Zeng
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia; (S.Z.); (M.C.M.); (M.C.G.); (L.Z.)
| | - Michele C. Madigan
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia; (S.Z.); (M.C.M.); (M.C.G.); (L.Z.)
- School of Optometry and Vision Science, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Xinyuan Zhang
- Department of Ocular Fundus Diseases, Beijing Tongren Eye Centre, Tongren Hospital, Capital Medical University, Beijing 100073, China;
- Beijing Retinal and Choroidal Vascular Study Group, Beijing 100073, China
| | - Mark C. Gillies
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia; (S.Z.); (M.C.M.); (M.C.G.); (L.Z.)
| | - Ling Zhu
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia; (S.Z.); (M.C.M.); (M.C.G.); (L.Z.)
| | - Ting Zhang
- Save Sight Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2000, Australia; (S.Z.); (M.C.M.); (M.C.G.); (L.Z.)
- Correspondence: (M.Z.); (T.Z.)
| |
Collapse
|
11
|
Bisbach CM, Hass DT, Thomas ED, Cherry TJ, Hurley JB. Monocarboxylate Transporter 1 (MCT1) Mediates Succinate Export in the Retina. Invest Ophthalmol Vis Sci 2022; 63:1. [PMID: 35363247 PMCID: PMC8976921 DOI: 10.1167/iovs.63.4.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 03/12/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose Succinate is exported by the retina and imported by eyecup tissue. The transporters mediating this process have not yet been identified. Recent studies showed that monocarboxylate transporter 1 (MCT1) can transport succinate across plasma membranes in cardiac and skeletal muscle. Retina and retinal pigment epithelium (RPE) both express multiple MCT isoforms including MCT1. We tested the hypothesis that MCTs facilitate retinal succinate export and RPE succinate import. Methods We assessed retinal succinate export and eyecup succinate import in short-term ex vivo culture using gas chromatography-mass spectrometry. We tested the dependence of succinate export and import on pH, proton ionophores, conventional MCT substrates, and the MCT inhibitors AZD3965, AR-C155858, and diclofenac. Results Succinate exits retinal tissue through MCT1 but does not enter the RPE through MCT1 or any other MCT. Intracellular succinate levels are a contributing factor that determines if an MCT1-expressing tissue will export succinate. Conclusions MCT1 facilitates export of succinate from retinas. An unidentified, non-MCT transporter facilitates import of succinate into RPE.
Collapse
Affiliation(s)
- Celia M. Bisbach
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Daniel T. Hass
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Eric D. Thomas
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, United States
| | - Timothy J. Cherry
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, Seattle, Washington, United States
| | - James B. Hurley
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| |
Collapse
|
12
|
Effect of the ethyl acetate extract of Sophora flavescens Aiton on diabetic retinopathy based on untargeted retinal metabolomics. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1198:123233. [DOI: 10.1016/j.jchromb.2022.123233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/15/2022] [Accepted: 03/26/2022] [Indexed: 12/22/2022]
|
13
|
Tsantilas KA, Cleghorn WM, Bisbach CM, Whitson JA, Hass DT, Robbings BM, Sadilek M, Linton JD, Rountree AM, Valencia AP, Sweetwyne MT, Campbell MD, Zhang H, Jankowski CSR, Sweet IR, Marcinek DJ, Rabinovitch PS, Hurley JB. An Analysis of Metabolic Changes in the Retina and Retinal Pigment Epithelium of Aging Mice. Invest Ophthalmol Vis Sci 2021; 62:20. [PMID: 34797906 PMCID: PMC8606884 DOI: 10.1167/iovs.62.14.20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose The purpose of this study was to present our hypothesis that aging alters metabolic function in ocular tissues. We tested the hypothesis by measuring metabolism in aged murine tissues alongside retinal responses to light. Methods Scotopic and photopic electroretinogram (ERG) responses in young (3–6 months) and aged (23–26 months) C57Bl/6J mice were recorded. Metabolic flux in retina and eyecup explants was quantified using U-13C-glucose or U-13C-glutamine with gas chromatography-mass spectrometry (GC-MS), O2 consumption rate (OCR) in a perifusion apparatus, and quantifying adenosine triphosphatase (ATP) with a bioluminescence assay. Results Scotopic and photopic ERG responses were reduced in aged mice. Glucose metabolism, glutamine metabolism, OCR, and ATP pools in retinal explants were mostly unaffected in aged mice. In eyecups, glutamine usage in the Krebs Cycle decreased while glucose metabolism, OCR, and ATP pools remained stable. Conclusions Our examination of metabolism showed negligible impact of age on retina and an impairment of glutamine anaplerosis in eyecups. The metabolic stability of these tissues ex vivo suggests age-related metabolic alterations may not be intrinsic. Future experiments should focus on determining whether external factors including nutrient supply, oxygen availability, or structural changes influence ocular metabolism in vivo.
Collapse
Affiliation(s)
- Kristine A Tsantilas
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Whitney M Cleghorn
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Celia M Bisbach
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Jeremy A Whitson
- Department of Biology, Davidson College, Davidson, North Carolina, United States
| | - Daniel T Hass
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Brian M Robbings
- Department of Biochemistry, University of Washington, Seattle, Washington, United States.,UW Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - Martin Sadilek
- Department of Chemistry, University of Washington, Seattle, Washington, United States
| | - Jonathan D Linton
- Department of Biochemistry, University of Washington, Seattle, Washington, United States
| | - Austin M Rountree
- UW Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - Ana P Valencia
- Department of Radiology, University of Washington, Seattle, Washington, United States
| | - Mariya T Sweetwyne
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington, United States
| | - Matthew D Campbell
- Department of Radiology, University of Washington, Seattle, Washington, United States
| | - Huiliang Zhang
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States
| | - Connor S R Jankowski
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, United States.,Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey, United States
| | - Ian R Sweet
- UW Diabetes Institute, University of Washington, Seattle, Washington, United States
| | - David J Marcinek
- Department of Radiology, University of Washington, Seattle, Washington, United States
| | - Peter S Rabinovitch
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington, United States
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, Washington, United States.,Department of Ophthalmology, University of Washington, Seattle, Washington, United States
| |
Collapse
|
14
|
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: 2.7] [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
|
15
|
Bisbach CM, Hass DT, Robbings BM, Rountree AM, Sadilek M, Sweet IR, Hurley JB. Succinate Can Shuttle Reducing Power from the Hypoxic Retina to the O 2-Rich Pigment Epithelium. Cell Rep 2021; 31:107606. [PMID: 32375026 DOI: 10.1016/j.celrep.2020.107606] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/21/2020] [Accepted: 04/10/2020] [Indexed: 12/19/2022] Open
Abstract
When O2 is plentiful, the mitochondrial electron transport chain uses it as a terminal electron acceptor. However, the mammalian retina thrives in a hypoxic niche in the eye. We find that mitochondria in retinas adapt to their hypoxic environment by reversing the succinate dehydrogenase reaction to use fumarate to accept electrons instead of O2. Reverse succinate dehydrogenase activity produces succinate and is enhanced by hypoxia-induced downregulation of cytochrome oxidase. Retinas can export the succinate they produce to the neighboring O2-rich retinal pigment epithelium-choroid complex. There, succinate enhances O2 consumption by severalfold. Malate made from succinate in the pigment epithelium can then be imported into the retina, where it is converted to fumarate to again accept electrons in the reverse succinate dehydrogenase reaction. This malate-succinate shuttle can sustain these two tissues by transferring reducing power from an O2-poor tissue (retina) to an O2-rich one (retinal pigment epithelium-choroid).
Collapse
Affiliation(s)
- Celia M Bisbach
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Daniel T Hass
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Brian M Robbings
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; UW Diabetes Institute, University of Washington, Seattle, WA 98195, USA
| | - Austin M Rountree
- UW Diabetes Institute, University of Washington, Seattle, WA 98195, USA
| | - Martin Sadilek
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Ian R Sweet
- UW Diabetes Institute, University of Washington, Seattle, WA 98195, USA
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Department of Ophthalmology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
16
|
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: 4.0] [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
|
17
|
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: 18] [Impact Index Per Article: 6.0] [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
|
18
|
Xu R, Wang Y, Du J. Tracing Nitrogen Metabolism in Mouse Tissues with Gas Chromatography-Mass Spectrometry. Bio Protoc 2021; 11:e3925. [PMID: 33732812 DOI: 10.21769/bioprotoc.3925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/30/2020] [Accepted: 01/06/2021] [Indexed: 11/02/2022] Open
Abstract
Nitrogen-containing metabolites including ammonia, amino acids, and nucleotides, are essential for cell metabolism, growth, and neural transmission. Nitrogen metabolism is tightly coordinated with carbon metabolism in the breakdown and biosynthesis of amino acids and nucleotides. Both nuclear magnetic resonance spectroscopy and mass spectrometry including gas chromatography-mass spectrometry (GC MS) and liquid chromatography (LC MS) have been used to measure nitrogen metabolism. Here we describe a protocol to trace nitrogen metabolism in multiple mouse tissues using 15N-ammonia coupled with GC MS. This protocol includes detailed procedures in tracer injection, tissue preparation, metabolite extraction, GC MS analysis and natural abundance corrections. This protocol will provide a useful tool to study tissue-specific nitrogen in metabolically active tissues such as the retina, brain, liver, and tumor.
Collapse
Affiliation(s)
- Rong Xu
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, USA.,Department of Biochemistry, West Virginia University, Morgantown, USA
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, USA.,Department of Biochemistry, West Virginia University, Morgantown, USA
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West Virginia University, Morgantown, USA.,Department of Biochemistry, West Virginia University, Morgantown, USA
| |
Collapse
|
19
|
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: 50] [Impact Index Per Article: 16.7] [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
|
20
|
Selective knockdown of hexokinase 2 in rods leads to age-related photoreceptor degeneration and retinal metabolic remodeling. Cell Death Dis 2020; 11:885. [PMID: 33082308 PMCID: PMC7576789 DOI: 10.1038/s41419-020-03103-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022]
Abstract
Photoreceptors, the primary site of phototransduction in the retina, require energy and metabolites to constantly renew their outer segments. They preferentially consume most glucose through aerobic glycolysis despite possessing abundant mitochondria and enzymes for oxidative phosphorylation (OXPHOS). Exactly how photoreceptors balance aerobic glycolysis and mitochondrial OXPHOS to regulate their survival is still unclear. We crossed rhodopsin-Cre mice with hexokinase 2 (HK2)-floxed mice to study the effect of knocking down HK2, the first rate-limiting enzyme in glycolysis, on retinal health and metabolic remodeling. Immunohistochemistry and Western blots were performed to study changes in photoreceptor-specific proteins and key enzymes in glycolysis and the tricarboxylic acid (TCA) cycle. Changes in retinal structure and function were studied by optical coherence tomography and electroretinography. Mass spectrometry was performed to profile changes in 13C-glucose-derived metabolites in glycolysis and the TCA cycle. We found that knocking down HK2 in rods led to age-related photoreceptor degeneration, evidenced by reduced expression of photoreceptor-specific proteins, age-related reductions of the outer nuclear layer, photoreceptor inner and outer segments and impaired electroretinographic responses. Loss of HK2 in rods led to upregulation of HK1, phosphorylation of pyruvate kinase muscle isozyme 2, mitochondrial stress proteins and enzymes in the TCA cycle. Mass spectrometry found that the deletion of HK2 in rods resulted in accumulation of 13C-glucose along with decreased pyruvate and increased metabolites in the TCA cycle. Our data suggest that HK2-mediated aerobic glycolysis is indispensable for the maintenance of photoreceptor structure and function and that long-term inhibition of glycolysis leads to photoreceptor degeneration.
Collapse
|
21
|
Zhang R, Engel AL, Wang Y, Li B, Shen W, Gillies MC, Chao JR, Du J. Inhibition of Mitochondrial Respiration Impairs Nutrient Consumption and Metabolite Transport in Human Retinal Pigment Epithelium. J Proteome Res 2020; 20:909-922. [PMID: 32975122 DOI: 10.1021/acs.jproteome.0c00690] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Mitochondrial respiration in mammalian cells not only generates ATP to meet their own energy needs but also couples with biosynthetic pathways to produce metabolites that can be exported to support neighboring cells. However, how defects in mitochondrial respiration influence these biosynthetic and exporting pathways remains poorly understood. Mitochondrial dysfunction in retinal pigment epithelium (RPE) cells is an emerging contributor to the death of their neighboring photoreceptors in degenerative retinal diseases including age-related macular degeneration. In this study, we used targeted-metabolomics and 13C tracing to investigate how inhibition of mitochondrial respiration influences the intracellular and extracellular metabolome. We found inhibition of mitochondrial respiration strikingly influenced both the intracellular and extracellular metabolome in primary RPE cells. Intriguingly, the extracellular metabolic changes sensitively reflected the intracellular changes. These changes included substantially enhanced glucose consumption and lactate production; reduced release of pyruvate, citrate, and ketone bodies; and massive accumulation of multiple amino acids and nucleosides. In conclusion, these findings reveal a metabolic signature of nutrient consumption and release in mitochondrial dysfunction in RPE cells. Testing medium metabolites provides a sensitive and noninvasive method to assess mitochondrial function in nutrient utilization and transport.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506, United States.,Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States.,Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Abbi L Engel
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109, United States
| | - Yekai Wang
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506, United States.,Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Bo Li
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506, United States.,Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| | - Weiyong Shen
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Mark C Gillies
- Save Sight Institute, Sydney Medical School, University of Sydney, Sydney, NSW 2000, Australia
| | - Jennifer R Chao
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109, United States
| | - Jianhai Du
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506, United States.,Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506, United States
| |
Collapse
|
22
|
Meschede IP, Ovenden NC, Seabra MC, Futter CE, Votruba M, Cheetham ME, Burgoyne T. Symmetric arrangement of mitochondria:plasma membrane contacts between adjacent photoreceptor cells regulated by Opa1. Proc Natl Acad Sci U S A 2020; 117:15684-15693. [PMID: 32571921 PMCID: PMC7355040 DOI: 10.1073/pnas.2000304117] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Mitochondria are known to play an essential role in photoreceptor function and survival that enables normal vision. Within photoreceptors, mitochondria are elongated and extend most of the inner-segment length, where they supply energy for protein synthesis and the phototransduction machinery in the outer segment, as well as acting as a calcium store. Here, we examined the arrangement of the mitochondria within the inner segment in detail using three-dimensional (3D) electron microscopy techniques and show they are tethered to the plasma membrane in a highly specialized arrangement. Remarkably, mitochondria and their cristae openings align with those of neighboring inner segments. The pathway by which photoreceptors meet their high energy demands is not fully understood. We propose this to be a mechanism to share metabolites and assist in maintaining homeostasis across the photoreceptor cell layer. In the extracellular space between photoreceptors, Müller glial processes were identified. Due to the often close proximity to the inner-segment mitochondria, they may, too, play a role in the inner-segment mitochondrial arrangement as well as metabolite shuttling. OPA1 is an important factor in mitochondrial homeostasis, including cristae remodeling; therefore, we examined the photoreceptors of a heterozygous Opa1 knockout mouse model. The cristae structure in the Opa1+/- photoreceptors was not greatly affected, but the mitochondria were enlarged and had reduced alignment to neighboring inner-segment mitochondria. This indicates the importance of key regulators in maintaining this specialized photoreceptor mitochondrial arrangement.
Collapse
Affiliation(s)
- Ingrid P Meschede
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, United Kingdom
| | - Nicholas C Ovenden
- Department of Mathematics, University College London, WC1E 6BT London, United Kingdom
| | - Miguel C Seabra
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, United Kingdom
- Centro de Estudos de Doenças Crónicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Clare E Futter
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, United Kingdom
| | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, CF24 4HQ Cardiff, United Kingdom
- Cardiff Eye Unit, University Hospital Wales, CF14 4XW Cardiff, United Kingdom
| | - Michael E Cheetham
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, United Kingdom
| | - Thomas Burgoyne
- UCL Institute of Ophthalmology, University College London, EC1V 9EL London, United Kingdom;
| |
Collapse
|
23
|
Luo Y, Cui HP, Liu Y, Chen L. Metabolomics and biomarkers in ocular matrix: beyond ocular diseases. Int J Ophthalmol 2020; 13:991-1003. [PMID: 32566514 DOI: 10.18240/ijo.2020.06.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 03/23/2020] [Indexed: 12/15/2022] Open
Abstract
According to the recent report, there are 870 million people suffer from ocular diseases worldwide. The present approaches for diagnosis are morphological examination, imaging examination and immunological examination, regrettably, they lack of sensitivity and difficult to make a definite diagnosis in the early stage. Systemic biology as an effective method has been used in clinical diagnosis and treatment for diseases, especially metabolomics which is more attractive with high sensitivity and accuracy. Although previous researches had been confirmed that endogenous metabolites in the ocular matrix play a crucial role in the progress of diseases related diseases, the standard protocols and systematic summary about the biomarker researches based on ocular matrix has not been established. This review article highlights the pretreatment for ocular matrix and the new biomarkers expressed by the eye diseases, expected to promote the application of biomarkers in the diagnosis and treatment of eye diseases.
Collapse
Affiliation(s)
- Yun Luo
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong Province, China
| | - Hong-Pei Cui
- Department of Ophthalmology, Henan Provincial People's Hospital, Zhengzhou 450003, Henan Province, China
| | - Yi Liu
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Lei Chen
- School of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong Province, China
| |
Collapse
|
24
|
Wubben TJ, Pawar M, Weh E, Smith A, Sajjakulnukit P, Zhang L, Dai L, Hager H, Pai MP, Lyssiotis CA, Besirli CG. Small molecule activation of metabolic enzyme pyruvate kinase muscle isozyme 2, PKM2, circumvents photoreceptor apoptosis. Sci Rep 2020; 10:2990. [PMID: 32076076 PMCID: PMC7031539 DOI: 10.1038/s41598-020-59999-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 02/06/2020] [Indexed: 01/22/2023] Open
Abstract
Photoreceptor cell death is the ultimate cause of vision loss in many retinal disorders, and there is an unmet need for neuroprotective modalities to improve photoreceptor survival. Similar to cancer cells, photoreceptors maintain pyruvate kinase muscle isoform 2 (PKM2) expression, which is a critical regulator in aerobic glycolysis. Unlike PKM1, which has constitutively high catalytic activity, PKM2 is under complex regulation. Recently, we demonstrated that genetically reprogramming photoreceptor metabolism via PKM2-to-PKM1 substitution is a promising neuroprotective strategy. Here, we explored the neuroprotective effects of pharmacologically activating PKM2 via ML-265, a small molecule activator of PKM2, during acute outer retinal stress. We found that ML-265 increased PKM2 activity in 661 W cells and in vivo in rat eyes without affecting the expression of genes involved in glucose metabolism. ML-265 treatment did, however, alter metabolic intermediates of glucose metabolism and those necessary for biosynthesis in cultured cells. Long-term exposure to ML-265 did not result in decreased photoreceptor function or survival under baseline conditions. Notably, though, ML-265-treatment did reduce entrance into the apoptotic cascade in in vitro and in vivo models of outer retinal stress. These data suggest that reprogramming metabolism via activation of PKM2 is a novel, and promising, therapeutic strategy for photoreceptor neuroprotection.
Collapse
Affiliation(s)
- Thomas J Wubben
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | - Mercy Pawar
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | - Eric Weh
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | - Andrew Smith
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | - Peter Sajjakulnukit
- University of Michigan, Department of Molecular and Integrative Physiology, Ann Arbor, USA
| | - Li Zhang
- University of Michigan Biomedical Research Core Facilities, Metabolomics Core, Ann Arbor, USA
| | - Lipeng Dai
- University of Michigan, College of Pharmacy, Ann Arbor, USA
| | - Heather Hager
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA
| | | | - Costas A Lyssiotis
- University of Michigan, Department of Molecular and Integrative Physiology, Ann Arbor, USA
| | - Cagri G Besirli
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, USA.
| |
Collapse
|
25
|
Xu R, Ritz BK, Wang Y, Huang J, Zhao C, Gong K, Liu X, Du J. The retina and retinal pigment epithelium differ in nitrogen metabolism and are metabolically connected. J Biol Chem 2020; 295:2324-2335. [PMID: 31953322 DOI: 10.1074/jbc.ra119.011727] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/15/2020] [Indexed: 01/07/2023] Open
Abstract
Defects in energy metabolism in either the retina or the immediately adjacent retinal pigment epithelium (RPE) underlie retinal degeneration, but the metabolic dependence between retina and RPE remains unclear. Nitrogen-containing metabolites such as amino acids are essential for energy metabolism. Here, we found that 15N-labeled ammonium is predominantly assimilated into glutamine in both the retina and RPE/choroid ex vivo [15N]Ammonium tracing in vivo show that, like the brain, the retina can synthesize asparagine from ammonium, but RPE/choroid and the liver cannot. However, unless present at toxic concentrations, ammonium cannot be recycled into glutamate in the retina and RPE/choroid. Tracing with 15N-labeled amino acids show that the retina predominantly uses aspartate transaminase for de novo synthesis of glutamate, glutamine, and aspartate, whereas RPE uses multiple transaminases to utilize and synthesize amino acids. Retina consumes more leucine than RPE, but little leucine is catabolized. The synthesis of serine and glycine is active in RPE but limited in the retina. RPE, but not the retina, uses alanine as mitochondrial substrates through mitochondrial pyruvate carrier. However, when the mitochondrial pyruvate carrier is inhibited, alanine may directly enter the retinal mitochondria but not those of RPE. In conclusion, our results demonstrate that the retina and RPE differ in nitrogen metabolism and highlight that the RPE supports retinal metabolism through active amino acid metabolism.
Collapse
Affiliation(s)
- Rong Xu
- Central Laboratory, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012 China; Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506; Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Brianna K Ritz
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506; Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Yekai Wang
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506; Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Jiancheng Huang
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506; Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506; Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433 China
| | - Chen Zhao
- Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, 200433 China
| | - Kaizheng Gong
- Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012 China
| | - Xinnong Liu
- Department of General Surgery, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012; Department of Cardiology, the Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012 China.
| | - Jianhai Du
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506; Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506.
| |
Collapse
|
26
|
Blond F, Léveillard T. Functional Genomics of the Retina to Elucidate its Construction and Deconstruction. Int J Mol Sci 2019; 20:E4922. [PMID: 31590277 PMCID: PMC6801968 DOI: 10.3390/ijms20194922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022] Open
Abstract
The retina is the light sensitive part of the eye and nervous tissue that have been used extensively to characterize the function of the central nervous system. The retina has a central position both in fundamental biology and in the physiopathology of neurodegenerative diseases. We address the contribution of functional genomics to the understanding of retinal biology by reviewing key events in their historical perspective as an introduction to major findings that were obtained through the study of the retina using genomics, transcriptomics and proteomics. We illustrate our purpose by showing that most of the genes of interest for retinal development and those involved in inherited retinal degenerations have a restricted expression to the retina and most particularly to photoreceptors cells. We show that the exponential growth of data generated by functional genomics is a future challenge not only in terms of storage but also in terms of accessibility to the scientific community of retinal biologists in the future. Finally, we emphasize on novel perspectives that emerge from the development of redox-proteomics, the new frontier in retinal biology.
Collapse
Affiliation(s)
- Frédéric Blond
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| | - Thierry Léveillard
- Department of Genetics, Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.
| |
Collapse
|
27
|
Fu Z, Chen CT, Cagnone G, Heckel E, Sun Y, Cakir B, Tomita Y, Huang S, Li Q, Britton W, Cho SS, Kern TS, Hellström A, Joyal JS, Smith LE. Dyslipidemia in retinal metabolic disorders. EMBO Mol Med 2019; 11:e10473. [PMID: 31486227 PMCID: PMC6783651 DOI: 10.15252/emmm.201910473] [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: 02/15/2019] [Revised: 06/10/2019] [Accepted: 08/15/2019] [Indexed: 12/24/2022] Open
Abstract
The light‐sensitive photoreceptors in the retina are extremely metabolically demanding and have the highest density of mitochondria of any cell in the body. Both physiological and pathological retinal vascular growth and regression are controlled by photoreceptor energy demands. It is critical to understand the energy demands of photoreceptors and fuel sources supplying them to understand neurovascular diseases. Retinas are very rich in lipids, which are continuously recycled as lipid‐rich photoreceptor outer segments are shed and reformed and dietary intake of lipids modulates retinal lipid composition. Lipids (as well as glucose) are fuel substrates for photoreceptor mitochondria. Dyslipidemia contributes to the development and progression of retinal dysfunction in many eye diseases. Here, we review photoreceptor energy demands with a focus on lipid metabolism in retinal neurovascular disorders.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.,Manton Center for Orphan Disease, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Chuck T Chen
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Gael Cagnone
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada.,Department of Pharmacology and Therapeutics, University of Montreal, Montreal, QC, Canada
| | - Emilie Heckel
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada.,Department of Pharmacology and Therapeutics, University of Montreal, Montreal, QC, Canada
| | - Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Bertan Cakir
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Yohei Tomita
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Shuo Huang
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Qian Li
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - William Britton
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Steve S Cho
- Department of Ophthalmology, Harvard Medical School, 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
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada.,Department of Pharmacology and Therapeutics, University of Montreal, Montreal, QC, Canada
| | - Lois Eh Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
28
|
Hutto RA, Bisbach CM, Abbas F, Brock DC, Cleghorn WM, Parker ED, Bauer BH, Ge W, Vinberg F, Hurley JB, Brockerhoff SE. Increasing Ca 2+ in photoreceptor mitochondria alters metabolites, accelerates photoresponse recovery, and reveals adaptations to mitochondrial stress. Cell Death Differ 2019; 27:1067-1085. [PMID: 31371786 PMCID: PMC7206026 DOI: 10.1038/s41418-019-0398-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 06/19/2019] [Accepted: 07/11/2019] [Indexed: 11/09/2022] Open
Abstract
Photoreceptors are specialized neurons that rely on Ca2+ to regulate phototransduction and neurotransmission. Photoreceptor dysfunction and degeneration occur when intracellular Ca2+ homeostasis is disrupted. Ca2+ homeostasis is maintained partly by mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniporter (MCU), which can influence cytosolic Ca2+ signals, stimulate energy production, and trigger apoptosis. Here we discovered that zebrafish cone photoreceptors express unusually low levels of MCU. We expected that this would be important to prevent mitochondrial Ca2+ overload and consequent cone degeneration. To test this hypothesis, we generated a cone-specific model of MCU overexpression. Surprisingly, we found that cones tolerate MCU overexpression, surviving elevated mitochondrial Ca2+ and disruptions to mitochondrial ultrastructure until late adulthood. We exploited the survival of MCU overexpressing cones to additionally demonstrate that mitochondrial Ca2+ uptake alters the distributions of citric acid cycle intermediates and accelerates recovery kinetics of the cone response to light. Cones adapt to mitochondrial Ca2+ stress by decreasing MICU3, an enhancer of MCU-mediated Ca2+ uptake, and selectively transporting damaged mitochondria away from the ellipsoid toward the synapse. Our findings demonstrate how mitochondrial Ca2+ can influence physiological and metabolic processes in cones and highlight the remarkable ability of cone photoreceptors to adapt to mitochondrial stress.
Collapse
Affiliation(s)
- Rachel A Hutto
- Biochemistry Department, University of Washington, Seattle, WA, 98109, USA
| | - Celia M Bisbach
- Biochemistry Department, University of Washington, Seattle, WA, 98109, USA
| | - Fatima Abbas
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, 84132, USA
| | - Daniel C Brock
- Biochemistry Department, University of Washington, Seattle, WA, 98109, USA
| | - Whitney M Cleghorn
- Biochemistry Department, University of Washington, Seattle, WA, 98109, USA
| | - Edward D Parker
- Opthalmology Department, University of Washington, Seattle, WA, 98109, USA
| | - Benjamin H Bauer
- Biochemistry Department, University of Washington, Seattle, WA, 98109, USA
| | - William Ge
- Biochemistry Department, University of Washington, Seattle, WA, 98109, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, 84132, USA
| | - James B Hurley
- Biochemistry Department, University of Washington, Seattle, WA, 98109, USA.,Opthalmology Department, University of Washington, Seattle, WA, 98109, USA
| | - Susan E Brockerhoff
- Biochemistry Department, University of Washington, Seattle, WA, 98109, USA. .,Opthalmology Department, University of Washington, Seattle, WA, 98109, USA.
| |
Collapse
|
29
|
Singh C, Hoppe G, Tran V, McCollum L, Bolok Y, Song W, Sharma A, Brunengraber H, Sears JE. Serine and 1-carbon metabolism are required for HIF-mediated protection against retinopathy of prematurity. JCI Insight 2019; 4:129398. [PMID: 31341109 DOI: 10.1172/jci.insight.129398] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
We determined which metabolic pathways are activated by hypoxia-inducible factor 1-mediated (HIF-1-mediated) protection against oxygen-induced retinopathy (OIR) in newborn mice, the experimental correlate to retinopathy of prematurity, a leading cause of infant blindness. HIF-1 coordinates the change from oxidative to glycolytic metabolism and mediates flux through serine and 1-carbon metabolism (1CM) in hypoxic and cancer cells. We used untargeted metabolite profiling in vivo to demonstrate that hypoxia mimesis activates serine/1CM. Both [13C6] glucose labeling of metabolites in ex vivo retinal explants as well as in vivo [13C3] serine labeling of metabolites followed in liver lysates strongly suggest that retinal serine is primarily derived from hepatic glycolytic carbon and not from retinal glycolytic carbon in newborn pups. In HIF-1α2lox/2lox albumin-Cre-knockout mice, reduced or near-0 levels of serine/glycine further demonstrate the hepatic origin of retinal serine. Furthermore, inhibition of 1CM by methotrexate blocked HIF-mediated protection against OIR. This demonstrated that 1CM participates in protection induced by HIF-1 stabilization. The urea cycle also dominated pathway enrichment analyses of plasma samples. The dependence of retinal serine on hepatic HIF-1 and the upregulation of the urea cycle emphasize the importance of the liver to remote protection of the retina.
Collapse
Affiliation(s)
| | - George Hoppe
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Vincent Tran
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Leah McCollum
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Youstina Bolok
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Weilin Song
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Amit Sharma
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Henri Brunengraber
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jonathan E Sears
- Cole Eye Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
30
|
Yam M, Engel AL, Wang Y, Zhu S, Hauer A, Zhang R, Lohner D, Huang J, Dinterman M, Zhao C, Chao JR, Du J. Proline mediates metabolic communication between retinal pigment epithelial cells and the retina. J Biol Chem 2019; 294:10278-10289. [PMID: 31110046 PMCID: PMC6664195 DOI: 10.1074/jbc.ra119.007983] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/16/2019] [Indexed: 01/16/2023] Open
Abstract
The retinal pigment epithelium (RPE) is a monolayer of pigmented cells between the choroid and the retina. RPE dysfunction underlies many retinal degenerative diseases, including age-related macular degeneration, the leading cause of age-related blindness. To perform its various functions in nutrient transport, phagocytosis of the outer segment, and cytokine secretion, the RPE relies on an active energy metabolism. We previously reported that human RPE cells prefer proline as a nutrient and transport proline-derived metabolites to the apical, or retinal, side. In this study, we investigated how RPE utilizes proline in vivo and why proline is a preferred substrate. By using [13C]proline labeling both ex vivo and in vivo, we found that the retina rarely uses proline directly, whereas the RPE utilizes it at a high rate, exporting proline-derived mitochondrial intermediates for use by the retina. We observed that in primary human RPE cell culture, proline is the only amino acid whose uptake increases with cellular maturity. In human RPE, proline was sufficient to stimulate de novo serine synthesis, increase reductive carboxylation, and protect against oxidative damage. Blocking proline catabolism in RPE impaired glucose metabolism and GSH production. Notably, in an acute model of RPE-induced retinal degeneration, dietary proline improved visual function. In conclusion, proline is an important nutrient that supports RPE metabolism and the metabolic demand of the retina.
Collapse
Affiliation(s)
- Michelle Yam
- From the Departments of Ophthalmology and
- Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Abbi L Engel
- the Department of Ophthalmology, University of Washington, Seattle, Washington 98109
| | - Yekai Wang
- From the Departments of Ophthalmology and
- Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Siyan Zhu
- From the Departments of Ophthalmology and
- Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Allison Hauer
- From the Departments of Ophthalmology and
- Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Rui Zhang
- From the Departments of Ophthalmology and
- the Save Sight Institute, University of Sydney, 8 Macquarie Street, Sydney, New South Wales 2000, Australia
| | - Daniel Lohner
- From the Departments of Ophthalmology and
- Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Jiancheng Huang
- From the Departments of Ophthalmology and
- the Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China, and
- the Department of Ophthalmology, State Key Laboratory of Reproductive Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Marlee Dinterman
- From the Departments of Ophthalmology and
- Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Chen Zhao
- the Eye Institute, Eye and ENT Hospital, Shanghai Medical College, Fudan University, Shanghai 200031, China, and
| | - Jennifer R Chao
- the Department of Ophthalmology, University of Washington, Seattle, Washington 98109,
| | - Jianhai Du
- From the Departments of Ophthalmology and
- Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| |
Collapse
|
31
|
|
32
|
Zhang T, Zhu L, Madigan MC, Liu W, Shen W, Cherepanoff S, Zhou F, Zeng S, Du J, Gillies MC. Human macular Müller cells rely more on serine biosynthesis to combat oxidative stress than those from the periphery. eLife 2019; 8:43598. [PMID: 31036157 PMCID: PMC6533082 DOI: 10.7554/elife.43598] [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: 11/15/2018] [Accepted: 04/08/2019] [Indexed: 11/18/2022] Open
Abstract
The human macula is more susceptible than the peripheral retina to developing blinding conditions such as age-related macular degeneration, diabetic retinopathy. A key difference between them may be the nature of their Müller cells. We found primary cultured Müller cells from macula and peripheral retina display significant morphological and transcriptomic differences. Macular Müller cells expressed more phosphoglycerate dehydrogenase (PHGDH, a rate-limiting enzyme in serine synthesis) than peripheral Müller cells. The serine synthesis, glycolytic and mitochondrial function were more activated in macular than peripheral Müller cells. Serine biosynthesis is critical in defending against oxidative stress. Intracellular reactive oxygen species and glutathione levels were increased in primary cultured macular Müller cells which were more susceptible to oxidative stress after inhibition of PHGDH. Our findings indicate serine biosynthesis is a critical part of the macular defence against oxidative stress and suggest dysregulation of this pathway as a potential cause of macular pathology.
Collapse
Affiliation(s)
- Ting Zhang
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Ling Zhu
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Michele C Madigan
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia.,School of Optometry and Vision Sciences, University of New South Wales, Sydney, Australia
| | - Wei Liu
- Clinical Genomics Laboratory, Sidra Medicine, Doha, Qatar
| | - Weiyong Shen
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Svetlana Cherepanoff
- Department of Anatomical Pathology, St Vincent's Hospital, Darlinghurst, Australia
| | - Fanfan Zhou
- Faculty of Pharmacy, The University of Sydney, Sydney, Australia
| | - Shaoxue Zeng
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Jianhai Du
- Department of Ophthalmology, West Virginia University, Morgantown, United States.,Department of Biochemistry, West Virginia University, Morgantown, United States
| | - Mark C Gillies
- Save Sight Institute, Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| |
Collapse
|
33
|
Loss of MPC1 reprograms retinal metabolism to impair visual function. Proc Natl Acad Sci U S A 2019; 116:3530-3535. [PMID: 30808746 DOI: 10.1073/pnas.1812941116] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Glucose metabolism in vertebrate retinas is dominated by aerobic glycolysis (the "Warburg Effect"), which allows only a small fraction of glucose-derived pyruvate to enter mitochondria. Here, we report evidence that the small fraction of pyruvate in photoreceptors that does get oxidized by their mitochondria is required for visual function, photoreceptor structure and viability, normal neuron-glial interaction, and homeostasis of retinal metabolism. The mitochondrial pyruvate carrier (MPC) links glycolysis and mitochondrial metabolism. Retina-specific deletion of MPC1 results in progressive retinal degeneration and decline of visual function in both rod and cone photoreceptors. Using targeted-metabolomics and 13C tracers, we found that MPC1 is required for cytosolic reducing power maintenance, glutamine/glutamate metabolism, and flexibility in fuel utilization.
Collapse
|
34
|
Huang J, Gu S, Chen M, Zhang SJ, Jiang Z, Chen X, Jiang C, Liu G, Radu RA, Sun X, Vollrath D, Du J, Yan B, Zhao C. Abnormal mTORC1 signaling leads to retinal pigment epithelium degeneration. Am J Cancer Res 2019; 9:1170-1180. [PMID: 30867823 PMCID: PMC6401408 DOI: 10.7150/thno.26281] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022] Open
Abstract
Retinal pigment epithelial (RPE) degeneration is potentially involved in the pathogenesis of several retinal degenerative diseases. mTORC1 signaling is shown as a crucial regulator of many biological processes and disease progression. In this study, we aimed at investigating the role of mTORC1 signaling in RPE degeneration. Methods: Western blots were conducted to detect mTORC1 expression pattern during RPE degeneration. Cre-loxP system was used to generate RPE-specific mTORC1 activation mice. Fundus, immunofluorescence staining, transmission electron microscopy, and targeted metabolomic analysis were conducted to determine the effects of mTORC1 activation on RPE degeneration in vivo. Electroretinography, spectral-domain optical coherence tomography, and histological experiments were conducted to determine the effects of mTORC1 activation on choroidal and retinal function in vivo. Results: RPE-specific activation of mTORC1 led to RPE degeneration as shown by the loss of RPE-specific marker, compromised cell junction integrity, and intracellular accumulation of lipid droplets. RPE degeneration further led to abnormal choroidal and retinal function. The inhibition of mTORC1 signaling with rapamycin could partially reverse RPE degeneration. Targeted metabolomics analysis further revealed that mTORC1 activation affected the metabolism of purine, carboxylic acid, and niacin in RPE. Conclusion: This study revealed that abnormal activation of mTORC1 signaling leads to RPE degeneration, which could provide a promising target for the treatment of RPE dysfunction-related diseases.
Collapse
|
35
|
Millman JR, Doggett T, Thebeau C, Zhang S, Semenkovich CF, Rajagopal R. Measurement of Energy Metabolism in Explanted Retinal Tissue Using Extracellular Flux Analysis. J Vis Exp 2019. [PMID: 30663677 DOI: 10.3791/58626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
High acuity vision is a heavily energy-consuming process, and the retina has developed several unique adaptations to precisely meet such demands while maintaining transparency of the visual axis. Perturbations to this delicate balance cause blinding illnesses, such as diabetic retinopathy. Therefore, the understanding of energy metabolism changes in the retina during disease is imperative to the development of rational therapies for various causes of vison loss. The recent advent of commercially-available extracellular flux analyzers has made the study of retinal energy metabolism more accessible. This protocol describes the use of such an analyzer to measure contributions to retinal energy supply through its two principle arms - oxidative phosphorylation and glycolysis - by quantifying changes in oxygen consumption rates (OCR) and extracellular acidification rates (ECAR) as proxies for these pathways. This technique is readily performed in explanted retinal tissue, facilitating assessment of responses to multiple pharmacologic agents in a single experiment. Metabolic signatures in retinas from animals lacking rod photoreceptor signaling are compared to wild-type controls using this method. A major limitation in this technique is the lack of ability to discriminate between light-adapted and dark-adapted energy utilization, an important physiologic consideration in retinal tissue.
Collapse
Affiliation(s)
- Jeffrey R Millman
- Division of Metabolism, Endocrinology and Lipid Research, Department of Medicine, Washington University School of Medicine; Department of Biomedical Engineering, Washington University in Saint Louis
| | - Teresa Doggett
- Department of Ophthalmology and Visual Science, Washington University School of Medicine
| | - Christina Thebeau
- Department of Ophthalmology and Visual Science, Washington University School of Medicine
| | - Sheng Zhang
- Department of Ophthalmology and Visual Science, Washington University School of Medicine
| | - Clay F Semenkovich
- Division of Metabolism, Endocrinology and Lipid Research, Department of Medicine, Washington University School of Medicine
| | - Rithwick Rajagopal
- Department of Ophthalmology and Visual Science, Washington University School of Medicine;
| |
Collapse
|
36
|
Brown CN, Green BD, Thompson RB, den Hollander AI, Lengyel I. Metabolomics and Age-Related Macular Degeneration. Metabolites 2018; 9:metabo9010004. [PMID: 30591665 PMCID: PMC6358913 DOI: 10.3390/metabo9010004] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/17/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Age-related macular degeneration (AMD) leads to irreversible visual loss, therefore, early intervention is desirable, but due to its multifactorial nature, diagnosis of early disease might be challenging. Identification of early markers for disease development and progression is key for disease diagnosis. Suitable biomarkers can potentially provide opportunities for clinical intervention at a stage of the disease when irreversible changes are yet to take place. One of the most metabolically active tissues in the human body is the retina, making the use of hypothesis-free techniques, like metabolomics, to measure molecular changes in AMD appealing. Indeed, there is increasing evidence that metabolic dysfunction has an important role in the development and progression of AMD. Therefore, metabolomics appears to be an appropriate platform to investigate disease-associated biomarkers. In this review, we explored what is known about metabolic changes in the retina, in conjunction with the emerging literature in AMD metabolomics research. Methods for metabolic biomarker identification in the eye have also been discussed, including the use of tears, vitreous, and aqueous humor, as well as imaging methods, like fluorescence lifetime imaging, that could be translated into a clinical diagnostic tool with molecular level resolution.
Collapse
Affiliation(s)
- Connor N Brown
- Wellcome-Wolfson Institute for Experimental Medicine (WWIEM), Queen's University Belfast, Belfast BT9 7BL, UK.
| | - Brian D Green
- Institute for Global Food Security (IGFS), Queen's University Belfast, Belfast BT9 6AG, UK.
| | - Richard B Thompson
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA.
| | - Anneke I den Hollander
- Department of Ophthalmology, Radboud University Nijmegen Medical Centre, Nijmegen 6525 EX, The Netherlands.
| | - Imre Lengyel
- Wellcome-Wolfson Institute for Experimental Medicine (WWIEM), Queen's University Belfast, Belfast BT9 7BL, UK.
| |
Collapse
|
37
|
Sinha T, Makia M, Du J, Naash MI, Al-Ubaidi MR. Flavin homeostasis in the mouse retina during aging and degeneration. J Nutr Biochem 2018; 62:123-133. [PMID: 30290331 PMCID: PMC7162609 DOI: 10.1016/j.jnutbio.2018.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/31/2018] [Accepted: 09/01/2018] [Indexed: 12/14/2022]
Abstract
Involvement of flavin adenine dinucleotide (FAD) and flavin mononucleotide (FMN) in cellular homeostasis has been well established for tissues other than the retina. Here, we present an optimized method to effectively extract and quantify FAD and FMN from a single neural retina and its corresponding retinal pigment epithelium (RPE). Optimizations led to detection efficiency of 0.1 pmol for FAD and FMN while 0.01 pmol for riboflavin. Interestingly, levels of FAD and FMN in the RPE were found to be 1.7- and 12.5-fold higher than their levels in the retina, respectively. Both FAD and FMN levels in the RPE and retina gradually decline with age and preceded the age-dependent drop in the functional competence of the retina as measured by electroretinography. Further, quantifications of retinal levels of FAD and FMN in different mouse models of retinal degeneration revealed differential metabolic requirements of these two factors in relation to the rate and degree of photoreceptor degeneration. We also found twofold reductions in retinal levels of FAD and FMN in two mouse models of diabetic retinopathy. Altogether, our results suggest that retinal levels of FAD and FMN can be used as potential markers to determine state of health of the retina in general and more specifically the photoreceptors.
Collapse
Affiliation(s)
- Tirthankar Sinha
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204
| | - Mustafa Makia
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204
| | - Jianhai Du
- Department of Ophthalmology and Department of Biochemistry, West Virginia University, Morgantown, WV 26506
| | - Muna I Naash
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204.
| | - Muayyad R Al-Ubaidi
- Department of Biomedical Engineering, University of Houston, Houston, TX 77204.
| |
Collapse
|
38
|
Swarup A, Bell BA, Du J, Han JYS, Soto J, Abel ED, Bravo-Nuevo A, FitzGerald PG, Peachey NS, Philp NJ. Deletion of GLUT1 in mouse lens epithelium leads to cataract formation. Exp Eye Res 2018; 172:45-53. [PMID: 29604281 PMCID: PMC6716165 DOI: 10.1016/j.exer.2018.03.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 03/21/2018] [Accepted: 03/22/2018] [Indexed: 10/17/2022]
Abstract
The primary energy substrate of the lens is glucose and uptake of glucose from the aqueous humor is dependent on glucose transporters. GLUT1, the facilitated glucose transporter encoded by Slc2a1 is expressed in the epithelium of bovine, human and rat lenses. In the current study, we examined the expression of GLUT1 in the mouse lens and determined its role in maintaining lens transparency by studying effects of postnatal deletion of Slc2a1. In situ hybridization and immunofluorescence labeling were used to determine the expression and subcellular distribution of GLUT1 in the lens. Slc2a1 was knocked out of the lens epithelium by crossing transgenic mice expressing Cre recombinase under control of the GFAP promoter with Slc2a1loxP/loxP mice to generate Slc2a1loxP/loxP;GFAP-Cre+/0 (LensΔGlut1) mice. LensΔGlut1 mice developed visible lens opacities by around 3 months of age, which corresponded temporally with the total loss of detectable GLUT1 expression in the lens. Spectral domain optical coherence tomography (SD-OCT) imaging was used to monitor the formation of cataracts over time. SD-OCT imaging revealed that small nuclear cataracts were first apparent in the lenses of LensΔGlut1 mice beginning at about 2.7 months of age. Longitudinal SD-OCT imaging of LensΔGlut1 mice revealed disruption of mature secondary fiber cells after 3 months of age. Histological sections of eyes from LensΔGlut1 mice confirmed the disruption of the secondary fiber cells. The structural changes were most pronounced in fiber cells that had lost their organelles. In contrast, the histology of the lens epithelium in these mice appeared normal. Lactate and ATP were measured in lenses from LensΔGlut1 and control mice at 2 and 3 months of age. At 2 months of age, when GLUT1 was still detectable in the lens epithelium, albeit at low levels, the amount of lactate and ATP were not significantly different from controls. However, in lenses isolated from 3-month-old LensΔGlut1 mice, when GLUT1 was no longer detectable, levels of lactate and ATP were 50% lower than controls. Our findings demonstrate that in vivo, the transparency of mature lens fiber cells was dependent on glycolysis for ATP and the loss of GLUT1 transporters led to cataract formation. In contrast, lens epithelium and cortical fiber cells have mitochondria and could utilize other substrates to support their anabolic and catabolic needs.
Collapse
Affiliation(s)
- Aditi Swarup
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Brent A Bell
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jianhai Du
- West Virginia University Eye Institute, Morgantown, WV, USA
| | - John Y S Han
- Department of Pathology, Anatomy & Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jamie Soto
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Division of Endocrinology & Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, USA; Division of Endocrinology & Metabolism, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Arturo Bravo-Nuevo
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Paul G FitzGerald
- Department of Cell Biology & Human Anatomy, University of California at Davis, Davis, CA, 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
|
39
|
Zhu S, Yam M, Wang Y, Linton JD, Grenell A, Hurley JB, Du J. Impact of euthanasia, dissection and postmortem delay on metabolic profile in mouse retina and RPE/choroid. Exp Eye Res 2018; 174:113-120. [PMID: 29864440 DOI: 10.1016/j.exer.2018.05.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 05/30/2018] [Accepted: 05/31/2018] [Indexed: 01/02/2023]
Abstract
Metabolomics studies in the retina and retinal pigment epithelium (RPE) in animal models or postmortem donors are essential to understanding the retinal metabolism and to revealing the underlying mechanisms of retinal degenerative diseases. We have studied how different methods of euthanasia (CO2 or cervical dislocation) different isolation procedures and postmortem delay affect metabolites in mouse retina and RPE/choroid using LC MS/MS and GC MS. Compared with cervical dislocation, CO2 exposure for 5 min dramatically degrades ATP and GTP into purine metabolites in the retina while raising intermediates in glucose metabolism and amino acids in the RPE/choroid. Isolation in cold buffer containing glucose has the least change in metabolites. Postmortem delay time-dependently and differentially impacts metabolites in the retina and RPE/choroid. In the postmortem retina, 18% of metabolites were changed at 0.5 h (h), 41% at 4 h and 51% at 8 h. However, only 6% of metabolites were changed in the postmortem RPE/choroid and it steadily increased to 20% at 8 h. Notably, both postmortem retina and RPE/choroid tissue showed increased purine metabolites. Storage of eyes in cold nutrient-rich medium substantially blocked the postmortem change in the retina and RPE/choroid. In conclusion, our study provides optimized methods to prepare fresh or postmortem retina and RPE/choroid tissue for metabolomics studies.
Collapse
Affiliation(s)
- Siyan Zhu
- Department of Ophthalmology, West Virginia University, Morgantown, WV 26506, USA; Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA
| | - Michelle Yam
- Department of Ophthalmology, West Virginia University, Morgantown, WV 26506, USA; Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA
| | - Yekai Wang
- Department of Ophthalmology, West Virginia University, Morgantown, WV 26506, USA; Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA
| | - Jonathan D Linton
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA
| | - Allison Grenell
- Department of Ophthalmology, West Virginia University, Morgantown, WV 26506, USA; Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, WA 98109, USA; Department of Ophthalmology, University of Washington, Seattle, WA 98109, USA
| | - Jianhai Du
- Department of Ophthalmology, West Virginia University, Morgantown, WV 26506, USA; Department of Biochemistry, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
40
|
Park KS, Xu CL, Cui X, Tsang SH. Reprogramming the metabolome rescues retinal degeneration. Cell Mol Life Sci 2018; 75:1559-1566. [PMID: 29332245 PMCID: PMC9377522 DOI: 10.1007/s00018-018-2744-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 12/27/2017] [Accepted: 01/02/2018] [Indexed: 02/03/2023]
Abstract
Metabolomics studies in the context of ophthalmology have largely focused on identifying metabolite concentrations that characterize specific retinal diseases. Studies involving mass spectrometry (MS) and nuclear magnetic resonance (NMR) spectroscopy have shown that individuals suffering from retinal diseases exhibit metabolic profiles that markedly differ from those of control individuals, supporting the notion that metabolites may serve as easily identifiable biomarkers for specific conditions. An emerging branch of metabolomics resulting from biomarker studies, however, involves the study of retinal metabolic dysfunction as causes of degeneration. Recent publications have identified a number of metabolic processes-including but not limited to glucose and oxygen metabolism-that, when perturbed, play a role in the degeneration of photoreceptor cells. As a result, such studies have led to further research elucidating methods for prolonging photoreceptor survival in an effort to halt degeneration in its early stages. This review will explore the ways in which metabolomics has deepened our understanding of the causes of retinal degeneration and discuss how metabolomics can be used to prevent retinal degeneration from progressing to its later disease stages.
Collapse
Affiliation(s)
- Karen Sophia Park
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Christine L Xu
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Xuan Cui
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, NY, USA.
- Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, NY, USA.
- Departments of Ophthalmology, Pathology, and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
41
|
Pyruvate kinase M2 regulates photoreceptor structure, function, and viability. Cell Death Dis 2018; 9:240. [PMID: 29445082 PMCID: PMC5833680 DOI: 10.1038/s41419-018-0296-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/18/2017] [Accepted: 12/27/2017] [Indexed: 01/30/2023]
Abstract
Pyruvate kinase M2 (PKM2) is a glycolytic enzyme that is expressed in cancer cells. Its role in tumor metabolism is not definitively established, but investigators have suggested that regulation of PKM2 activity can cause accumulation of glycolytic intermediates and increase flux through the pentose phosphate pathway. Recent evidence suggests that PKM2 also may have non-metabolic functions, including as a transcriptional co-activator in gene regulation. We reported previously that PKM2 is abundant in photoreceptor cells in mouse retinas. In the present study, we conditionally deleted PKM2 (rod-cre PKM2-KO) in rod photoreceptors and found that the absence of PKM2 causes increased expression of PKM1 in rods. Analysis of metabolic flux from U-13C glucose shows that rod-cre PKM2-KO retinas accumulate glycolytic intermediates, consistent with an overall reduction in the amount of pyruvate kinase activity. Rod-cre PKM2-KO mice also have an increased NADPH availability could favor lipid synthesis, but we found no difference in phospholipid synthesis between rod-cre PKM2 KO and PKM2-positive controls. As rod-cre PKM2-KO mice aged, we observed a significant loss of rod function, reduced thickness of the photoreceptor outer segment layer, and reduced expression of photoreceptor proteins, including PDE6β. The rod-cre PKM2-KO retinas showed greater TUNEL staining than wild-type retinas, indicating a slow retinal degeneration. In vitro analysis showed that PKM2 can regulate transcriptional activity from the PDE6β promoter in vitro. Our findings indicate that both the metabolic and transcriptional regulatory functions of PKM2 may contribute to photoreceptor structure, function, and viability.
Collapse
|
42
|
Wubben TJ, Pawar M, Smith A, Toolan K, Hager H, Besirli CG. Photoreceptor metabolic reprogramming provides survival advantage in acute stress while causing chronic degeneration. Sci Rep 2017; 7:17863. [PMID: 29259242 PMCID: PMC5736549 DOI: 10.1038/s41598-017-18098-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 12/05/2017] [Indexed: 11/16/2022] Open
Abstract
Photoreceptor death is the root cause of vision loss in many retinal disorders, and there is an unmet need for neuroprotective modalities to improve photoreceptor survival. The biosynthetic requirement of photoreceptors is among the highest in the body, and to meet this demand, photoreceptors maintain their ability to perform aerobic glycolysis. This highly regulated form of glycolysis allows cells to efficiently budget their metabolic needs and may be a critical link between photoreceptor function and survival. Pyruvate kinase muscle isozyme 2 (PKM2) is a key regulator of aerobic glycolysis. In the present study, we characterized the effect of PKM2 deletion on baseline functioning and survival of photoreceptors over time by utilizing a photoreceptor-specific, PKM2 knockout mouse model. We found that upon PKM2 deletion, PKM1 is upregulated in the outer retina and there is increased expression of genes involved in glucose metabolism, which led to chronic degenerative changes in the outer retina of these mice. We also discovered that this metabolic reprogramming provided a survival advantage to photoreceptors in an experimental model of retinal detachment. This study strongly supports the hypothesis that reprogramming metabolism may be a novel therapeutic strategy for photoreceptor neuroprotection during acute stress.
Collapse
Affiliation(s)
- Thomas J Wubben
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, Michigan, USA
| | - Mercy Pawar
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, Michigan, USA
| | - Andrew Smith
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, Michigan, USA
| | - Kevin Toolan
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, Michigan, USA
| | - Heather Hager
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, Michigan, USA
| | - Cagri G Besirli
- University of Michigan, Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, Ann Arbor, Michigan, USA.
| |
Collapse
|
43
|
Kanow MA, Giarmarco MM, Jankowski CS, Tsantilas K, Engel AL, Du J, Linton JD, Farnsworth CC, Sloat SR, Rountree A, Sweet IR, Lindsay KJ, Parker ED, Brockerhoff SE, Sadilek M, Chao JR, Hurley JB. Biochemical adaptations of the retina and retinal pigment epithelium support a metabolic ecosystem in the vertebrate eye. eLife 2017; 6:28899. [PMID: 28901286 PMCID: PMC5617631 DOI: 10.7554/elife.28899] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/12/2017] [Indexed: 12/12/2022] Open
Abstract
Here we report multiple lines of evidence for a comprehensive model of energy metabolism in the vertebrate eye. Metabolic flux, locations of key enzymes, and our finding that glucose enters mouse and zebrafish retinas mostly through photoreceptors support a conceptually new model for retinal metabolism. In this model, glucose from the choroidal blood passes through the retinal pigment epithelium to the retina where photoreceptors convert it to lactate. Photoreceptors then export the lactate as fuel for the retinal pigment epithelium and for neighboring Müller glial cells. We used human retinal epithelial cells to show that lactate can suppress consumption of glucose by the retinal pigment epithelium. Suppression of glucose consumption in the retinal pigment epithelium can increase the amount of glucose that reaches the retina. This framework for understanding metabolic relationships in the vertebrate retina provides new insights into the underlying causes of retinal disease and age-related vision loss.
Collapse
Affiliation(s)
- Mark A Kanow
- Department of Biochemistry, University of Washington, Seattle, United States
| | | | - Connor Sr Jankowski
- Department of Biochemistry, University of Washington, Seattle, United States
| | - Kristine Tsantilas
- Department of Biochemistry, University of Washington, Seattle, United States
| | - Abbi L Engel
- Department of Ophthalmology, University of Washington, Seattle, United States
| | - Jianhai Du
- Department of Ophthalmology, West Virginia University, Morgantown, United States.,Department of Biochemistry, West Virginia University, Morgantown, United States
| | - Jonathan D Linton
- Department of Biochemistry, University of Washington, Seattle, United States.,Department of Ophthalmology, University of Washington, Seattle, United States
| | | | - Stephanie R Sloat
- Department of Biochemistry, University of Washington, Seattle, United States
| | - Austin Rountree
- Department of Medicine, UW Diabetes Institute, University of Washington, Seattle, United States
| | - Ian R Sweet
- Department of Medicine, UW Diabetes Institute, University of Washington, Seattle, United States
| | - Ken J Lindsay
- Department of Biochemistry, University of Washington, Seattle, United States.,Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Edward D Parker
- Department of Ophthalmology, University of Washington, Seattle, United States
| | - Susan E Brockerhoff
- Department of Biochemistry, University of Washington, Seattle, United States.,Department of Ophthalmology, University of Washington, Seattle, United States
| | - Martin Sadilek
- Department of Chemistry, University of Washington, Seattle, United States
| | - Jennifer R Chao
- Department of Ophthalmology, University of Washington, Seattle, United States
| | - James B Hurley
- Department of Biochemistry, University of Washington, Seattle, United States.,Department of Ophthalmology, University of Washington, Seattle, United States
| |
Collapse
|
44
|
Chao JR, Knight K, Engel AL, Jankowski C, Wang Y, Manson MA, Gu H, Djukovic D, Raftery D, Hurley JB, Du J. Human retinal pigment epithelial cells prefer proline as a nutrient and transport metabolic intermediates to the retinal side. J Biol Chem 2017; 292:12895-12905. [PMID: 28615447 DOI: 10.1074/jbc.m117.788422] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/30/2017] [Indexed: 11/06/2022] Open
Abstract
Metabolite transport is a major function of the retinal pigment epithelium (RPE) to support the neural retina. RPE dysfunction plays a significant role in retinal degenerative diseases. We have used mass spectrometry with 13C tracers to systematically study nutrient consumption and metabolite transport in cultured human fetal RPE. LC/MS-MS detected 120 metabolites in the medium from either the apical or basal side. Surprisingly, more proline is consumed than any other nutrient, including glucose, taurine, lipids, vitamins, or other amino acids. Besides being oxidized through the Krebs cycle, proline is used to make citrate via reductive carboxylation. Citrate, made either from 13C proline or from 13C glucose, is preferentially exported to the apical side and is taken up by the retina. In conclusion, RPE cells consume multiple nutrients, including glucose and taurine, but prefer proline, and they actively synthesize and export metabolic intermediates to the apical side to nourish the outer retina.
Collapse
Affiliation(s)
- Jennifer R Chao
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109.
| | - Kaitlen Knight
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109
| | - Abbi L Engel
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109
| | - Connor Jankowski
- Department of Biochemistry, University of Washington, Seattle, Washington 98109
| | - Yekai Wang
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506; Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506
| | - Megan A Manson
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109
| | - Haiwei Gu
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
| | - Danijel Djukovic
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
| | - Daniel Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington 98109
| | - James B Hurley
- Department of Ophthalmology, University of Washington, Seattle, Washington 98109; Department of Biochemistry, University of Washington, Seattle, Washington 98109
| | - Jianhai Du
- Department of Ophthalmology, West Virginia University, Morgantown, West Virginia 26506; Department of Biochemistry, West Virginia University, Morgantown, West Virginia 26506.
| |
Collapse
|
45
|
de la Barca JMC, Huang NT, Jiao H, Tessier L, Gadras C, Simard G, Natoli R, Tcherkez G, Reynier P, Valter K. Retinal metabolic events in preconditioning light stress as revealed by wide-spectrum targeted metabolomics. Metabolomics 2017; 13:22. [PMID: 28706468 PMCID: PMC5486622 DOI: 10.1007/s11306-016-1156-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 12/20/2016] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Light is the primary stimulus for vision, but may also cause damage to the retina. Pre-exposing the retina to sub-lethal amount of light (or preconditioning) improves chances for retinal cells to survive acute damaging light stress. OBJECTIVES This study aims at exploring the changes in retinal metabolome after mild light stress and identifying mechanisms that may be involved in preconditioning. METHODS Retinas from 12 rats exposed to mild light stress (1000 lux × for 12 h) and 12 controls were collected one and seven days after light stress (LS). One retina was used for targeted metabolomics analysis using the Biocrates p180 kit while the fellow retina was used for histological and immunohistochemistry analysis. RESULTS Immunohistochemistry confirmed that in this experiment, a mild LS with retinal immune response and minimal photoreceptor loss occurred. Compared to controls, LS induced an increased concentration in phosphatidylcholines. The concentration in some amino acids and biogenic amines, particularly those related to the nitric oxide pathway (like asymmetric dimethylarginine (ADMA), arginine and citrulline) also increased 1 day after LS. 7 days after LS, the concentration in two sphingomyelins and phenylethylamine was found to be higher. We further found that in controls, retina metabolome was different between males and females: male retinas had an increased concentration in tyrosine, acetyl-ornithine, phosphatidylcholines and (acyl)-carnitines. CONCLUSIONS Besides retinal sexual metabolic dimorphism, this study shows that preconditioning is mostly associated with re-organisation of lipid metabolism and changes in amino acid composition, likely reflecting the involvement of arginine-dependent NO signalling.
Collapse
Affiliation(s)
- Juan Manuel Chao de la Barca
- 0000 0001 2248 3363grid.7252.2PREMMi/Pôle de Recherche et d’Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d’Angers, 49933 Angers, France
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
| | - Nuan-Ting Huang
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
| | - Haihan Jiao
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
| | - Lydie Tessier
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
| | - Cédric Gadras
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
| | - Gilles Simard
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
- 0000 0001 2248 3363grid.7252.2INSERM U1063, Université d’Angers, 49933 Angers, France
| | - Riccardo Natoli
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
- 0000 0001 2180 7477grid.1001.0Medical School, Australian National University, Canberra, ACT 2601 Australia
| | - Guillaume Tcherkez
- 0000 0001 2180 7477grid.1001.0Research School of Biology, College of Medicine, Biology and Environment, Australian National University, Canberra, ACT 2601 Australia
| | - Pascal Reynier
- 0000 0001 2248 3363grid.7252.2PREMMi/Pôle de Recherche et d’Enseignement en Médecine Mitochondriale, Institut MITOVASC, CNRS 6214, INSERM U1083, Université d’Angers, 49933 Angers, France
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, 4 rue Larrey, 49933 Angers cedex 9, France
| | - Krisztina Valter
- 0000 0001 2180 7477grid.1001.0Eccles Institute of Neuroscience, John Curtin School of Medical research, Australian National University, Canberra, ACT 2601 Australia
- 0000 0001 2180 7477grid.1001.0Medical School, Australian National University, Canberra, ACT 2601 Australia
| |
Collapse
|
46
|
Reductive carboxylation is a major metabolic pathway in the retinal pigment epithelium. Proc Natl Acad Sci U S A 2016; 113:14710-14715. [PMID: 27911769 DOI: 10.1073/pnas.1604572113] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The retinal pigment epithelium (RPE) is a monolayer of pigmented cells that requires an active metabolism to maintain outer retinal homeostasis and compensate for oxidative stress. Using 13C metabolic flux analysis in human RPE cells, we found that RPE has an exceptionally high capacity for reductive carboxylation, a metabolic pathway that has recently garnered significant interest because of its role in cancer cell survival. The capacity for reductive carboxylation in RPE exceeds that of all other cells tested, including retina, neural tissue, glial cells, and a cancer cell line. Loss of reductive carboxylation disrupts redox balance and increases RPE sensitivity to oxidative damage, suggesting that deficiencies of reductive carboxylation may contribute to RPE cell death. Supporting reductive carboxylation by supplementation with an NAD+ precursor or its substrate α-ketoglutarate or treatment with a poly(ADP ribose) polymerase inhibitor protects reductive carboxylation and RPE viability from excessive oxidative stress. The ability of these treatments to rescue RPE could be the basis for an effective strategy to treat blinding diseases caused by RPE dysfunction.
Collapse
|
47
|
Zhang L, Du J, Justus S, Hsu CW, Bonet-Ponce L, Wu WH, Tsai YT, Wu WP, Jia Y, Duong JK, Mahajan VB, Lin CS, Wang S, Hurley JB, Tsang SH. Reprogramming metabolism by targeting sirtuin 6 attenuates retinal degeneration. J Clin Invest 2016; 126:4659-4673. [PMID: 27841758 DOI: 10.1172/jci86905] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 10/06/2016] [Indexed: 12/16/2022] Open
Abstract
Retinitis pigmentosa (RP) encompasses a diverse group of Mendelian disorders leading to progressive degeneration of rods and then cones. For reasons that remain unclear, diseased RP photoreceptors begin to deteriorate, eventually leading to cell death and, consequently, loss of vision. Here, we have hypothesized that RP associated with mutations in phosphodiesterase-6 (PDE6) provokes a metabolic aberration in rod cells that promotes the pathological consequences of elevated cGMP and Ca2+, which are induced by the Pde6 mutation. Inhibition of sirtuin 6 (SIRT6), a histone deacetylase repressor of glycolytic flux, reprogrammed rods into perpetual glycolysis, thereby driving the accumulation of biosynthetic intermediates, improving outer segment (OS) length, enhancing photoreceptor survival, and preserving vision. In mouse retinae lacking Sirt6, effectors of glycolytic flux were dramatically increased, leading to upregulation of key intermediates in glycolysis, TCA cycle, and glutaminolysis. Both transgenic and AAV2/8 gene therapy-mediated ablation of Sirt6 in rods provided electrophysiological and anatomic rescue of both rod and cone photoreceptors in a preclinical model of RP. Due to the extensive network of downstream effectors of Sirt6, this study motivates further research into the role that these pathways play in retinal degeneration. Because reprogramming metabolism by enhancing glycolysis is not gene specific, this strategy may be applicable to a wide range of neurodegenerative disorders.
Collapse
|
48
|
Du J, Rountree A, Cleghorn WM, Contreras L, Lindsay KJ, Sadilek M, Gu H, Djukovic D, Raftery D, Satrústegui J, Kanow M, Chan L, Tsang SH, Sweet IR, Hurley JB. Phototransduction Influences Metabolic Flux and Nucleotide Metabolism in Mouse Retina. J Biol Chem 2015; 291:4698-710. [PMID: 26677218 DOI: 10.1074/jbc.m115.698985] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Indexed: 01/20/2023] Open
Abstract
Production of energy in a cell must keep pace with demand. Photoreceptors use ATP to maintain ion gradients in darkness, whereas in light they use it to support phototransduction. Matching production with consumption can be accomplished by coupling production directly to consumption. Alternatively, production can be set by a signal that anticipates demand. In this report we investigate the hypothesis that signaling through phototransduction controls production of energy in mouse retinas. We found that respiration in mouse retinas is not coupled tightly to ATP consumption. By analyzing metabolic flux in mouse retinas, we also found that phototransduction slows metabolic flux through glycolysis and through intermediates of the citric acid cycle. We also evaluated the relative contributions of regulation of the activities of α-ketoglutarate dehydrogenase and the aspartate-glutamate carrier 1. In addition, a comprehensive analysis of the retinal metabolome showed that phototransduction also influences steady-state concentrations of 5'-GMP, ribose-5-phosphate, ketone bodies, and purines.
Collapse
Affiliation(s)
- Jianhai Du
- From the Department of Biochemistry, Department of Ophthalmology, University of Washington, Seattle, Washington 98109
| | | | | | - Laura Contreras
- Department of Molecular Biology, Centre for Molecular Biology Severo Ochoa, Universidad Autonoma de Madrid-Consejo Superior de Investigaciones Científicas, CIBER of Rare Diseases (CIBERER), and Health Research Institute Jimenez Diaz Foundation, Autonomous University of Madrid, 28049 Madrid, Spain
| | | | | | - Haiwei Gu
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine
| | - Danijel Djukovic
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine
| | - Dan Raftery
- Northwest Metabolomics Research Center, Department of Anesthesiology and Pain Medicine
| | - Jorgina Satrústegui
- Department of Molecular Biology, Centre for Molecular Biology Severo Ochoa, Universidad Autonoma de Madrid-Consejo Superior de Investigaciones Científicas, CIBER of Rare Diseases (CIBERER), and Health Research Institute Jimenez Diaz Foundation, Autonomous University of Madrid, 28049 Madrid, Spain
| | | | - Lawrence Chan
- Bernard and Shirlee Brown Glaucoma Laboratory and Barbara and Donald Jonas Stem Cell Laboratory, Department of Ophthalmology, Columbia University, New York, New York, and
| | - Stephen H Tsang
- Bernard and Shirlee Brown Glaucoma Laboratory and Barbara and Donald Jonas Stem Cell Laboratory, Department of Ophthalmology, Columbia University, New York, New York, and Department of Pathology and Cell Biology and Institute of Human Nutrition, Columbia University, New York, New York 10032
| | | | - James B Hurley
- From the Department of Biochemistry, Department of Ophthalmology, University of Washington, Seattle, Washington 98109,
| |
Collapse
|