Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
J Biol Chem ; 299(11): 105275, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37741457

RESUMO

It is known that metabolic defects in the retinal pigment epithelium (RPE) can cause degeneration of its neighboring photoreceptors in the retina, leading to retinal degenerative diseases such as age-related macular degeneration. However, how RPE metabolism supports the health of the neural retina remains unclear. The retina requires exogenous nitrogen sources for protein synthesis, neurotransmission, and energy metabolism. Using 15N tracing coupled with mass spectrometry, we found human RPE can utilize the nitrogen in proline to produce and export 13 amino acids, including glutamate, aspartate, glutamine, alanine, and serine. Similarly, we found this proline nitrogen utilization in the mouse RPE/choroid but not in the neural retina of explant cultures. Coculture of human RPE with the retina showed that the retina can take up the amino acids, especially glutamate, aspartate, and glutamine, generated from proline nitrogen in the RPE. Intravenous delivery of 15N proline in vivo demonstrated 15N-derived amino acids appear earlier in the RPE before the retina. We also found proline dehydrogenase, the key enzyme in proline catabolism is highly enriched in the RPE but not the retina. The deletion of proline dehydrogenase blocks proline nitrogen utilization in RPE and the import of proline nitrogen-derived amino acids in the retina. Our findings highlight the importance of RPE metabolism in supporting nitrogen sources for the retina, providing insight into understanding the mechanisms of the retinal metabolic ecosystem and RPE-initiated retinal degenerative diseases.


Assuntos
Aminoácidos , Epitélio Pigmentado da Retina , Animais , Humanos , Camundongos , Aminoácidos/metabolismo , Ácido Aspártico/metabolismo , Glutamatos/metabolismo , Glutamina/metabolismo , Nitrogênio/metabolismo , Prolina/metabolismo , Prolina Oxidase/metabolismo , Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo
2.
Exp Eye Res ; 245: 109966, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38857822

RESUMO

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 (ß-HB). Prior work, based on detecting ß-HB via enzymatic assays, suggested that differentiated cultures of primary RPE preferentially export ß-HB across the apical membrane. Here, we compare the accuracy of measuring ß-HB by enzymatic assay kits to mass spectrometry analysis. We found that commercial kits lack the sensitivity to accurately measure the levels of ß-HB in RPE cultures and are prone to artifact. Using mass spectrometry, we found that while RPE cultures secrete ß-HB, they do so equally to both apical and basal sides. We also find RPE is capable of consuming ß-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 ß-HB. Altogether, we substantiate ß-HB secretion in RPE but find that the secretion is equal apically and basally, RPE ß-HB can derive from ketogenic amino acids or fatty acids, and accurate ß-HB assessment requires mass spectrometric analysis.

3.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-33526685

RESUMO

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.


Assuntos
Proteínas do Olho/genética , Piruvato Quinase/genética , Retina/metabolismo , Degeneração Retiniana/genética , Animais , Ciclo do Ácido Cítrico/genética , Modelos Animais de Doenças , Proteínas do Olho/metabolismo , Flavinas/metabolismo , Glicólise/genética , Homeostase , Humanos , Camundongos , Piruvato Quinase/metabolismo , Retina/patologia , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia
4.
Adv Exp Med Biol ; 1415: 435-441, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37440069

RESUMO

Metabolism is adapted to meet energetic needs. Based on the amount of ATP required to maintain plasma membrane potential, photoreceptor energy demands must be high. The available evidence suggests that photoreceptors primarily generate metabolic energy through aerobic glycolysis, though this evidence is based primarily on protein expression and not measurement of metabolic flux. Aerobic glycolysis can be validated by measuring flux of glucose to lactate. Aerobic glycolysis is also inefficient and thus an unexpected adaptation for photoreceptors to make. We measured metabolic rates to determine the energy-generating pathways that support photoreceptor metabolism. We found that photoreceptors indeed perform aerobic glycolysis and this is associated with mitochondrial uncoupling.


Assuntos
Glicólise , Células Fotorreceptoras , Células Fotorreceptoras/metabolismo , Mitocôndrias/metabolismo , Ácido Láctico/metabolismo , Metabolismo Energético , Glucose/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(46): 28816-28827, 2020 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-33144507

RESUMO

Cone photoreceptors in the retina are exposed to intense daylight and have higher energy demands in darkness. Cones produce energy using a large cluster of mitochondria. Mitochondria are susceptible to oxidative damage, and healthy mitochondrial populations are maintained by regular turnover. Daily cycles of light exposure and energy consumption suggest that mitochondrial turnover is important for cone health. We investigated the three-dimensional (3D) ultrastructure and metabolic function of zebrafish cone mitochondria throughout the day. At night retinas undergo a mitochondrial biogenesis event, corresponding to an increase in the number of smaller, simpler mitochondria and increased metabolic activity in cones. In the daytime, endoplasmic reticula (ER) and autophagosomes associate more with mitochondria, and mitochondrial size distribution across the cluster changes. We also report dense material shared between cone mitochondria that is extruded from the cell at night, sometimes forming extracellular structures. Our findings reveal an elaborate set of daily changes to cone mitochondrial structure and function.


Assuntos
Mitocôndrias/metabolismo , Dinâmica Mitocondrial/fisiologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Animais , Ritmo Circadiano/fisiologia , Adaptação à Escuridão/fisiologia , Retículo Endoplasmático/metabolismo , Retina/metabolismo , Sinapses/metabolismo , Peixe-Zebra
6.
Exp Eye Res ; 215: 108899, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34929159

RESUMO

Sorsby Fundus Dystrophy (SFD) is a rare form of macular degeneration that is clinically similar to age-related macular degeneration (AMD), and a histologic hallmark of SFD is a thick layer of extracellular deposits beneath the retinal pigment epithelium (RPE). Previous studies of SFD patient-induced pluripotent stem cell (iPSC) derived RPE differ as to whether these cultures recapitulate this key clinical feature by forming increased drusenoid deposits. The primary purpose of this study is to examine whether SFD patient-derived iPSC-RPE form basal deposits similar to what is found in affected family member SFD globes and to determine whether SFD iPSC RPE may be more oxidatively stressed. We performed a careful comparison of iPSC RPE from three control individuals, multiple iPSC clones from two SFD patients' iPSC RPE, and post-mortem eyes of affected SFD family members. We also examined the effect of CRISPR-Cas9 gene correction of the S204C TIMP3 mutation on RPE phenotype. Finally, targeted metabolomics with liquid chromatography and mass spectrometry analysis and stable isotope-labeled metabolite analysis were performed to determine whether SFD RPE are more oxidatively stressed. We found that SFD iPSC-RPE formed significantly more sub-RPE deposits (∼6-90 µm in height) compared to control RPE at 8 weeks. These deposits were similar in composition to the thick layer of sub-RPE deposits found in SFD family member globes by immunofluorescence staining and TEM imaging. S204C TIMP3 correction by CRISPR-Cas9 gene editing in SFD iPSC RPE cells resulted in significantly reduced basal laminar and sub-RPE calcium deposits. We detected a ∼18-fold increase in TIMP3 accumulation in the extracellular matrix (ECM) of SFD RPE, and targeted metabolomics showed that intracellular 4-hydroxyproline, a major breakdown product of collagen, is significantly elevated in SFD RPE, suggesting increased ECM turnover. Finally, SFD RPE cells have decreased intracellular reduced glutathione and were found to be more vulnerable to oxidative stress. Our findings suggest that elements of SFD pathology can be demonstrated in culture which may lead to insights into disease mechanisms.


Assuntos
Células-Tronco Pluripotentes Induzidas , Degeneração Macular , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Degeneração Macular/metabolismo , Epitélio Pigmentado da Retina/metabolismo
7.
Proc Natl Acad Sci U S A ; 116(9): 3530-3535, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808746

RESUMO

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.


Assuntos
Mitocôndrias/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/genética , Retina/metabolismo , Visão Ocular/genética , Animais , Glucose/metabolismo , Glicólise/genética , Humanos , Camundongos , Mitocôndrias/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos , Ácido Pirúvico/metabolismo , Retina/patologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Células Fotorreceptoras Retinianas Cones/patologia , Degeneração Retiniana , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia
8.
J Biol Chem ; 295(19): 6482-6497, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32238432

RESUMO

Cone photoreceptors in the retina enable vision over a wide range of light intensities. However, the processes enabling cone vision in bright light (i.e. photopic vision) are not adequately understood. Chromophore regeneration of cone photopigments may require the retinal pigment epithelium (RPE) and/or retinal Müller glia. In the RPE, isomerization of all-trans-retinyl esters to 11-cis-retinol is mediated by the retinoid isomerohydrolase Rpe65. A putative alternative retinoid isomerase, dihydroceramide desaturase-1 (DES1), is expressed in RPE and Müller cells. The retinol-isomerase activities of Rpe65 and Des1 are inhibited by emixustat and fenretinide, respectively. Here, we tested the effects of these visual cycle inhibitors on immediate, early, and late phases of cone photopic vision. In zebrafish larvae raised under cyclic light conditions, fenretinide impaired late cone photopic vision, while the emixustat-treated zebrafish unexpectedly had normal vision. In contrast, emixustat-treated larvae raised under extensive dark-adaptation displayed significantly attenuated immediate photopic vision concomitant with significantly reduced 11-cis-retinaldehyde (11cRAL). Following 30 min of light, early photopic vision was recovered, despite 11cRAL levels remaining significantly reduced. Defects in immediate cone photopic vision were rescued in emixustat- or fenretinide-treated larvae following exogenous 9-cis-retinaldehyde supplementation. Genetic knockout of Des1 (degs1) or retinaldehyde-binding protein 1b (rlbp1b) did not eliminate photopic vision in zebrafish. Our findings define molecular and temporal requirements of the nonphotopic or photopic visual cycles for mediating vision in bright light.


Assuntos
Visão de Cores , Células Ependimogliais/metabolismo , Células Fotorreceptoras Retinianas Cones/metabolismo , Peixe-Zebra/metabolismo , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Ependimogliais/citologia , Ácidos Graxos Dessaturases/genética , Ácidos Graxos Dessaturases/metabolismo , Deleção de Genes , Células Fotorreceptoras Retinianas Cones/citologia , Vitamina A/genética , Vitamina A/metabolismo , Peixe-Zebra/genética , cis-trans-Isomerases/genética , cis-trans-Isomerases/metabolismo
9.
Glia ; 69(8): 1966-1986, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33835598

RESUMO

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.


Assuntos
Células Fotorreceptoras , Degeneração Retiniana , Animais , Células Ependimogliais/metabolismo , Camundongos , Neuroglia/metabolismo , Células Fotorreceptoras/metabolismo , Retina/metabolismo , Degeneração Retiniana/metabolismo
10.
Adv Exp Med Biol ; 1256: 237-264, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33848005

RESUMO

Strong experimental evidence from studies in human donor retinas and animal models supports the idea that the retinal pathology associated with age-related macular degeneration (AMD) involves mitochondrial dysfunction and consequent altered retinal metabolism. This chapter provides a brief overview of mitochondrial structure and function, summarizes evidence for mitochondrial defects in AMD, and highlights the potential ramifications of these defects on retinal health and function. Discussion of mitochondrial haplogroups and their association with AMD brings to light how mitochondrial genetics can influence disease outcome. As one of the most metabolically active tissues in the human body, there is strong evidence that disruption in key metabolic pathways contributes to AMD pathology. The section on retinal metabolism reviews cell-specific metabolic differences and how the metabolic interdependence of each retinal cell type creates a unique ecosystem that is disrupted in the diseased retina. The final discussion includes strategies for therapeutic interventions that target key mitochondrial pathways as a treatment for AMD.


Assuntos
DNA Mitocondrial , Degeneração Macular , Animais , DNA Mitocondrial/metabolismo , Ecossistema , Humanos , Degeneração Macular/genética , Degeneração Macular/metabolismo , Mitocôndrias/genética , Retina , Epitélio Pigmentado da Retina/metabolismo
11.
Proc Natl Acad Sci U S A ; 113(51): 14710-14715, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-27911769

RESUMO

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.


Assuntos
Carbono/química , Olho/embriologia , Ácidos Cetoglutáricos/química , Degeneração Macular/metabolismo , Epitélio Pigmentado da Retina/embriologia , Epitélio Pigmentado da Retina/metabolismo , Idoso de 80 Anos ou mais , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Ácidos Graxos/química , Feminino , Células HeLa , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Isocitrato Desidrogenase/metabolismo , Degeneração Macular/patologia , Camundongos , NAD/química , Neoplasias/metabolismo , Neoplasias/patologia , Oxirredução , Estresse Oxidativo , Oxigênio/química , Poli(ADP-Ribose) Polimerases/metabolismo
12.
J Neurosci ; 37(8): 2061-2072, 2017 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-28115482

RESUMO

Ca2+ ions have distinct roles in the outer segment, cell body, and synaptic terminal of photoreceptors. We tested the hypothesis that distinct Ca2+ domains are maintained by Ca2+ uptake into mitochondria. Serial block face scanning electron microscopy of zebrafish cones revealed that nearly 100 mitochondria cluster at the apical side of the inner segment, directly below the outer segment. The endoplasmic reticulum surrounds the basal and lateral surfaces of this cluster, but does not reach the apical surface or penetrate into the cluster. Using genetically encoded Ca2+ sensors, we found that mitochondria take up Ca2+ when it accumulates either in the cone cell body or outer segment. Blocking mitochondrial Ca2+ uniporter activity compromises the ability of mitochondria to maintain distinct Ca2+ domains. Together, our findings indicate that mitochondria can modulate subcellular functional specialization in photoreceptors.SIGNIFICANCE STATEMENT Ca2+ homeostasis is essential for the survival and function of retinal photoreceptors. Separate pools of Ca2+ regulate phototransduction in the outer segment, metabolism in the cell body, and neurotransmitter release at the synaptic terminal. We investigated the role of mitochondria in compartmentalization of Ca2+ We found that mitochondria form a dense cluster that acts as a diffusion barrier between the outer segment and cell body. The cluster is surprisingly only partially surrounded by the endoplasmic reticulum, a key mediator of mitochondrial Ca2+ uptake. Blocking the uptake of Ca2+ by mitochondria causes redistribution of Ca2+ throughout the cell. Our results show that mitochondrial Ca2+ uptake in photoreceptors is complex and plays an essential role in normal function.


Assuntos
Cálcio/metabolismo , Mitocôndrias/metabolismo , Retina/citologia , Células Fotorreceptoras Retinianas Cones/ultraestrutura , Animais , Animais Geneticamente Modificados , Antiarrítmicos/farmacologia , Compostos de Boro/farmacocinética , Calmodulina/genética , Calmodulina/metabolismo , Citosol/metabolismo , Corantes Fluorescentes/farmacocinética , Proteínas Heterotriméricas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Técnicas In Vitro , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mitocôndrias/genética , Mitocôndrias/ultraestrutura , Cloreto de Potássio/farmacologia , Frações Subcelulares/metabolismo , Frações Subcelulares/ultraestrutura , Sinapses/metabolismo , Tioureia/análogos & derivados , Tioureia/farmacologia , Peixe-Zebra , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
13.
J Biol Chem ; 292(31): 12895-12905, 2017 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-28615447

RESUMO

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.


Assuntos
Prolina/metabolismo , Retina/metabolismo , Epitélio Pigmentado da Retina/metabolismo , Animais , Transporte Biológico , Isótopos de Carbono , Polaridade Celular , Células Cultivadas , Ácido Cítrico/metabolismo , Ciclo do Ácido Cítrico , Técnicas de Cocultura , Embrião de Mamíferos/citologia , Glucose/metabolismo , Humanos , Cinética , Metabolômica/métodos , Camundongos , Retina/citologia , Retina/enzimologia , Epitélio Pigmentado da Retina/citologia , Epitélio Pigmentado da Retina/enzimologia , Taurina/metabolismo , Técnicas de Cultura de Tecidos
14.
Exp Eye Res ; 174: 113-120, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29864440

RESUMO

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.


Assuntos
Dióxido de Carbono/farmacologia , Corioide , Dissecação , Eutanásia , Metaboloma/efeitos dos fármacos , Epitélio Pigmentado da Retina , Trifosfato de Adenosina/metabolismo , Animais , Corioide/efeitos dos fármacos , Corioide/metabolismo , Cromatografia Líquida , Modelos Animais de Doenças , Glucose/metabolismo , Guanosina Trifosfato/metabolismo , Camundongos , Epitélio Pigmentado da Retina/efeitos dos fármacos , Epitélio Pigmentado da Retina/metabolismo , Espectrometria de Massas em Tandem , Fatores de Tempo
15.
Adv Exp Med Biol ; 1074: 289-295, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721955

RESUMO

Aryl-hydrocarbon receptor interacting protein-like 1 (AIPL1) is essential to stabilize cGMP phosphodiesterase 6 (PDE6) in rod photoreceptors. Mutation of AIPL1 leads to loss of PDE6, accumulation of intracellular cGMP, and rapid degeneration of rods. To understand the metabolic basis for the photoreceptor degeneration caused by excessive cGMP, we performed proteomics and phosphoproteomics analyses on retinas from AIPL1-/- mice at the onset of rod cell death. AIPL1-/- retinas have about 18 times less than normal PDE6a and no detectable PDE6b. We identified twelve other proteins and thirty-nine phosphorylated proteins related to cell metabolism that are significantly altered preceding the massive degeneration of rods. They include transporters, kinases, phosphatases, transferases, and proteins involved in mitochondrial bioenergetics and metabolism of glucose, lipids, amino acids, nucleotides, and RNA. In AIPLI-/- retinas mTOR and proteins involved in mitochondrial energy production and lipid synthesis are more dephosphorylated, but glycolysis proteins and proteins involved in leucine catabolism are more phosphorylated than in normal retinas. Our findings indicate that elevating cGMP rewires cellular metabolism prior to photoreceptor degeneration and that targeting metabolism may be a productive strategy to prevent or slow retinal degeneration.


Assuntos
GMP Cíclico/metabolismo , Metabolismo Energético/genética , Proteínas do Olho/metabolismo , Fosfoproteínas/metabolismo , Processamento de Proteína Pós-Traducional , Retina/metabolismo , Degeneração Retiniana/metabolismo , Células Fotorreceptoras Retinianas Bastonetes/patologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Retina/patologia , Degeneração Retiniana/patologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo
16.
J Biol Chem ; 291(9): 4698-710, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26677218

RESUMO

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.


Assuntos
Sinalização do Cálcio/efeitos da radiação , Metabolismo Energético/efeitos da radiação , Proteínas do Olho/metabolismo , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Proteínas Heterotriméricas de Ligação ao GTP/metabolismo , Transdução de Sinal Luminoso , Retina/efeitos da radiação , Transducina/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animais , Antiporters/metabolismo , Ciclo do Ácido Cítrico/efeitos da radiação , GMP Cíclico/metabolismo , Transporte de Elétrons/efeitos da radiação , Proteínas do Olho/genética , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Glicólise/efeitos da radiação , Proteínas Heterotriméricas de Ligação ao GTP/genética , Complexo Cetoglutarato Desidrogenase/metabolismo , Luz , Metaboloma/efeitos da radiação , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Consumo de Oxigênio/efeitos da radiação , Retina/enzimologia , Retina/metabolismo , Técnicas de Cultura de Tecidos , Transducina/genética
17.
Proc Natl Acad Sci U S A ; 111(43): 15579-84, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25313047

RESUMO

Symbiotic relationships between neurons and glia must adapt to structures, functions, and metabolic roles of the tissues they are in. We show here that Müller glia in retinas have specific enzyme deficiencies that can enhance their ability to synthesize Gln. The metabolic cost of these deficiencies is that they impair the Müller cell's ability to metabolize Glc. We show here that the cells can compensate for this deficiency by using metabolites produced by neurons. Müller glia are deficient for pyruvate kinase (PK) and for aspartate/glutamate carrier 1 (AGC1), a key component of the malate-aspartate shuttle. In contrast, photoreceptor neurons express AGC1 and the M2 isoform of pyruvate kinase, which is commonly associated with aerobic glycolysis in tumors, proliferating cells, and some other cell types. Our findings reveal a previously unidentified type of metabolic relationship between neurons and glia. Müller glia compensate for their unique metabolic adaptations by using lactate and aspartate from neurons as surrogates for their missing PK and AGC1.


Assuntos
Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Antiporters/metabolismo , Neuroglia/metabolismo , Piruvato Quinase/metabolismo , Neurônios Retinianos/metabolismo , Animais , Ácido Aspártico/metabolismo , Isótopos de Carbono , Células Cultivadas , Células Ependimogliais/metabolismo , Células Ependimogliais/efeitos da radiação , Glucose/metabolismo , Glutamina/metabolismo , Glicólise , Células HeLa , Humanos , Isoenzimas/metabolismo , Lactose/metabolismo , Luz , Camundongos , Modelos Biológicos , Neuroglia/efeitos da radiação , Oxirredução/efeitos da radiação , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/efeitos da radiação , Neurônios Retinianos/efeitos da radiação
18.
Mol Vis ; 22: 1198-1212, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27746674

RESUMO

PURPOSE: To characterize the vision phenotype of mice lacking Aralar/AGC1/Slc25a12, the mitochondrial aspartate-glutamate carrier mutated in global cerebral hypomyelination (OMIM 612949). METHODS: We tested overnight dark-adapted control and aralar-deficient mice for the standard full electroretinogram (ERG) response. The metabolic stress of dark-adaptation was reduced by 5 min illumination after which the ERG response was monitored in darkness. We used the electrical response to two identical saturating light flashes (paired-flash stimulation) to isolate the inner retina and photoreceptor responses. Retinal morphology was examined with hematoxylin and eosin staining, immunohistochemistry of antibodies against retinal cells, and 4',6-diamidino-2-phenylindole (DAPI) labeling. RESULTS: Aralar plays a pivotal role in retina metabolism as aralar provides de novo synthesis pathway for glutamine, protects glutamate from oxidation, and is required for efficient glucose oxidative metabolism. Aralar-deficient mice are not blind as their retinas have light-evoked activity. However, we report an approximate 50% decrease in the ERG amplitude response in the light-evoked activity of dark-adapted retinas from aralar-deficient mice, in spite of normal retina histology. The defective response is partly reversed by exposure to a brief illumination period, which lowers the metabolic stress of dark-adaptation. The metabolic stress and ERG alteration takes place primarily in photoreceptors, but the response to two flashes applied in fast succession also revealed an alteration in synaptic transmission consistent with an imbalance of glutamate and an energy deficit in the inner retina neurons. CONCLUSIONS: We propose that compromised glucose oxidation and altered glutamine and glutamate metabolism in the absence of aralar are responsible for the phenotype reported.


Assuntos
Agrecanas/fisiologia , Glucose/metabolismo , Glutamina/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/fisiologia , Retina/metabolismo , Transtornos da Visão/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/deficiência , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animais , Antiporters/deficiência , Antiporters/metabolismo , Adaptação à Escuridão , Eletrorretinografia , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Doenças Mitocondriais/metabolismo , Estimulação Luminosa , Transtornos Psicomotores/metabolismo , Retina/fisiopatologia , Células Bipolares da Retina/fisiologia , Transmissão Sináptica , Transtornos da Visão/fisiopatologia
19.
Proc Natl Acad Sci U S A ; 110(46): 18501-6, 2013 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-24127593

RESUMO

Glutamate in neurons is an important excitatory neurotransmitter, but it also is a key metabolite. We investigated how glutamate in a neural tissue is protected from catabolism. Flux analysis using (13)C-labeled fuels revealed that retinas use activities of the malate aspartate shuttle to protect >98% of their glutamate from oxidation in mitochondria. Isolation of glutamate from the oxidative pathway relies on cytosolic NADH/NAD(+), which is influenced by extracellular glucose, lactate, and pyruvate.


Assuntos
Citosol/metabolismo , Ácido Glutâmico/metabolismo , Retina/metabolismo , Análise de Variância , Animais , Isótopos de Carbono/metabolismo , Cromatografia Gasosa-Espectrometria de Massas , Células HeLa , Humanos , Análise do Fluxo Metabólico , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Oxirredução
20.
J Neurosci Res ; 93(7): 1079-92, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25801286

RESUMO

The vertebrate retina has specific functions and structures that give it a unique set of constraints on the way in which it can produce and use metabolic energy. The retina's response to illumination influences its energy requirements, and the retina's laminated structure influences the extent to which neurons and glia can access metabolic fuels. There are fundamental differences between energy metabolism in retina and that in brain. The retina relies on aerobic glycolysis much more than the brain does, and morphological differences between retina and brain limit the types of metabolic relationships that are possible between neurons and glia. This Mini-Review summarizes the unique metabolic features of the retina with a focus on the role of lactate shuttling.


Assuntos
Metabolismo Energético/fisiologia , Glucose/metabolismo , Ácido Láctico/metabolismo , Retina/metabolismo , Animais , Humanos , Vertebrados
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA