1
|
Maurya M, Liu CH, Bora K, Kushwah N, Pavlovich MC, Wang Z, Chen J. Animal Models of Retinopathy of Prematurity: Advances and Metabolic Regulators. Biomedicines 2024; 12:1937. [PMID: 39335451 PMCID: PMC11428941 DOI: 10.3390/biomedicines12091937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 09/30/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a primary cause of visual impairment and blindness in premature newborns, characterized by vascular abnormalities in the developing retina, with microvascular alteration, neovascularization, and in the most severe cases retinal detachment. To elucidate the pathophysiology and develop therapeutics for ROP, several pre-clinical experimental models of ROP were developed in different species. Among them, the oxygen-induced retinopathy (OIR) mouse model has gained the most popularity and critically contributed to our current understanding of pathological retinal angiogenesis and the discovery of potential anti-angiogenic therapies. A deeper comprehension of molecular regulators of OIR such as hypoxia-inducible growth factors including vascular endothelial growth factors as primary perpetrators and other new metabolic modulators such as lipids and amino acids influencing pathological retinal angiogenesis is also emerging, indicating possible targets for treatment strategies. This review delves into the historical progressions that gave rise to the modern OIR models with a focus on the mouse model. It also reviews the fundamental principles of OIR, recent advances in its automated assessment, and a selected summary of metabolic investigation enabled by OIR models including amino acid transport and metabolism.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jing Chen
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
2
|
Zhang C, Gu L, Xie H, Liu Y, Huang P, Zhang J, Luo D, Zhang J. Glucose transport, transporters and metabolism in diabetic retinopathy. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166995. [PMID: 38142757 DOI: 10.1016/j.bbadis.2023.166995] [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: 08/02/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 12/26/2023]
Abstract
Diabetic retinopathy (DR) is the most common reason for blindness in working-age individuals globally. Prolonged high blood glucose is a main causative factor for DR development, and glucose transport is prerequisite for the disturbances in DR caused by hyperglycemia. Glucose transport is mediated by its transporters, including the facilitated transporters (glucose transporter, GLUTs), the "active" glucose transporters (sodium-dependent glucose transporters, SGLTs), and the SLC50 family of uniporters (sugars will eventually be exported transporters, SWEETs). Glucose transport across the blood-retinal barrier (BRB) is crucial for nourishing the neuronal retina in the context of retinal physiology. This physiological process primarily relies on GLUTs and SGLTs, which mediate the glucose transportation across both the cell membrane of retinal capillary endothelial cells and the retinal pigment epithelium (RPE). Under diabetic conditions, increased accumulation of extracellular glucose enhances the retinal cellular glucose uptake and metabolism via both glycolysis and glycolytic side branches, which activates several biochemical pathways, including the protein kinase C (PKC), advanced glycation end-products (AGEs), polyol pathway and hexosamine biosynthetic pathway (HBP). These activated biochemical pathways further increase the production of reactive oxygen species (ROS), leading to oxidative stress and activation of Poly (ADP-ribose) polymerase (PARP). The activated PARP further affects all the cellular components in the retina, and finally resulting in microangiopathy, neurodegeneration and low-to-moderate grade inflammation in DR. This review aims to discuss the changes of glucose transport, glucose transporters, as well as its metabolism in DR, which influences the retinal neurovascular unit (NVU) and implies the possible therapeutic strategies for treating DR.
Collapse
Affiliation(s)
- Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Limin Gu
- Department of Ophthalmology, Shanghai Aier Eye Hospital, Shanghai, China.
| | - Hai Xie
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Yan Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Peirong Huang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Jingting Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Dawei Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China; National Clinical Research Center for Eye Diseases; Shanghai Clinical Research Center for Eye Diseases; Shanghai Key Clinical Specialty; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine; Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Eye Research Institute, Shanghai, China.
| |
Collapse
|
3
|
Xu L, Ruddick WN, Bolch SN, Klingeborn M, Dyka FM, Kulkarni MM, Simpson CP, Beltran WA, Bowes Rickman C, Smith WC, Dinculescu A. Distinct Phenotypic Consequences of Pathogenic Mutants Associated with Late-Onset Retinal Degeneration. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1706-1720. [PMID: 36328299 PMCID: PMC10726427 DOI: 10.1016/j.ajpath.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/23/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
A pathologic feature of late-onset retinal degeneration caused by the S163R mutation in C1q-tumor necrosis factor-5 (C1QTNF5) is the presence of unusually thick deposits between the retinal pigmented epithelium (RPE) and the vascular choroid, considered a hallmark of this disease. Following its specific expression in mouse RPE, the S163R mutant exhibits a reversed polarized distribution relative to the apically secreted wild-type C1QTNF5, and forms widespread, prominent deposits that gradually increase in size with aging. The current study shows that S163R deposits expand to a considerable thickness through a progressive increase in the basolateral RPE membrane, substantially raising the total RPE height, and enabling their clear imaging as a distinct hyporeflective layer by noninvasive optical coherence tomography in advanced age animals. This phenotype bears a striking resemblance to ocular pathology previously documented in patients harboring the S163R mutation. Therefore, a similar viral vector-based gene delivery approach was used to also investigate the behavior of P188T and G216C, two novel pathogenic C1QTNF5 mutants recently reported in patients for which histopathologic data are lacking. Both mutants primarily impacted the RPE/photoreceptor interface and did not generate basal laminar deposits. Distinct distribution patterns and phenotypic consequences of C1QTNF5 mutants were observed in vivo, which suggested that multiple pathobiological mechanisms contribute to RPE dysfunction and vision loss in this disorder.
Collapse
Affiliation(s)
- Lei Xu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida
| | - William N Ruddick
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida
| | - Susan N Bolch
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida
| | - Mikael Klingeborn
- McLaughlin Research Institute, Great Falls, Montana; Helen Wills Neuroscience Institute, Berkeley, California
| | - Frank M Dyka
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida
| | - Manoj M Kulkarni
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Chiab P Simpson
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida
| | - William A Beltran
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina
| | - Catherine Bowes Rickman
- Helen Wills Neuroscience Institute, Berkeley, California; Department of Cell Biology, Duke University Medical Center, Durham, North Carolina
| | - W Clay Smith
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida
| | - Astra Dinculescu
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, Florida.
| |
Collapse
|
4
|
Aiello JJ, Bogart MC, Chan WT, Holoman NC, Trobenter TD, Relf CE, Kleinman DM, De Vivo DC, Samuels IS. Systemic Reduction of Glut1 Normalizes Retinal Dysfunction, Inflammation, and Oxidative Stress in the Retina of Spontaneous Type 2 Diabetic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:927-938. [PMID: 37062410 PMCID: PMC10294444 DOI: 10.1016/j.ajpath.2023.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 04/18/2023]
Abstract
Defects in the light-evoked responses of the retina occur early in the sequalae of diabetic retinopathy (DR). These defects, identified through the electroretinogram (ERG), represent dysfunction of retinal neurons and the retinal pigment epithelium and are commonly identifiable at the timing of, or almost immediately following, diabetes diagnosis. Recently, systemic reduction of the facilitated glucose transporter type 1, Glut1, in type 1 diabetic mice was shown to reduce retinal sorbitol accumulation, mitigate ERG defects, and prevent retinal oxidative stress and inflammation. Herein, the study investigated whether systemic reduction of Glut1 also diminished hallmarks of DR in type 2 diabetic mice. Transgenic nondiabetic Leprdb/+ and spontaneously diabetic Leprdb/db mice that expressed wild-type (Glut1+/+) or systemically reduced levels of Glut1 (Glut1+/-) were aged and subjected to standard strobe flash electroretinography and c-wave analysis before evaluation of inflammatory cytokines and oxidative stress molecules. Although Leprdb/dbGlut1+/- mice still displayed overt obesity and diabetes, no scotopic, photopic, or c-wave ERG defects were present through 16 weeks of age, and expression of inflammatory cytokines and oxidative stress molecules was also normalized. These findings suggest that systemic reduction of Glut1 is sufficient to prevent functional retinal pathophysiology in type 2 diabetes. Targeted, moderate reductions of Glut1 or inhibition of Glut1 activity in the retina of diabetic patients should be considered as a novel therapeutic strategy to prevent development and progression of DR.
Collapse
Affiliation(s)
- Jacob J Aiello
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | - Maislin C Bogart
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | - Wai-Ting Chan
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | - Nicholas C Holoman
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | - Timothy D Trobenter
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | - Chloe E Relf
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | - Dana M Kleinman
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, Ohio
| | - Darryl C De Vivo
- Departments of Neurology and Pediatrics, Columbia University Irving Medical Center, New York, New York
| | - Ivy S Samuels
- Louis Stokes Cleveland VA Medical Center, VA Northeast Ohio Healthcare System, Cleveland, Ohio; Department of Ophthalmic Research, Cole Eye Institute, Cleveland, Ohio.
| |
Collapse
|
5
|
Liu Y, Wang X, Gong R, Xu G, Zhu M. Overexpression of Rhodopsin or Its Mutants Leads to Energy Metabolism Dysfunction in 661w Cells. Invest Ophthalmol Vis Sci 2022; 63:2. [PMID: 36469028 PMCID: PMC9730732 DOI: 10.1167/iovs.63.13.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Retinitis pigmentosa (RP) is a heterogeneous group of inherited disorders characterized by photoreceptor degeneration. The rhodopsin gene (RHO) is the most frequent cause of autosomal dominant RP (ADRP), yet it remains unclear how RHO mutations cause heterogeneous phenotypes. Energy failure is a main cause of the secondary cone death during RP progression; however, its role in primary rod death induced by ADRP RHO mutants is unknown. Methods Three RHO missense mutations were chosen from different clinical classes. Wild-type (WT) RHO and its mutants, P23H (class B1), R135L (class A), and G188R (class B2), were overexpressed in 661w cells, a mouse photoreceptor cell line, and their effects on oxidative phosphorylation (OXPHOS) and aerobic glycolysis were compared separately. Results Here, we report that energy failure is an early event in the cell death caused by overexpression of WT RHO and its mutants. RHO overexpression leads to OXPHOS deficiency, which might be a result of mitochondrial loss. Nonetheless, only in WT RHO and P23H groups, energy stress triggers AMP-activated protein kinase activation and metabolic reprogramming to increase glycolysis. Metabolic reprogramming impairment in R135L and G188R groups might be the reason why energy failure and cell injury are much more severe in those groups. Conclusions Our results imply that overexpression of RHO missense mutants have distinct impacts on the two energy metabolic pathways, which might be related to their heterogeneous phenotypes.
Collapse
Affiliation(s)
- Yang Liu
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Xin Wang
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Ruowen Gong
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Gezhi Xu
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| | - Min Zhu
- Shanghai Key Laboratory of Visual Impairment and Restoration, Eye & ENT Hospital, Fudan University, Shanghai, China,NHC Key Laboratory of Myopia (Fudan University); Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
| |
Collapse
|
6
|
Daniele LL, Han JY, Samuels IS, Komirisetty R, Mehta N, McCord JL, Yu M, Wang Y, Boesze-Battaglia K, Bell BA, Du J, Peachey NS, Philp NJ. Glucose uptake by GLUT1 in photoreceptors is essential for outer segment renewal and rod photoreceptor survival. FASEB J 2022; 36:e22428. [PMID: 35766190 PMCID: PMC9438481 DOI: 10.1096/fj.202200369r] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/27/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
Photoreceptors consume glucose supplied by the choriocapillaris to support phototransduction and outer segment (OS) renewal. Reduced glucose supply underlies photoreceptor cell death in inherited retinal degeneration and age-related retinal disease. We have previously shown that restricting glucose transport into the outer retina by conditional deletion of Slc2a1 encoding GLUT1 resulted in photoreceptor loss and impaired OS renewal. However, retinal neurons, glia, and the retinal pigment epithelium play specialized, synergistic roles in metabolite supply and exchange, and the cell-specific map of glucose uptake and utilization in the retina is incomplete. In these studies, we conditionally deleted Slc2a1 in a pan-retinal or rod-specific manner to better understand how glucose is utilized in the retina. Using non-invasive ocular imaging, electroretinography, and histochemical and biochemical analyses we show that genetic deletion of Slc2a1 from retinal neurons and Müller glia results in reduced OS growth and progressive rod but not cone photoreceptor cell death. Rhodopsin levels were severely decreased even at postnatal day 20 when OS length was relatively normal. Arrestin levels were not changed suggesting that glucose uptake is required to synthesize membrane glycoproteins. Rod-specific deletion of Slc2a1 resulted in similar changes in OS length and rod photoreceptor cell death. These studies demonstrate that glucose is an essential carbon source for rod photoreceptor cell OS maintenance and viability.
Collapse
Affiliation(s)
- Lauren L. Daniele
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - John Y.S. Han
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - Ivy S. Samuels
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH
- Louis Stokes Cleveland VA Medical Center, Cleveland,
OH
| | - Ravikiran Komirisetty
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - Nikhil Mehta
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - Jessica L. McCord
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| | - Minzhong Yu
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH
- Department of Ophthalmology, Cleveland Clinic Lerner
College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Yekai Wang
- Department of Ophthalmology and Visual Sciences, West
Virginia University, Morgantown, WV
- Department of Biochemistry, West Virginia University,
Morgantown, WV
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, Penn Dental
Medicine, University of Pennsylvania, Philadelphia, PA
| | - Brent A. Bell
- Department of Ophthalmology, University of Pennsylvania,
Philadelphia, PA
| | - Jianhai Du
- Department of Ophthalmology and Visual Sciences, West
Virginia University, Morgantown, WV
- Department of Biochemistry, West Virginia University,
Morgantown, WV
| | - Neal S. Peachey
- Cole Eye Institute, Cleveland Clinic, Cleveland, OH
- Louis Stokes Cleveland VA Medical Center, Cleveland,
OH
- Department of Ophthalmology, Cleveland Clinic Lerner
College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Nancy J. Philp
- Department of Pathology, Anatomy, and Cell Biology, Thomas
Jefferson University, Philadelphia, PA
| |
Collapse
|
7
|
Functional compartmentalization of photoreceptor neurons. Pflugers Arch 2021; 473:1493-1516. [PMID: 33880652 DOI: 10.1007/s00424-021-02558-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/15/2021] [Accepted: 03/22/2021] [Indexed: 12/16/2022]
Abstract
Retinal photoreceptors are neurons that convert dynamically changing patterns of light into electrical signals that are processed by retinal interneurons and ultimately transmitted to vision centers in the brain. They represent the essential first step in seeing without which the remainder of the visual system is rendered moot. To support this role, the major functions of photoreceptors are segregated into three main specialized compartments-the outer segment, the inner segment, and the pre-synaptic terminal. This compartmentalization is crucial for photoreceptor function-disruption leads to devastating blinding diseases for which therapies remain elusive. In this review, we examine the current understanding of the molecular and physical mechanisms underlying photoreceptor functional compartmentalization and highlight areas where significant knowledge gaps remain.
Collapse
|
8
|
Viegas FO, Neuhauss SCF. A Metabolic Landscape for Maintaining Retina Integrity and Function. Front Mol Neurosci 2021; 14:656000. [PMID: 33935647 PMCID: PMC8081888 DOI: 10.3389/fnmol.2021.656000] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/22/2021] [Indexed: 01/27/2023] Open
Abstract
Neurons have high metabolic demands that are almost exclusively met by glucose supplied from the bloodstream. Glucose is utilized in complex metabolic interactions between neurons and glia cells, described by the astrocyte-neuron lactate shuttle (ANLS) hypothesis. The neural retina faces similar energy demands to the rest of the brain, with additional high anabolic needs to support continuous renewal of photoreceptor outer segments. This demand is met by a fascinating variation of the ANLS in which photoreceptors are the central part of a metabolic landscape, using glucose and supplying surrounding cells with metabolic intermediates. In this review we summarize recent evidence on how neurons, in particular photoreceptors, meet their energy and biosynthetic requirements by comprising a metabolic landscape of interdependent cells.
Collapse
Affiliation(s)
- Filipe O Viegas
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Life Science Zurich Graduate School, Ph.D. Program in Molecular Life Sciences, Zurich, Switzerland
| | - Stephan C F Neuhauss
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| |
Collapse
|
9
|
Xue Y, Wang SK, Rana P, West ER, Hong CM, Feng H, Wu DM, Cepko CL. AAV-Txnip prolongs cone survival and vision in mouse models of retinitis pigmentosa. eLife 2021; 10:e66240. [PMID: 33847261 PMCID: PMC8081528 DOI: 10.7554/elife.66240] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/30/2021] [Indexed: 01/14/2023] Open
Abstract
Retinitis pigmentosa (RP) is an inherited retinal disease affecting >20 million people worldwide. Loss of daylight vision typically occurs due to the dysfunction/loss of cone photoreceptors, the cell type that initiates our color and high-acuity vision. Currently, there is no effective treatment for RP, other than gene therapy for a limited number of specific disease genes. To develop a disease gene-agnostic therapy, we screened 20 genes for their ability to prolong cone photoreceptor survival in vivo. Here, we report an adeno-associated virus vector expressing Txnip, which prolongs the survival of cone photoreceptors and improves visual acuity in RP mouse models. A Txnip allele, C247S, which blocks the association of Txnip with thioredoxin, provides an even greater benefit. Additionally, the rescue effect of Txnip depends on lactate dehydrogenase b (Ldhb) and correlates with the presence of healthier mitochondria, suggesting that Txnip saves RP cones by enhancing their lactate catabolism.
Collapse
Affiliation(s)
- Yunlu Xue
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
- Department of Ophthalmology, Harvard Medical SchoolBostonUnited States
| | - Sean K Wang
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
- Department of Ophthalmology, Harvard Medical SchoolBostonUnited States
- Howard Hughs Medical InstituteChevy ChaseUnited States
| | - Parimal Rana
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
| | - Emma R West
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
- Howard Hughs Medical InstituteChevy ChaseUnited States
| | - Christin M Hong
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
- Howard Hughs Medical InstituteChevy ChaseUnited States
| | - Helian Feng
- Department of Biostatistics, Harvard T.H. Chan School of Public HealthBostonUnited States
| | - David M Wu
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
- Department of Ophthalmology, Harvard Medical SchoolBostonUnited States
- Retina Service, Massachusetts Eye and Ear Infirmary, Harvard Medical SchoolBostonUnited States
| | - Constance L Cepko
- Department of Genetics, Blavatnik Institute, Harvard Medical SchoolBostonUnited States
- Department of Ophthalmology, Harvard Medical SchoolBostonUnited States
- Howard Hughs Medical InstituteChevy ChaseUnited States
| |
Collapse
|
10
|
Reduction of Glut1 in the Neural Retina But Not the RPE Alleviates Polyol Accumulation and Normalizes Early Characteristics of Diabetic Retinopathy. J Neurosci 2021; 41:3275-3299. [PMID: 33622781 DOI: 10.1523/jneurosci.2010-20.2021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/16/2020] [Accepted: 02/11/2021] [Indexed: 01/04/2023] Open
Abstract
Hyperglycemia is a key determinant for development of diabetic retinopathy (DR). Inadequate glycemic control exacerbates retinopathy, while normalization of glucose levels delays its progression. In hyperglycemia, hexokinase is saturated and excess glucose is metabolized to sorbitol by aldose reductase via the polyol pathway. Therapies to reduce retinal polyol accumulation for the prevention of DR have been elusive because of low sorbitol dehydrogenase levels in the retina and inadequate inhibition of aldose reductase. Using systemic and conditional genetic inactivation, we targeted the primary facilitative glucose transporter in the retina, Glut1, as a preventative therapeutic in diabetic male and female mice. Unlike WT diabetics, diabetic Glut1 +/- mice did not display elevated Glut1 levels in the retina. Furthermore, diabetic Glut1 +/- mice exhibited ameliorated ERG defects, inflammation, and oxidative stress, which was correlated with a significant reduction in retinal sorbitol accumulation. Retinal pigment epithelium-specific reduction of Glut1 did not prevent an increase in retinal sorbitol content or early hallmarks of DR. However, like diabetic Glut1 +/- mice, reduction of Glut1 specifically in the retina mitigated polyol accumulation and diminished retinal dysfunction and the elevation of markers for oxidative stress and inflammation associated with diabetes. These results suggest that modulation of retinal polyol accumulation via Glut1 in photoreceptors can circumvent the difficulties in regulating systemic glucose metabolism and be exploited to prevent DR.SIGNIFICANCE STATEMENT Diabetic retinopathy affects one-third of diabetic patients and is the primary cause of vision loss in adults 20-74 years of age. While anti-VEGF and photocoagulation treatments for the late-stage vision threatening complications can prevent vision loss, a significant proportion of patients do not respond to anti-VEGF therapies, and mechanisms to stop progression of early-stage symptoms remain elusive. Glut1 is the primary facilitative glucose transporter for the retina. We determined that a moderate reduction in Glut1 levels, specifically in the retina, but not the retinal pigment epithelium, was sufficient to prevent retinal polyol accumulation and the earliest functional defects to be identified in the diabetic retina. Our study defines modulation of Glut1 in retinal neurons as a targetable molecule for prevention of diabetic retinopathy.
Collapse
|
11
|
Jaroszynska N, Harding P, Moosajee M. Metabolism in the Zebrafish Retina. J Dev Biol 2021; 9:10. [PMID: 33804189 PMCID: PMC8006245 DOI: 10.3390/jdb9010010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Retinal photoreceptors are amongst the most metabolically active cells in the body, consuming more glucose as a metabolic substrate than even the brain. This ensures that there is sufficient energy to establish and maintain photoreceptor functions during and after their differentiation. Such high dependence on glucose metabolism is conserved across vertebrates, including zebrafish from early larval through to adult retinal stages. As the zebrafish retina develops rapidly, reaching an adult-like structure by 72 hours post fertilisation, zebrafish larvae can be used to study metabolism not only during retinogenesis, but also in functionally mature retinae. The interplay between rod and cone photoreceptors and the neighbouring retinal pigment epithelium (RPE) cells establishes a metabolic ecosystem that provides essential control of their individual functions, overall maintaining healthy vision. The RPE facilitates efficient supply of glucose from the choroidal vasculature to the photoreceptors, which produce metabolic products that in turn fuel RPE metabolism. Many inherited retinal diseases (IRDs) result in photoreceptor degeneration, either directly arising from photoreceptor-specific mutations or secondary to RPE loss, leading to sight loss. Evidence from a number of vertebrate studies suggests that the imbalance of the metabolic ecosystem in the outer retina contributes to metabolic failure and disease pathogenesis. The use of larval zebrafish mutants with disease-specific mutations that mirror those seen in human patients allows us to uncover mechanisms of such dysregulation and disease pathology with progression from embryonic to adult stages, as well as providing a means of testing novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Philippa Harding
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
| | - Mariya Moosajee
- Institute of Ophthalmology, University College London, London EC1V 9EL, UK;
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
12
|
Madrakhimov SB, Yang JY, Kim JH, Han JW, Park TK. mTOR-dependent dysregulation of autophagy contributes to the retinal ganglion cell loss in streptozotocin-induced diabetic retinopathy. Cell Commun Signal 2021; 19:29. [PMID: 33637094 PMCID: PMC7913405 DOI: 10.1186/s12964-020-00698-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Neurodegeneration, an early event in the pathogenesis of diabetic retinopathy (DR), precedes clinically detectable microvascular damage. Autophagy dysregulation is considered a potential cause of neuronal cell loss, however underlying mechanisms remain unclear. The mechanistic target of rapamycin (mTOR) integrates diverse environmental signals to coordinate biological processes, including autophagy. Here, we investigated the role of mTOR signaling in neuronal cell death in DR. METHODS Diabetes was induced by a single intraperitoneal injection of streptozotocin and tissue samples were harvested at 1, 2, 3, 4, and 6 months of diabetes. Early-stage of DR was investigated in 1-month-diabetic mice treated with phlorizin (two daily subcutaneous injections at a dose of 200 mg/kg of body weight during the last 7 full days of the experiment and the morning of the 8th day, 3 h before sacrifice) or rapamycin (daily intraperitoneal injections, at a dose of 3 mg/kg for the same period as for phlorizin treatment). The effect of autophagy modulation on retinal ganglion cells was investigated in 3-months-diabetic mice treated with phlorizin (two daily subcutaneous injections during the last 10 full days of the experiment and the morning of the 11th day, 3 h before sacrifice) or MHY1485 (daily i.p. injections, at a dose of 10 mg/kg for the same period as for phlorizin treatment). Tissue samples obtained from treated/untreated diabetic mice and age-matched controls were used for Western blot and histologic analysis. RESULTS mTOR-related proteins and glucose transporter 1 (GLUT1) was upregulated at 1 month and downregulated in the following period up to 6 months. Diabetes-induced neurodegeneration was characterized by an increase of apoptotic marker-cleaved caspase 3, a decrease of the total number of cells, and NeuN immunoreactivity in the ganglion cell layer, as well as an increase of autophagic protein. Insulin-independent glycemic control restored the mTOR pathway activity and GLUT1 expression, along with a decrease of autophagic and apoptotic proteins in 3-months-diabetic mice neuroretina. However, blockade of autophagy using MHY1485 resulted in a more protective effect on ganglion cells compared with phlorizin treatment. CONCLUSION Collectively, our study describes the mechanisms of neurodegeneration through the hyperglycemia/ mTOR/ autophagy/ apoptosis pathway. Video Abstract.
Collapse
Affiliation(s)
- Sanjar Batirovich Madrakhimov
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research On Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jin Young Yang
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research On Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jin Ha Kim
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Jung Woo Han
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
| | - Tae Kwann Park
- Department of Interdisciplinary Program in Biomedical Science, Soonchunhyang Graduate School, Bucheon Hospital, Bucheon, South Korea
- Laboratory for Translational Research On Retinal and Macular Degeneration, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
- Department of Ophthalmology, Soonchunhyang University Hospital Bucheon, Bucheon, South Korea
- Department of Ophthalmology, College of Medicine, Soonchunhyang University, Choongchungnam-do, Cheonan, South Korea
- Department of Ophthalmology, College of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
- Ex Lumina Therapeutics and Technologies. Co., Ltd., Bucheon, South Korea
| |
Collapse
|
13
|
Mitochondria: The Retina's Achilles' Heel in AMD. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1256:237-264. [PMID: 33848005 DOI: 10.1007/978-3-030-66014-7_10] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Strong experimental evidence from studies in human donor retinas and animal models supports the idea that the retinal pathology associated with age-related macular degeneration (AMD) involves mitochondrial dysfunction and consequent altered retinal metabolism. This chapter provides a brief overview of mitochondrial structure and function, summarizes evidence for mitochondrial defects in AMD, and highlights the potential ramifications of these defects on retinal health and function. Discussion of mitochondrial haplogroups and their association with AMD brings to light how mitochondrial genetics can influence disease outcome. As one of the most metabolically active tissues in the human body, there is strong evidence that disruption in key metabolic pathways contributes to AMD pathology. The section on retinal metabolism reviews cell-specific metabolic differences and how the metabolic interdependence of each retinal cell type creates a unique ecosystem that is disrupted in the diseased retina. The final discussion includes strategies for therapeutic interventions that target key mitochondrial pathways as a treatment for AMD.
Collapse
|
14
|
Peng X, Gao L, Liu Y. Cell-based therapies for age-related macular degeneration: cell replacement versus paracrine effects. Neural Regen Res 2021; 16:1214-1215. [PMID: 33269782 PMCID: PMC8224131 DOI: 10.4103/1673-5374.300443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Affiliation(s)
- Xiaoyan Peng
- Department of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Ling Gao
- Department of Ophthalmology, Second Affiliated Hospital of Xiangya Medical School, Central South China University, Changsha, Hunan Province, China
| | - Yongqing Liu
- James Graham Brown Cancer Center; Department of Ophthalmology and Visual Sciences; Birth Defects Center, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
15
|
Villar PS, Vergara C, Bacigalupo J. Energy sources that fuel metabolic processes in protruding finger-like organelles. FEBS J 2020; 288:3799-3812. [PMID: 33142020 DOI: 10.1111/febs.15620] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/16/2020] [Accepted: 11/02/2020] [Indexed: 12/19/2022]
Abstract
Cells possess a variety of organelles with characteristic structure and subcellular localization intimately linked to their specific function. While most are intracellular and found in virtually all eukaryotic cells, there is a small group of organelles of elongated cylindrical shapes in highly specialized cells that protrude into the extracellular space, such as cilia, flagella, and microvilli. The ATP required by intracellular organelles is amply available in the cytosol, largely generated by mitochondria. However, such is not the case for cilia and flagella, whose slender structures cannot accommodate mitochondria. These organelles consume massive amounts of ATP to carry out high energy-demanding functions, such as sensory transduction or motility. ATP from the nearest mitochondria or other reactions within the cell body is severely limited by diffusion and generally insufficient to fuel the entire length of cilia and flagella. These organelles overcome this fuel restriction by local generation of ATP, using mechanisms that vary depending on the nutrients that are available in their particular external environment. Here, we review, with emphasis in mammals, the remarkable adaptations that cilia and flagella use to fuel their metabolic needs. Additionally, we discuss how a decrease in nutrients surrounding olfactory cilia might impair olfaction in COVID-19 patients.
Collapse
Affiliation(s)
- Pablo S Villar
- Department of Biology, University of Maryland, College Park, MD, USA
| | - Cecilia Vergara
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Juan Bacigalupo
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
16
|
Mori S, Kurimoto T, Miki A, Maeda H, Kusuhara S, Nakamura M. Aqp9 Gene Deletion Enhances Retinal Ganglion Cell (RGC) Death and Dysfunction Induced by Optic Nerve Crush: Evidence that Aquaporin 9 Acts as an Astrocyte-to-Neuron Lactate Shuttle in Concert with Monocarboxylate Transporters To Support RGC Function and Survival. Mol Neurobiol 2020; 57:4530-4548. [PMID: 32748371 PMCID: PMC7515957 DOI: 10.1007/s12035-020-02030-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 07/22/2020] [Indexed: 12/27/2022]
Abstract
Aquaporin 9 (AQP9) is an aquaglyceroporin that can transport lactate. Accumulating evidence suggests that astrocyte-to-neuron lactate shuttle (ANLS) plays a critical role in energy metabolism in neurons, including retinal ganglion cells (RGCs). To test the hypothesis that AQP9, in concert with monocarboxylate transporters (MCTs), participates in ANLS to maintain function and survival of RGCs, Aqp9-null mice and wild-type (WT) littermates were subjected to optic nerve crush (ONC) with or without intravitreal injection of an MCT2 inhibitor. RGC density was similar between the Aqp9-null mice and WT mice without ONC, while ONC resulted in significantly more RGC density reduction in the Aqp9-null mice than in the WT mice at day 7. Positive scotopic threshold response (pSTR) amplitude values were similar between the two groups without ONC, but were significantly more reduced in the Aqp9-null mice than in the WT mice 7days after ONC. MCT2 inhibitor injection accelerated RGC death and pSTR amplitude reduction only in the WT mice with ONC. Immunolabeling revealed that both RGCs and astrocytes expressed AQP9, that ONC predominantly reduced astrocytic AQP9 expression, and that MCTs 1, 2, and 4 were co-localized with AQP9 at the ganglion cell layer. These retinal MCTs were also co-immunoprecipitated with AQP9 in the WT mice. ONC decreased the co-immunoprecipitation of MCTs 1 and 4, but did not impact co-immunoprecipitation of MCT2. Retinal glucose transporter 1 expression was increased in Aqp9-null mice. Aqp9 gene deletion reduced and increased the intraretinal L-lactate and D-glucose concentrations, respectively. Results suggest that AQP9 acts as the ANLS to maintain function and survival of RGCs.
Collapse
Affiliation(s)
- Sotaro Mori
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takuji Kurimoto
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Akiko Miki
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hidetaka Maeda
- Maeda Eye Clinic, 1-1-1, Uchihonmachi, Chuo-ku, Osaka, 540-0012, Japan
| | - Sentaro Kusuhara
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Makoto Nakamura
- Division of Ophthalmology, Department of Surgery, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| |
Collapse
|
17
|
Narayan DS, Chidlow G, Wood JPM, Casson RJ. Investigations Into Bioenergetic Neuroprotection of Cone Photoreceptors: Relevance to Retinitis Pigmentosa. Front Neurosci 2019; 13:1234. [PMID: 31803010 PMCID: PMC6872495 DOI: 10.3389/fnins.2019.01234] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022] Open
Abstract
Recent studies suggest cone degeneration in retinitis pigmentosa (RP) may result from intracellular energy depletion. We tested the hypothesis that cones die when depleted of energy by examining the effect of two bioenergetic, nutraceutical agents on cone survival. The study had three specific aims: firstly, we, studied the neuroprotective efficacies of glucose and creatine in an in vitro model of RP. Next, we utilized a well-characterized mouse model of RP to examine whether surviving cones, devoid of their inner segments, continue to express genes vital for glucose, and creatine utilization. Finally, we analyzed the neuroprotective properties of glucose and creatine on cone photoreceptors in a mouse model of RP. Two different bioenergy-based therapies were tested in rd1 mice: repeated local delivery of glucose and systemic creatine. Optomotor responses were tested and cone density was quantified on retinal wholemounts. The results showed that glucose supplementation increased survival of cones in culture subjected to mitochondrial stress or oxidative insult. Despite losing their inner segments, surviving cones in the rd1 retina continued to express the various glycolytic enzymes. Following a single subconjunctival injection, the mean vitreous glucose concentration was significantly elevated at 1 and 8 h, but not at 16 h after injection; however, daily subconjunctival injection of glucose neither enhanced spatial visual performance nor slowed cone cell degeneration in rd1 mice relative to isotonic saline. Creatine dose-dependently increased survival of cones in culture subjected to mitochondrial dysfunction, but not to oxidative stress. Despite the loss of their mitochondrial-rich inner segments, cone somas and axonal terminals in the rd1 retina were strongly positive for both the mitochondrial and cytosolic forms of creatine kinase at each time point examined. Creatine-fed rd1 mice displayed enhanced optomotor responses compared to mice fed normal chow. Moreover, cone density was significantly greater in creatine-treated mice compared to controls. The overall results of this study provide tentative support for the hypothesis that creatine supplementation may delay secondary degeneration of cones in individuals with RP.
Collapse
Affiliation(s)
- Daniel S Narayan
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Glyn Chidlow
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia
| | - John P M Wood
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Robert J Casson
- Ophthalmic Research Laboratories, Discipline of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
18
|
Power M, Das S, Schütze K, Marigo V, Ekström P, Paquet-Durand F. Cellular mechanisms of hereditary photoreceptor degeneration - Focus on cGMP. Prog Retin Eye Res 2019; 74:100772. [PMID: 31374251 DOI: 10.1016/j.preteyeres.2019.07.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 07/25/2019] [Accepted: 07/29/2019] [Indexed: 12/21/2022]
Abstract
The cellular mechanisms underlying hereditary photoreceptor degeneration are still poorly understood, a problem that is exacerbated by the enormous genetic heterogeneity of this disease group. However, the last decade has yielded a wealth of new knowledge on degenerative pathways and their diversity. Notably, a central role of cGMP-signalling has surfaced for photoreceptor cell death triggered by a subset of disease-causing mutations. In this review, we examine key aspects relevant for photoreceptor degeneration of hereditary origin. The topics covered include energy metabolism, epigenetics, protein quality control, as well as cGMP- and Ca2+-signalling, and how the related molecular and metabolic processes may trigger photoreceptor demise. We compare and integrate evidence on different cell death mechanisms that have been associated with photoreceptor degeneration, including apoptosis, necrosis, necroptosis, and PARthanatos. A special focus is then put on the mechanisms of cGMP-dependent cell death and how exceedingly high photoreceptor cGMP levels may cause activation of Ca2+-dependent calpain-type proteases, histone deacetylases and poly-ADP-ribose polymerase. An evaluation of the available literature reveals that a large group of patients suffering from hereditary photoreceptor degeneration carry mutations that are likely to trigger cGMP-dependent cell death, making this pathway a prime target for future therapy development. Finally, an outlook is given into technological and methodological developments that will with time likely contribute to a comprehensive overview over the entire metabolic complexity of photoreceptor cell death. Building on such developments, new imaging technology and novel biomarkers may be used to develop clinical test strategies, that fully consider the genetic heterogeneity of hereditary retinal degenerations, in order to facilitate clinical testing of novel treatment approaches.
Collapse
Affiliation(s)
- Michael Power
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Germany; Centre for Integrative Neurosciences (CIN), University of Tübingen, Germany; Graduate Training Centre of Neuroscience (GTC), University of Tübingen, Germany
| | - Soumyaparna Das
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Germany; Graduate Training Centre of Neuroscience (GTC), University of Tübingen, Germany
| | | | - Valeria Marigo
- Department of Life Sciences, University of Modena and Reggio Emilia, Italy
| | - Per Ekström
- Ophthalmology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sweden
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, University of Tübingen, Germany.
| |
Collapse
|
19
|
Gospe SM, Travis AM, Kolesnikov AV, Klingeborn M, Wang L, Kefalov VJ, Arshavsky VY. Photoreceptors in a mouse model of Leigh syndrome are capable of normal light-evoked signaling. J Biol Chem 2019; 294:12432-12443. [PMID: 31248988 DOI: 10.1074/jbc.ra119.007945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/12/2019] [Indexed: 11/06/2022] Open
Abstract
Mitochondrial dysfunction is an important cause of heritable vision loss. Mutations affecting mitochondrial bioenergetics may lead to isolated vision loss or life-threatening systemic disease, depending on a mutation's severity. Primary optic nerve atrophy resulting from death of retinal ganglion cells is the most prominent ocular manifestation of mitochondrial disease. However, dysfunction of other retinal cell types has also been described, sometimes leading to a loss of photoreceptors and retinal pigment epithelium that manifests clinically as pigmentary retinopathy. A popular mouse model of mitochondrial disease that lacks NADH:ubiquinone oxidoreductase subunit S4 (NDUFS4), a subunit of mitochondrial complex I, phenocopies many traits of the human disease Leigh syndrome, including the development of optic atrophy. It has also been reported that ndufs4 -/- mice display diminished light responses at the level of photoreceptors or bipolar cells. By conducting electroretinography (ERG) recordings in live ndufs4 -/- mice, we now demonstrate that this defect occurs at the level of retinal photoreceptors. We found that this deficit does not arise from retinal developmental anomalies, photoreceptor degeneration, or impaired regeneration of visual pigment. Strikingly, the impairment of ndufs4 -/- photoreceptor function was not observed in ex vivo ERG recordings from isolated retinas, indicating that photoreceptors with complex I deficiency are intrinsically capable of normal signaling. The difference in electrophysiological phenotypes in vivo and ex vivo suggests that the energy deprivation associated with severe mitochondrial impairment in the outer retina renders ndufs4 -/- photoreceptors unable to maintain the homeostatic conditions required to operate at their normal capacity.
Collapse
Affiliation(s)
- Sidney M Gospe
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710.
| | - Amanda M Travis
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710
| | - Alexander V Kolesnikov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Mikael Klingeborn
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710
| | - Luyu Wang
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710
| | - Vladimir J Kefalov
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, Missouri 63110
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University, Durham, North Carolina 27710; Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710
| |
Collapse
|
20
|
Spencer WJ, Ding JD, Lewis TR, Yu C, Phan S, Pearring JN, Kim KY, Thor A, Mathew R, Kalnitsky J, Hao Y, Travis AM, Biswas SK, Lo WK, Besharse JC, Ellisman MH, Saban DR, Burns ME, Arshavsky VY. PRCD is essential for high-fidelity photoreceptor disc formation. Proc Natl Acad Sci U S A 2019; 116:13087-13096. [PMID: 31189593 PMCID: PMC6601265 DOI: 10.1073/pnas.1906421116] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Progressive rod-cone degeneration (PRCD) is a small protein residing in the light-sensitive disc membranes of the photoreceptor outer segment. Until now, the function of PRCD has remained enigmatic despite multiple demonstrations that its mutations cause blindness in humans and dogs. Here, we generated a PRCD knockout mouse and observed a striking defect in disc morphogenesis, whereby newly forming discs do not properly flatten. This leads to the budding of disc-derived vesicles, specifically at the site of disc morphogenesis, which accumulate in the interphotoreceptor matrix. The defect in nascent disc flattening only minimally alters the photoreceptor outer segment architecture beyond the site of new disc formation and does not affect the abundance of outer segment proteins and the photoreceptor's ability to generate responses to light. Interestingly, the retinal pigment epithelium, responsible for normal phagocytosis of shed outer segment material, lacks the capacity to clear the disc-derived vesicles. This deficiency is partially compensated by a unique pattern of microglial migration to the site of disc formation where they actively phagocytize vesicles. However, the microglial response is insufficient to prevent vesicular accumulation and photoreceptors of PRCD knockout mice undergo slow, progressive degeneration. Taken together, these data show that the function of PRCD is to keep evaginating membranes of new discs tightly apposed to each other, which is essential for the high fidelity of photoreceptor disc morphogenesis and photoreceptor survival.
Collapse
Affiliation(s)
- William J Spencer
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710
| | - Jin-Dong Ding
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
| | - Tylor R Lewis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
| | - Chen Yu
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
| | - Sebastien Phan
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Jillian N Pearring
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Andrea Thor
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Rose Mathew
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
| | - Joan Kalnitsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
| | - Ying Hao
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
| | - Amanda M Travis
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710
| | - Sondip K Biswas
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310
| | - Woo-Kuen Lo
- Department of Neurobiology, Morehouse School of Medicine, Atlanta, GA 30310
| | - Joseph C Besharse
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI 53226
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, School of Medicine, University of California San Diego, La Jolla, CA 92093
| | - Daniel R Saban
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710
| | - Marie E Burns
- Department of Cell Biology and Human Anatomy, University of California, Davis, CA 95616
- Department of Ophthalmology, University of California, Davis, CA 95616
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710;
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
21
|
Swarup A, Samuels IS, Bell BA, Han JYS, Du J, Massenzio E, Abel ED, Boesze-Battaglia K, Peachey NS, Philp NJ. Modulating GLUT1 expression in retinal pigment epithelium decreases glucose levels in the retina: impact on photoreceptors and Müller glial cells. Am J Physiol Cell Physiol 2018; 316:C121-C133. [PMID: 30462537 DOI: 10.1152/ajpcell.00410.2018] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The retina is one of the most metabolically active tissues in the body and utilizes glucose to produce energy and intermediates required for daily renewal of photoreceptor cell outer segments. Glucose transporter 1 (GLUT1) facilitates glucose transport across outer blood retinal barrier (BRB) formed by the retinal pigment epithelium (RPE) and the inner BRB formed by the endothelium. We used conditional knockout mice to study the impact of reducing glucose transport across the RPE on photoreceptor and Müller glial cells. Transgenic mice expressing Cre recombinase under control of the Bestrophin1 ( Best1) promoter were bred with Glut1flox/flox mice to generate Tg-Best1-Cre:Glut1flox/flox mice ( RPEΔGlut1). The RPEΔGlut1 mice displayed a mosaic pattern of Cre expression within the RPE that allowed us to analyze mice with ~50% ( RPEΔGlut1m) recombination and mice with >70% ( RPEΔGlut1h) recombination separately. Deletion of GLUT1 from the RPE did not affect its carrier or barrier functions, indicating that the RPE utilizes other substrates to support its metabolic needs thereby sparing glucose for the outer retina. RPEΔGlut1m mice had normal retinal morphology, function, and no cell death; however, where GLUT1 was absent from a span of RPE greater than 100 µm, there was shortening of the photoreceptor cell outer segments. RPEΔGlut1h mice showed outer segment shortening, cell death of photoreceptors, and activation of Müller glial cells. The severe phenotype seen in RPEΔGlut1h mice indicates that glucose transport via the GLUT1 transporter in the RPE is required to meet the anabolic and catabolic requirements of photoreceptors and maintain Müller glial cells in a quiescent state.
Collapse
Affiliation(s)
- Aditi Swarup
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Ivy S Samuels
- Louis Stokes Cleveland VA Medical Center , Cleveland, Ohio.,Cole Eye Institute, Cleveland Clinic , Cleveland, Ohio
| | - Brent A Bell
- Department of Ophthalmology, University of Pennsylvania , Philadelphia, Pennsylvania
| | - John Y S Han
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - Jianhai Du
- Department of Ophthalmology, Department of Biochemistry, West Virginia University Eye Institute , Morgantown, West Virginia
| | - Erik Massenzio
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University , Philadelphia, Pennsylvania
| | - E Dale Abel
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa , Iowa City, Iowa.,Division of Endocrinology and Metabolism, Carver College of Medicine, University of Iowa , Iowa City, Iowa
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, Penn Dental Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Neal S Peachey
- Louis Stokes Cleveland VA Medical Center , Cleveland, Ohio.,Cole Eye Institute, Cleveland Clinic , Cleveland, Ohio.,Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University , Cleveland, Ohio
| | - Nancy J Philp
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University , Philadelphia, Pennsylvania
| |
Collapse
|
22
|
Arslan U, Özmert E, Demirel S, Örnek F, Şermet F. Effects of subtenon-injected autologous platelet-rich plasma on visual functions in eyes with retinitis pigmentosa: preliminary clinical results. Graefes Arch Clin Exp Ophthalmol 2018; 256:893-908. [PMID: 29546474 DOI: 10.1007/s00417-018-3953-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 02/26/2018] [Accepted: 03/02/2018] [Indexed: 12/19/2022] Open
Abstract
PURPOSE One of the main reasons for apoptosis and dormant cell phases in degenerative retinal diseases such as retinitis pigmentosa (RP) is growth factor withdrawal in the cellular microenvironment. Growth factors and neurotrophins can significantly slow down retinal degeneration and cell death in animal models. One possible source of autologous growth factors is platelet-rich plasma. The purpose of this study was to determine if subtenon injections of autologous platelet-rich plasma (aPRP) can have beneficial effects on visual function in RP patients by reactivating dormant photoreceptors. MATERIAL AND METHODS This prospective open-label clinical trial, conducted between September 2016 and February 2017, involved 71 eyes belonging to 48 RP patients with various degrees of narrowed visual field. Forty-nine eyes belonging to 37 patients were injected with aPRP. A comparison group was made up of 11 patients who had symmetrical bilateral narrowed visual field (VF) of both eyes. Among these 11 patients, one eye was injected with aPRP, while the other eye was injected with autologous platelet-poor plasma (aPPP) to serve as a control. The total duration of the study was 9 weeks: the aPRP or aPPP subtenon injections were applied three times, with 3-week intervals between injections, and the patients were followed for three more weeks after the third injection. Visual acuity (VA) tests were conducted on all patients, and VF, microperimetry (MP), and multifocal electroretinography (mfERG) tests were conducted on suitable patients to evaluate the visual function changes before and after the aPRP or aPPP injections. RESULTS The best-corrected visual acuity values in the ETDRS chart improved by 11.6 letters (from 70 to 81.6 letters) in 19 of 48 eyes following aPRP application; this result, however, was not statistically significant (p = 0.056). Following aPRP injections in 48 eyes, the mean deviation of the VF values improved from - 25.3 to - 23.1 dB (p = 0.0001). Results regarding the mfERG P1 amplitudes improved in ring 1 from 24.4 to 38.5 nv/deg2 (p = 0.0001), in ring 2 from 6.7 to 9.3 nv/deg2 (p = 0.0301), and in ring 3 from 3.5 to 4.5 nv/deg2 (p = 0.0329). The mfERG P1 implicit times improved in ring 1 from 40.0 to 34.4 ms (p = 0.01), in ring 2 from 42.5 to 33.2 ms (p = 0.01), and in ring 3 from 42.1 to 37.9 ms (p = 0.04). The mfERG N1 amplitudes improved in ring 1 from 0.18 to 0.25 nv/deg2 (p = 0.011) and in ring 2 from 0.05 to 0.08 nv/deg2 (p = 0.014). The mfERG N1 implicit time also improved in ring 1 from 18.9 to 16.2 ms (p = 0.040) and in ring 2 from 20.9 to 15.5 ms (p = 0.002). No improvement was seen in the 11 control eyes into which aPPP was injected. In the 23 RP patients with macular involvement, the MP average threshold values improved with aPRP injections from 15.0 to 16.4 dB (p = 0.0001). No ocular or systemic adverse events related to the injections or aPRP were observed during the follow-up period. CONCLUSION Preliminary clinical results are encouraging in terms of statistically significant improvements in VF, mfERG values, and MP. The subtenon injection of aPRP seems to be a therapeutic option for treatment and might lead to positive results in the vision of RP patients. Long-term results regarding adverse events are unknown. There have not been any serious adverse events and any ophthalmic or systemic side effects for 1 year follow-up. Further studies with long-term follow-up are needed to determine the duration of efficacy and the frequency of application.
Collapse
Affiliation(s)
- Umut Arslan
- Department of Ophthalmology, Ankara Training and Research Hospital, Ankara, Turkey
| | - Emin Özmert
- Faculty of Medicine, Department of Ophthalmology, Ankara University, Ankara, Turkey
| | - Sibel Demirel
- Faculty of Medicine, Department of Ophthalmology, Ankara University, Ankara, Turkey. .,Cebeci Tıp Fakültesi, Vehbi Koç Göz hastanesi, Göz Hastalıkları Ana Bilimdalı, Mamak caddesi, Dikimevi/Ankara, Dikimevi/Ankara, Turkey.
| | - Firdevs Örnek
- Department of Ophthalmology, Ankara Training and Research Hospital, Ankara, Turkey
| | - Figen Şermet
- Faculty of Medicine, Department of Ophthalmology, Ankara University, Ankara, Turkey
| |
Collapse
|
23
|
Seo S, Datta P. Photoreceptor outer segment as a sink for membrane proteins: hypothesis and implications in retinal ciliopathies. Hum Mol Genet 2017; 26:R75-R82. [PMID: 28453661 DOI: 10.1093/hmg/ddx163] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 04/24/2017] [Indexed: 12/28/2022] Open
Abstract
The photoreceptor outer segment (OS) is a unique modification of the primary cilium, specialized for light perception. Being homologous organelles, the primary cilium and the OS share common building blocks and molecular machinery to construct and maintain them. The OS, however, has several unique structural features that are not seen in primary cilia. Although these unique features of the OS have been well documented, their implications in protein localization have been under-appreciated. In this review, we compare the structural properties of the primary cilium and the OS, and propose a hypothesis that the OS can act as a sink for membrane proteins. We further discuss the implications of this hypothesis in polarized protein localization in photoreceptors and mechanisms of photoreceptor degeneration in retinal ciliopathies.
Collapse
Affiliation(s)
- Seongjin Seo
- Department of Ophthalmology and Visual Sciences, Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Poppy Datta
- Department of Ophthalmology and Visual Sciences, Wynn Institute for Vision Research, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
24
|
Joyal JS, Gantner ML, Smith LEH. Retinal energy demands control vascular supply of the retina in development and disease: The role of neuronal lipid and glucose metabolism. Prog Retin Eye Res 2017; 64:131-156. [PMID: 29175509 DOI: 10.1016/j.preteyeres.2017.11.002] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/11/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Affiliation(s)
- 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, McGill University, Montreal, Qc, Canada.
| | - Marin L Gantner
- The Lowy Medical Research Institute, La Jolla, United States
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston MA 02115, United States.
| |
Collapse
|
25
|
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: 246] [Impact Index Per Article: 30.8] [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
|
26
|
Kaplan HJ, Wang W, Dean DC. Restoration of Cone Photoreceptor Function in Retinitis Pigmentosa. Transl Vis Sci Technol 2017; 6:5. [PMID: 28900578 PMCID: PMC5588910 DOI: 10.1167/tvst.6.5.5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 07/17/2017] [Indexed: 01/12/2023] Open
Affiliation(s)
- Henry J Kaplan
- University of Louisville, Department of Ophthalmology and Visual Sciences, Louisville, KY, USA
| | - Wei Wang
- University of Louisville, Department of Ophthalmology and Visual Sciences, Louisville, KY, USA
| | - Douglas C Dean
- University of Louisville, Department of Ophthalmology and Visual Sciences, Louisville, KY, USA.,University of Louisville, Molecular Targets Program, James Graham Brown Cancer Center, Louisville, KY, USA
| |
Collapse
|
27
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Aerobic Glycolysis Hypothesis Through WNT/Beta-Catenin Pathway in Exudative Age-Related Macular Degeneration. J Mol Neurosci 2017; 62:368-379. [PMID: 28689265 DOI: 10.1007/s12031-017-0947-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022]
Abstract
Exudative age-related macular degeneration (AMD) is characterized by molecular mechanisms responsible for the initiation of choroidal neovascularization (CNV). Inflammatory processes are associated with upregulation of the canonical WNT/beta-catenin pathway in exudative AMD. We focus this review on the link between WNT/beta-catenin pathway activation and neovascular progression in exudative AMD through activation of aerobic glycolysis for production of angiogenic factors. Increased WNT/beta-catenin pathway involves hexokinase 2 (HK2) and pyruvate kinase M2 (PKM2). WNT/beta-catenin pathway stimulates PI3K/Akt pathway and then HIF-1alpha which activates glycolytic enzymes: glucose transporter (Glut), pyruvate dehydrogenase kinase 1 (PDK1), lactate dehydrogenase A (LDH-A), and monocarboxylate lactate transporter (MCT-1). This phenomenon is called aerobic glycolysis or the Warburg effect. Consequently, phosphorylation of PDK-1 inhibits the pyruvate dehydrogenase complex (PDH). Thus, a large part of pyruvate cannot be converted into acetyl-CoA in mitochondria and only a part of acetyl-CoA can enter the tricarboxylic acid cycle. Cytosolic pyruvate is converted into lactate through the action of LDH-A. In exudative AMD, high level of cytosolic lactate is correlated with increase of VEGF expression, the angiogenic factor of CNV. Photoreceptors in retina cells can metabolize glucose through aerobic glycolysis to protect them against oxidative damage, as cancer cells do.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, INSERM U1084, University of Poitiers, Poitiers, France.
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, 11 Boulevard Marie et Pierre Curie, Poitiers, France.
| | | | - Rémy Guillevin
- DACTIM, Laboratoire de Mathématiques et Applications, Université de Poitiers et CHU de Poitiers, UMR CNRS 7348, SP2MI Futuroscope, Chasseneuil-du-Poitou, France
| | - Jean-Noël Vallée
- Laboratoire de Mathématiques et Applications (LMA), UMR CNRS 7348, Université de Poitiers, 11 Boulevard Marie et Pierre Curie, Poitiers, France
- CHU Amiens Picardie, Université Picardie Jules Verne (UPJV), Amiens, France
| |
Collapse
|
28
|
Narayan DS, Chidlow G, Wood JP, Casson RJ. Glucose metabolism in mammalian photoreceptor inner and outer segments. Clin Exp Ophthalmol 2017; 45:730-741. [PMID: 28334493 DOI: 10.1111/ceo.12952] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 02/25/2017] [Accepted: 03/20/2017] [Indexed: 12/22/2022]
Abstract
Photoreceptors are the first-order neurons of the visual pathway, converting light into electrical signals. Rods and cones are the two main types of photoreceptors in the mammalian retina. Rods are specialized for sensitivity at the expense of resolution and are responsible for vision in dimly lit conditions. Cones are responsible for high acuity central vision and colour vision. Many human retinal diseases are characterized by a progressive loss of photoreceptors. Photoreceptors consist of four primary regions: outer segments, inner segments, cell bodies and synaptic terminals. Photoreceptors consume large amounts of energy, and therefore, energy metabolism may be a critical juncture that links photoreceptor function and survival. Cones require more energy than rods, and cone degeneration is the main cause of clinically significant vision loss in retinal diseases. Photoreceptor segments are capable of utilizing various energy substrates, including glucose, to meet their large energy demands. The pathways by which photoreceptor segments meet their energy demands remain incompletely understood. Improvements in the understanding of glucose metabolism in photoreceptor segments may provide insight into the reasons why photoreceptors degenerate due to energy failure. This may, in turn, assist in developing bio-energetic therapies aimed at protecting photoreceptors.
Collapse
Affiliation(s)
- Daniel S Narayan
- Ophthalmic Research Laboratories, Hanson Institute Centre for Neurological Diseases, Adelaide, South Austalia, Australia.,South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Austalia, Australia
| | - Glyn Chidlow
- Ophthalmic Research Laboratories, Hanson Institute Centre for Neurological Diseases, Adelaide, South Austalia, Australia.,South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Austalia, Australia
| | - John Pm Wood
- Ophthalmic Research Laboratories, Hanson Institute Centre for Neurological Diseases, Adelaide, South Austalia, Australia.,South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Austalia, Australia
| | - Robert J Casson
- Ophthalmic Research Laboratories, Hanson Institute Centre for Neurological Diseases, Adelaide, South Austalia, Australia.,South Australian Institute of Ophthalmology, University of Adelaide, Adelaide, South Austalia, Australia
| |
Collapse
|
29
|
Salinas RY, Pearring JN, Ding JD, Spencer WJ, Hao Y, Arshavsky VY. Photoreceptor discs form through peripherin-dependent suppression of ciliary ectosome release. J Cell Biol 2017; 216:1489-1499. [PMID: 28381413 PMCID: PMC5412563 DOI: 10.1083/jcb.201608081] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 12/22/2016] [Accepted: 02/14/2017] [Indexed: 01/27/2023] Open
Abstract
The primary cilium is a highly conserved organelle housing specialized molecules responsible for receiving and processing extracellular signals. A recently discovered property shared across many cilia is the ability to release small vesicles called ectosomes, which are used for exchanging protein and genetic material among cells. In this study, we report a novel role for ciliary ectosomes in building the elaborate photoreceptor outer segment filled with hundreds of tightly packed "disc" membranes. We demonstrate that the photoreceptor cilium has an innate ability to release massive amounts of ectosomes. However, this process is suppressed by the disc-specific protein peripherin, which enables retained ectosomes to be morphed into discs. This new function of peripherin is performed independently from its well-established role in maintaining the high curvature of disc edges, and each function is fulfilled by a separate part of peripherin's molecule. Our findings explain how the outer segment structure evolved from the primary cilium to provide photoreceptor cells with vast membrane surfaces for efficient light capture.
Collapse
Affiliation(s)
- Raquel Y Salinas
- Department of Ophthalmology, Duke University, Durham, NC 27710
- Department of Pharmacology, Duke University, Durham, NC 27710
| | | | - Jin-Dong Ding
- Department of Ophthalmology, Duke University, Durham, NC 27710
| | - William J Spencer
- Department of Ophthalmology, Duke University, Durham, NC 27710
- Department of Pharmacology, Duke University, Durham, NC 27710
| | - Ying Hao
- Department of Ophthalmology, Duke University, Durham, NC 27710
| | - Vadim Y Arshavsky
- Department of Ophthalmology, Duke University, Durham, NC 27710
- Department of Pharmacology, Duke University, Durham, NC 27710
| |
Collapse
|
30
|
Adamus G. Impact of Autoantibodies against Glycolytic Enzymes on Pathogenicity of Autoimmune Retinopathy and Other Autoimmune Disorders. Front Immunol 2017; 8:505. [PMID: 28503176 PMCID: PMC5408022 DOI: 10.3389/fimmu.2017.00505] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/12/2017] [Indexed: 12/20/2022] Open
Abstract
Autoantibodies (AAbs) against glycolytic enzymes: aldolase, α-enolase, glyceraldehyde-3-phosphate dehydrogenase, and pyruvate kinase are prevalent in sera of patients with blinding retinal diseases, such as paraneoplastic [cancer-associated retinopathy (CAR)] and non-paraneoplastic autoimmune retinopathies, as well as in many other autoimmune diseases. CAR is a degenerative disease of the retina characterized by sudden vision loss in patients with cancer and serum anti-retinal AAbs. In this review, we discuss the widespread serum presence of anti-glycolytic enzyme AAbs and their significance in autoimmune diseases. There are multiple mechanisms responsible for antibody generation, including the innate anti-microbial response, anti-tumor response, or autoimmune response against released self-antigens from damaged, inflamed tissue. AAbs against enolase, GADPH, and aldolase exist in a single patient in elevated titers, suggesting their participation in pathogenicity. The lack of restriction of AAbs to one disease may be related to an increased expression of glycolytic enzymes in various metabolically active tissues that triggers an autoimmune response and generation of AAbs with the same specificity in several chronic and autoimmune conditions. In CAR, the importance of serum anti-glycolytic enzyme AAbs had been previously dismissed, but the retina may be without pathological consequence until a failure of the blood–retinal barrier function, which would then allow pathogenic AAbs access to their retinal targets, ultimately leading to damaging effects.
Collapse
Affiliation(s)
- Grazyna Adamus
- School of Medicine, Casey Eye Institute, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
31
|
Garcia-Gonzalo FR, Reiter JF. Open Sesame: How Transition Fibers and the Transition Zone Control Ciliary Composition. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a028134. [PMID: 27770015 DOI: 10.1101/cshperspect.a028134] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cilia are plasma membrane protrusions that act as cellular propellers or antennae. To perform these functions, cilia must maintain a composition distinct from those of the contiguous cytosol and plasma membrane. The specialized composition of the cilium depends on the ciliary gate, the region at the ciliary base separating the cilium from the rest of the cell. The ciliary gate's main structural features are electron dense struts connecting microtubules to the adjacent membrane. These structures include the transition fibers, which connect the distal basal body to the base of the ciliary membrane, and the Y-links, which connect the proximal axoneme and ciliary membrane within the transition zone. Both transition fibers and Y-links form early during ciliogenesis and play key roles in ciliary assembly and trafficking. Accordingly, many human ciliopathies are caused by mutations that perturb ciliary gate function.
Collapse
Affiliation(s)
- Francesc R Garcia-Gonzalo
- Departamento de Bioquímica, Facultad de Medicina, and Instituto de Investigaciones Biomédicas Alberto Sols UAM-CSIC, Universidad Autónoma de Madrid, 28029 Madrid, Spain
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94158
| |
Collapse
|
32
|
Rueda EM, Johnson JE, Giddabasappa A, Swaroop A, Brooks MJ, Sigel I, Chaney SY, Fox DA. The cellular and compartmental profile of mouse retinal glycolysis, tricarboxylic acid cycle, oxidative phosphorylation, and ~P transferring kinases. Mol Vis 2016; 22:847-85. [PMID: 27499608 PMCID: PMC4961465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Accepted: 07/21/2016] [Indexed: 10/26/2022] Open
Abstract
PURPOSE The homeostatic regulation of cellular ATP is achieved by the coordinated activity of ATP utilization, synthesis, and buffering. Glucose is the major substrate for ATP synthesis through glycolysis and oxidative phosphorylation (OXPHOS), whereas intermediary metabolism through the tricarboxylic acid (TCA) cycle utilizes non-glucose-derived monocarboxylates, amino acids, and alpha ketoacids to support mitochondrial ATP and GTP synthesis. Cellular ATP is buffered by specialized equilibrium-driven high-energy phosphate (~P) transferring kinases. Our goals were twofold: 1) to characterize the gene expression, protein expression, and activity of key synthesizing and regulating enzymes of energy metabolism in the whole mouse retina, retinal compartments, and/or cells and 2) to provide an integrative analysis of the results related to function. METHODS mRNA expression data of energy-related genes were extracted from our whole retinal Affymetrix microarray data. Fixed-frozen retinas from adult C57BL/6N mice were used for immunohistochemistry, laser scanning confocal microscopy, and enzymatic histochemistry. The immunoreactivity levels of well-characterized antibodies, for all major retinal cells and their compartments, were obtained using our established semiquantitative confocal and imaging techniques. Quantitative cytochrome oxidase (COX) and lactate dehydrogenase (LDH) activity was determined histochemically. RESULTS The Affymetrix data revealed varied gene expression patterns of the ATP synthesizing and regulating enzymes found in the muscle, liver, and brain. Confocal studies showed differential cellular and compartmental distribution of isozymes involved in glucose, glutamate, glutamine, lactate, and creatine metabolism. The pattern and intensity of the antibodies and of the COX and LDH activity showed the high capacity of photoreceptors for aerobic glycolysis and OXPHOS. Competition assays with pyruvate revealed that LDH-5 was localized in the photoreceptor inner segments. The combined results indicate that glycolysis is regulated by the compartmental expression of hexokinase 2, pyruvate kinase M1, and pyruvate kinase M2 in photoreceptors, whereas the inner retinal neurons exhibit a lower capacity for glycolysis and aerobic glycolysis. Expression of nucleoside diphosphate kinase, mitochondria-associated adenylate kinase, and several mitochondria-associated creatine kinase isozymes was highest in the outer retina, whereas expression of cytosolic adenylate kinase and brain creatine kinase was higher in the cones, horizontal cells, and amacrine cells indicating the diversity of ATP-buffering strategies among retinal neurons. Based on the antibody intensities and the COX and LDH activity, Müller glial cells (MGCs) had the lowest capacity for glycolysis, aerobic glycolysis, and OXPHOS. However, they showed high expression of glutamate dehydrogenase, alpha-ketoglutarate dehydrogenase, succinate thiokinase, GABA transaminase, and ~P transferring kinases. This suggests that MGCs utilize TCA cycle anaplerosis and cataplerosis to generate GTP and ~P transferring kinases to produce ATP that supports MGC energy requirements. CONCLUSIONS Our comprehensive and integrated results reveal that the adult mouse retina expresses numerous isoforms of ATP synthesizing, regulating, and buffering genes; expresses differential cellular and compartmental levels of glycolytic, OXPHOS, TCA cycle, and ~P transferring kinase proteins; and exhibits differential layer-by-layer LDH and COX activity. New insights into cell-specific and compartmental ATP and GTP production, as well as utilization and buffering strategies and their relationship with known retinal and cellular functions, are discussed. Developing therapeutic strategies for neuroprotection and treating retinal deficits and degeneration in a cell-specific manner will require such knowledge. This work provides a platform for future research directed at identifying the molecular targets and proteins that regulate these processes.
Collapse
Affiliation(s)
- Elda M. Rueda
- College of Optometry, University of Houston, Houston TX
| | - Jerry E. Johnson
- Department of Natural Sciences, University of Houston-Downtown, Houston TX
- Department of Biology and Biochemistry, University of Houston, Houston TX
| | - Anand Giddabasappa
- Department of Biology and Biochemistry, University of Houston, Houston TX
| | | | | | - Irena Sigel
- College of Optometry, University of Houston, Houston TX
| | - Shawnta Y. Chaney
- Department of Biology and Biochemistry, University of Houston, Houston TX
| | - Donald A. Fox
- College of Optometry, University of Houston, Houston TX
- Department of Biology and Biochemistry, University of Houston, Houston TX
- Department of Pharmacology and Pharmaceutical Sciences, University of Houston, Houston TX
| |
Collapse
|
33
|
Aït-Ali N, Fridlich R, Millet-Puel G, Clérin E, Delalande F, Jaillard C, Blond F, Perrocheau L, Reichman S, Byrne LC, Olivier-Bandini A, Bellalou J, Moyse E, Bouillaud F, Nicol X, Dalkara D, van Dorsselaer A, Sahel JA, Léveillard T. Rod-derived cone viability factor promotes cone survival by stimulating aerobic glycolysis. Cell 2016; 161:817-32. [PMID: 25957687 DOI: 10.1016/j.cell.2015.03.023] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/21/2015] [Accepted: 03/05/2015] [Indexed: 12/31/2022]
Abstract
Rod-derived cone viability factor (RdCVF) is an inactive thioredoxin secreted by rod photoreceptors that protects cones from degeneration. Because the secondary loss of cones in retinitis pigmentosa (RP) leads to blindness, the administration of RdCVF is a promising therapy for this untreatable neurodegenerative disease. Here, we investigated the mechanism underlying the protective role of RdCVF in RP. We show that RdCVF acts through binding to Basigin-1 (BSG1), a transmembrane protein expressed specifically by photoreceptors. BSG1 binds to the glucose transporter GLUT1, resulting in increased glucose entry into cones. Increased glucose promotes cone survival by stimulation of aerobic glycolysis. Moreover, a missense mutation of RdCVF results in its inability to bind to BSG1, stimulate glucose uptake, and prevent secondary cone death in a model of RP. Our data uncover an entirely novel mechanism of neuroprotection through the stimulation of glucose metabolism.
Collapse
Affiliation(s)
- Najate Aït-Ali
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Ram Fridlich
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Géraldine Millet-Puel
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Emmanuelle Clérin
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - François Delalande
- BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, Université de Strasbourg, 25 rue Becquerel, 67087 Strasbourg, France; IPHC, CNRS, UMR7178, 67087 Strasbourg, France
| | - Céline Jaillard
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Frédéric Blond
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Ludivine Perrocheau
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Sacha Reichman
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Leah C Byrne
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | | | - Jacques Bellalou
- Institut Pasteur, Platform 5 Production of Recombinant Proteins and Antibodies, 75724 Paris Cedex 15, France
| | - Emmanuel Moyse
- Unité de Physiologie de la Reproduction et des Comportements (PRC), UMR-85 INRA, Centre INRA de Tours, Université François Rabelais de Tours, 37380 Nouzilly, France
| | - Frédéric Bouillaud
- Inserm, U1016, Institut Cochin, 75014 Paris, France; Cnrs, UMR8104, 75014 Paris, France; Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Xavier Nicol
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Deniz Dalkara
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Alain van Dorsselaer
- BioOrganic Mass Spectrometry Laboratory (LSMBO), IPHC, Université de Strasbourg, 25 rue Becquerel, 67087 Strasbourg, France; IPHC, CNRS, UMR7178, 67087 Strasbourg, France
| | - José-Alain Sahel
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France
| | - Thierry Léveillard
- INSERM, U968, 75012 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR_S 968, Institut de la Vision, 75012 Paris, France; CNRS, UMR_7210, 75012 Paris, France.
| |
Collapse
|
34
|
Joyal JS, Sun Y, Gantner ML, Shao Z, Evans LP, Saba N, Fredrick T, Burnim S, Kim JS, Patel G, Juan AM, Hurst CG, Hatton CJ, Cui Z, Pierce KA, Bherer P, Aguilar E, Powner MB, Vevis K, Boisvert M, Fu Z, Levy E, Fruttiger M, Packard A, Rezende FA, Maranda B, Sapieha P, Chen J, Friedlander M, Clish CB, Smith LE. Retinal lipid and glucose metabolism dictates angiogenesis through the lipid sensor Ffar1. Nat Med 2016; 22:439-45. [PMID: 26974308 PMCID: PMC4823176 DOI: 10.1038/nm.4059] [Citation(s) in RCA: 185] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 02/05/2016] [Indexed: 02/06/2023]
Abstract
Tissues with high metabolic rates often use lipids, as well as glucose, for energy, conferring a survival advantage during feast and famine. Current dogma suggests that high-energy-consuming photoreceptors depend on glucose. Here we show that the retina also uses fatty acid β-oxidation for energy. Moreover, we identify a lipid sensor, free fatty acid receptor 1 (Ffar1), that curbs glucose uptake when fatty acids are available. Very-low-density lipoprotein receptor (Vldlr), which is present in photoreceptors and is expressed in other tissues with a high metabolic rate, facilitates the uptake of triglyceride-derived fatty acid. In the retinas of Vldlr(-/-) mice with low fatty acid uptake but high circulating lipid levels, we found that Ffar1 suppresses expression of the glucose transporter Glut1. Impaired glucose entry into photoreceptors results in a dual (lipid and glucose) fuel shortage and a reduction in the levels of the Krebs cycle intermediate α-ketoglutarate (α-KG). Low α-KG levels promotes stabilization of hypoxia-induced factor 1a (Hif1a) and secretion of vascular endothelial growth factor A (Vegfa) by starved Vldlr(-/-) photoreceptors, leading to neovascularization. The aberrant vessels in the Vldlr(-/-) retinas, which invade normally avascular photoreceptors, are reminiscent of the vascular defects in retinal angiomatous proliferation, a subset of neovascular age-related macular degeneration (AMD), which is associated with high vitreous VEGFA levels in humans. Dysregulated lipid and glucose photoreceptor energy metabolism may therefore be a driving force in macular telangiectasia, neovascular AMD and other retinal diseases.
Collapse
Affiliation(s)
- Jean-Sébastien Joyal
- Department of Pediatrics, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Ca
- Department of Pharmacology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Ca
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Ca
| | - Ye Sun
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Ca
| | | | - Zhuo Shao
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Lucy P. Evans
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Nicholas Saba
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Thomas Fredrick
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Samuel Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Jin Sung Kim
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Ca
| | - Gauri Patel
- Department of Pharmacology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Ca
| | - Aimee M. Juan
- Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Ca
| | - Christian G. Hurst
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Colman J. Hatton
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Zhenghao Cui
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Kerry A. Pierce
- Metabolite Profiling Platform, The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Patrick Bherer
- Department of Genetics, Université de Sherbrooke, Sherbrooke, Qc, Ca
| | - Edith Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Kristis Vevis
- Institute of Ophthalmology, University College London, London, UK
| | - Michel Boisvert
- Department of Nutrition, CHU Sainte-Justine Research Center, Université de Montreal, Montreal, Qc, Ca
| | - Zhongjie Fu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Emile Levy
- Department of Nutrition, CHU Sainte-Justine Research Center, Université de Montreal, Montreal, Qc, Ca
| | - Marcus Fruttiger
- Institute of Ophthalmology, University College London, London, UK
| | - Alan Packard
- Radiology, Harvard Medical School, Boston Children's Hospital, MA. USA
| | - Flavio A. Rezende
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Qc, Ca
| | - Bruno Maranda
- Department of Genetics, Université de Sherbrooke, Sherbrooke, Qc, Ca
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Université de Montréal, Montreal, Qc, Ca
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| | - Martin Friedlander
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Clary B. Clish
- Metabolite Profiling Platform, The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Lois E.H. Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, MA, USA
| |
Collapse
|
35
|
Wang W, Lee SJ, Scott PA, Lu X, Emery D, Liu Y, Ezashi T, Roberts MR, Ross JW, Kaplan HJ, Dean DC. Two-Step Reactivation of Dormant Cones in Retinitis Pigmentosa. Cell Rep 2016; 15:372-85. [PMID: 27050517 DOI: 10.1016/j.celrep.2016.03.022] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/02/2016] [Accepted: 03/04/2016] [Indexed: 01/01/2023] Open
Abstract
Most retinitis pigmentosa (RP) mutations arise in rod photoreceptor genes, leading to diminished peripheral and nighttime vision. Using a pig model of autosomal-dominant RP, we show glucose becomes sequestered in the retinal pigment epithelium (RPE) and, thus, is not transported to photoreceptors. The resulting starvation for glucose metabolites impairs synthesis of cone visual pigment-rich outer segments (OSs), and then their mitochondrial-rich inner segments dissociate. Loss of these functional structures diminishes cone-dependent high-resolution central vision, which is utilized for most daily tasks. By transplanting wild-type rods, to restore glucose transport, or directly replacing glucose in the subretinal space, to bypass its retention in the RPE, we can regenerate cone functional structures, reactivating the dormant cells. Beyond providing metabolic building blocks for cone functional structures, we show glucose induces thioredoxin-interacting protein (Txnip) to regulate Akt signaling, thereby shunting metabolites toward aerobic glucose metabolism and regenerating cone OS synthesis.
Collapse
Affiliation(s)
- Wei Wang
- Department of Ophthalmology & Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Sang Joon Lee
- Department of Ophthalmology & Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA; Department of Ophthalmology, College of Medicine, Kosin University, Busan, South Korea
| | - Patrick A Scott
- Department of Ophthalmology & Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Xiaoqin Lu
- Department of Ophthalmology & Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Douglas Emery
- Department of Ophthalmology & Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Yongqin Liu
- Department of Ophthalmology & Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA; Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Toshihiko Ezashi
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Michael R Roberts
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Jason W Ross
- Department of Animal Sciences, Iowa State University, Ames, IA 50011, USA
| | - Henry J Kaplan
- Department of Ophthalmology & Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA
| | - Douglas C Dean
- Department of Ophthalmology & Visual Sciences, University of Louisville Health Sciences Center, Louisville, KY 40202, USA; Molecular Targets Program, James Graham Brown Cancer Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA; Birth Defects Center, University of Louisville Health Sciences Center, Louisville, KY 40202, USA.
| |
Collapse
|
36
|
Hurley JB, Lindsay KJ, Du J. Glucose, lactate, and shuttling of metabolites in vertebrate retinas. J Neurosci Res 2015; 93:1079-92. [PMID: 25801286 PMCID: PMC4720126 DOI: 10.1002/jnr.23583] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 02/11/2015] [Accepted: 02/12/2015] [Indexed: 02/06/2023]
Abstract
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.
Collapse
Affiliation(s)
- James B. Hurley
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington
| | - Kenneth J. Lindsay
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington
| | - Jianhai Du
- Departments of Biochemistry and Ophthalmology, University of Washington, Seattle, Washington
| |
Collapse
|
37
|
Pearring JN, Lieu EC, Winter JR, Baker SA, Arshavsky VY. R9AP targeting to rod outer segments is independent of rhodopsin and is guided by the SNARE homology domain. Mol Biol Cell 2014; 25:2644-9. [PMID: 25009288 PMCID: PMC4148253 DOI: 10.1091/mbc.e14-02-0747] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
R9AP, the membrane anchor for transducin's GTPase-activating complex, contains targeting information within its SNARE homology domain that is both necessary and sufficient for R9AP delivery to photoreceptor outer segments. R9AP's targeting is independent of rhodopsin, the most abundant protein residing in the outer segment organelle. In vertebrate photoreceptor cells, rapid recovery from light excitation is dependent on the RGS9⋅Gβ5 GTPase-activating complex located in the light-sensitive outer segment organelle. RGS9⋅Gβ5 is tethered to the outer segment membranes by its membrane anchor, R9AP. Recent studies indicated that RGS9⋅Gβ5 possesses targeting information that excludes it from the outer segment and that this information is overridden by association with R9AP, which allows outer segment targeting of the entire complex. It was also proposed that R9AP itself does not contain specific targeting information and instead is delivered to the outer segment in the same post-Golgi vesicles as rhodopsin, because they are the most abundant transport vesicles in photoreceptor cells. In this study, we revisited this concept by analyzing R9AP targeting in rods of wild-type and rhodopsin-knockout mice. We found that the R9AP targeting mechanism does not require the presence of rhodopsin and further demonstrated that R9AP is actively targeted in rods by its SNARE homology domain.
Collapse
Affiliation(s)
- Jillian N Pearring
- Albert Eye Research Institute, Duke Eye Center, Duke University, Durham, NC 27710
| | - Eric C Lieu
- Albert Eye Research Institute, Duke Eye Center, Duke University, Durham, NC 27710
| | - Joan R Winter
- Albert Eye Research Institute, Duke Eye Center, Duke University, Durham, NC 27710
| | - Sheila A Baker
- Department of Biochemistry and Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA 52242
| | - Vadim Y Arshavsky
- Albert Eye Research Institute, Duke Eye Center, Duke University, Durham, NC 27710
| |
Collapse
|
38
|
Adler L, Chen C, Koutalos Y. Mitochondria contribute to NADPH generation in mouse rod photoreceptors. J Biol Chem 2013; 289:1519-28. [PMID: 24297174 DOI: 10.1074/jbc.m113.511295] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NADPH is the primary source of reducing equivalents in the cytosol. Its major source is considered to be the pentose phosphate pathway, but cytosolic NADP(+)-dependent dehydrogenases using intermediates of mitochondrial pathways for substrates have been known to contribute. Photoreceptors, a nonproliferating cell type, provide a unique model for measuring the functional utilization of NADPH at the single cell level. In these cells, NADPH availability can be monitored from the reduction of the all-trans-retinal generated by light to all-trans-retinol using single cell fluorescence imaging. We have used mouse rod photoreceptors to investigate the generation of NADPH by different metabolic pathways. In the absence of extracellular metabolic substrates, NADPH generation was severely compromised. Extracellular glutamine supported NADPH generation to levels comparable to those of glucose, but pyruvate and lactate were relatively ineffective. At low extracellular substrate concentrations, partial inhibition of ATP synthesis lowered, whereas suppression of ATP consumption augmented NADPH availability. Blocking pyruvate transport into mitochondria decreased NADPH availability, and addition of glutamine restored it. Our findings demonstrate that in a nonproliferating cell type, mitochondria-linked pathways can generate substantial amounts of NADPH and do so even when the pentose phosphate pathway is operational. Competing demands for ATP and NADPH at low metabolic substrate concentrations indicate a vulnerability to nutrient shortages. By supporting substantial NADPH generation, mitochondria provide alternative metabolic pathways that may support cell function and maintain viability under transient nutrient shortages. Such pathways may play an important role in protecting against retinal degeneration.
Collapse
|
39
|
Pearring JN, Salinas RY, Baker SA, Arshavsky VY. Protein sorting, targeting and trafficking in photoreceptor cells. Prog Retin Eye Res 2013; 36:24-51. [PMID: 23562855 DOI: 10.1016/j.preteyeres.2013.03.002] [Citation(s) in RCA: 137] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 03/22/2013] [Accepted: 03/26/2013] [Indexed: 01/24/2023]
Abstract
Vision is the most fundamental of our senses initiated when photons are absorbed by the rod and cone photoreceptor neurons of the retina. At the distal end of each photoreceptor resides a light-sensing organelle, called the outer segment, which is a modified primary cilium highly enriched with proteins involved in visual signal transduction. At the proximal end, each photoreceptor has a synaptic terminal, which connects this cell to the downstream neurons for further processing of the visual information. Understanding the mechanisms involved in creating and maintaining functional compartmentalization of photoreceptor cells remains among the most fascinating topics in ocular cell biology. This review will discuss how photoreceptor compartmentalization is supported by protein sorting, targeting and trafficking, with an emphasis on the best-studied cases of outer segment-resident proteins.
Collapse
Affiliation(s)
- Jillian N Pearring
- Department of Ophthalmology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
40
|
Salinas RY, Baker SA, Gospe SM, Arshavsky VY. A single valine residue plays an essential role in peripherin/rds targeting to photoreceptor outer segments. PLoS One 2013; 8:e54292. [PMID: 23342122 PMCID: PMC3544770 DOI: 10.1371/journal.pone.0054292] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 12/10/2012] [Indexed: 12/28/2022] Open
Abstract
Peripherin/retinal degeneration slow (rds) is an integral membrane protein specifically localized to the light-sensing organelle of the photoreceptor cell, the outer segment. Within the outer segment, peripherin is found at the edges of photoreceptor discs, where it plays a critical role in disc morphogenesis and maintenance. Peripherin loss or mutations are often associated with severe forms of visual impairments. Like all other resident outer segment proteins, peripherin is synthesized in the photoreceptor cell body and subsequently transported to the outer segment. In an effort to further examine peripherin’s delivery to outer segments, we undertook a careful examination of its targeting sequence. Using a fluorescently labeled reporter expressed in the rods of transgenic tadpoles, we narrowed peripherin’s targeting sequence to ten amino acids within its C-terminal tail. This small stretch of amino acid residues is both necessary and sufficient for outer segment targeting. We also conducted alanine scanning of all residues within this sequence and found that only a single residue, valine at position 332, is essential for outer segment targeting. This valine is conserved in all species and its mutation is sufficient to completely abrogate the targeting of full-length peripherin in mouse rods.
Collapse
Affiliation(s)
- Raquel Y. Salinas
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Sheila A. Baker
- Department of Biochemistry, University of Iowa, Iowa City, Iowa, United States of America
| | - Sidney M. Gospe
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Vadim Y. Arshavsky
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Ophthalmology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
41
|
Abstract
Photoreceptors are exquisitely adapted to transform light stimuli into electrical signals that modulate neurotransmitter release. These cells are organized into several compartments including the unique outer segment (OS). Its whole function is to absorb light and transduce this signal into a change of membrane potential. Another compartment is the inner segment where much of metabolism and regulation of membrane potential takes place and that connects the OS and synapse. The synapse is the compartment where changes in membrane potentials are relayed to other neurons in the retina via release of neurotransmitter. The composition of the plasma membrane surrounding these compartments varies to accommodate their specific functions. In this chapter, we discuss the organization of the plasma membrane emphasizing the protein composition of each region as it relates to visual signaling. We also point out examples where mutations in these proteins cause visual impairment.
Collapse
Affiliation(s)
- Sheila A Baker
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA.
| | | |
Collapse
|
42
|
Garcia-Gonzalo FR, Reiter JF. Scoring a backstage pass: mechanisms of ciliogenesis and ciliary access. ACTA ACUST UNITED AC 2012; 197:697-709. [PMID: 22689651 PMCID: PMC3373398 DOI: 10.1083/jcb.201111146] [Citation(s) in RCA: 177] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cilia are conserved, microtubule-based cell surface projections that emanate from basal bodies, membrane-docked centrioles. The beating of motile cilia and flagella enables cells to swim and epithelia to displace fluids. In contrast, most primary cilia do not beat but instead detect environmental or intercellular stimuli. Inborn defects in both kinds of cilia cause human ciliopathies, diseases with diverse manifestations such as heterotaxia and kidney cysts. These diseases are caused by defects in ciliogenesis or ciliary function. The signaling functions of cilia require regulation of ciliary composition, which depends on the control of protein traffic into and out of cilia.
Collapse
Affiliation(s)
- Francesc R Garcia-Gonzalo
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
43
|
Adijanto J, Philp NJ. The SLC16A family of monocarboxylate transporters (MCTs)--physiology and function in cellular metabolism, pH homeostasis, and fluid transport. CURRENT TOPICS IN MEMBRANES 2012. [PMID: 23177990 DOI: 10.1016/b978-0-12-394316-3.00009-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The SLC16A family of monocarboxylate transporters (MCTs) is composed of 14 members. MCT1 through MCT4 (MCTs 1-4) are H(+)-coupled monocarboxylate transporters, MCT8 and MCT10 transport thyroid hormone and aromatic amino acids, while the substrate specificity and function of other MCTs have yet to be determined. The focus of this review is on MCTs 1-4 because their role in lactate transport is intrinsically linked to cellular metabolism in various biological systems, including skeletal muscle, brain, retina, and testis. Although MCTs 1-4 all transport lactate, they differ in their transport kinetics and vary in tissue and subcellular distribution, where they facilitate "lactate-shuttling" between glycolytic and oxidative cells within tissues and across blood-tissue barriers. However, the role of MCTs 1-4 is not confined to cellular metabolism. By interacting with bicarbonate transport proteins and carbonic anhydrases, MCTs participate in the regulation of pH homeostasis and fluid transport in renal proximal tubule and corneal endothelium, respectively. Here, we provide a comprehensive review of MCTs 1-4, linking their cellular distribution to their functions in various parts of the human body, so that we can better understand the physiological roles of MCTs at the systemic level.
Collapse
Affiliation(s)
- Jeffrey Adijanto
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| | | |
Collapse
|
44
|
Chertov AO, Holzhausen L, Kuok IT, Couron D, Parker E, Linton JD, Sadilek M, Sweet IR, Hurley JB. Roles of glucose in photoreceptor survival. J Biol Chem 2011; 286:34700-11. [PMID: 21840997 PMCID: PMC3186402 DOI: 10.1074/jbc.m111.279752] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Revised: 08/04/2011] [Indexed: 11/06/2022] Open
Abstract
Vertebrate photoreceptor neurons have a high demand for metabolic energy, and their viability is very sensitive to genetic and environmental perturbations. We investigated the relationship between energy metabolism and cell death by evaluating the metabolic effects of glucose deprivation on mouse photoreceptors. Oxygen consumption, lactate production, ATP, NADH/NAD(+), TCA cycle intermediates, morphological changes, autophagy, and viability were evaluated. We compared retinas incubated with glucose to retinas deprived of glucose or retinas treated with a mixture of mitochondrion-specific fuels. Rapid and slow phases of cell death were identified. The rapid phase is linked to reduced mitochondrial activity, and the slower phase reflects a need for substrates for cell maintenance and repair.
Collapse
Affiliation(s)
| | | | | | - Drew Couron
- Medicine, Diabetes, Obesity Center of Excellence
| | | | | | - Martin Sadilek
- Chemistry, University of Washington, Seattle, Washington 98195
| | - Ian R. Sweet
- Medicine, Diabetes, Obesity Center of Excellence
| | | |
Collapse
|
45
|
Reidel B, Thompson JW, Farsiu S, Moseley MA, Skiba NP, Arshavsky VY. Proteomic profiling of a layered tissue reveals unique glycolytic specializations of photoreceptor cells. Mol Cell Proteomics 2010; 10:M110.002469. [PMID: 21173383 DOI: 10.1074/mcp.m110.002469] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The retina is a highly ordered tissue whose outermost layers are formed by subcellular compartments of photoreceptors generating light-evoked electrical responses. We studied protein distributions among individual photoreceptor compartments by separating the entire photoreceptor layer of a flat-mounted frozen retina into a series of thin tangential cryosections and analyzing protein compositions of each section by label-free quantitative mass spectrometry. Based on 5038 confidently identified peptides assigned to 896 protein database entries, we generated a quantitative proteomic database (a "map") correlating the distribution profiles of identified proteins with the profiles of marker proteins representing individual compartments of photoreceptors and adjacent cells. We evaluated the applicability of several common peptide-to-protein quantification algorithms in the context of our database and found that the highest reliability was obtained by summing the intensities of all peptides representing a given protein, using at least the 5-6 most intense peptides when applicable. We used this proteome map to investigate the distribution of glycolytic enzymes, critical in fulfilling the extremely high metabolic demands of photoreceptor cells, and obtained two major findings. First, unlike the majority of neurons rich in hexokinase I, but similar to other highly metabolically active cells, photoreceptors express hexokinase II. Hexokinase II has a very high catalytic activity when associated with mitochondria, and indeed we found it colocalized with mitochondria in photoreceptors. Second, photoreceptors contain very little triosephosphate isomerase, an enzyme converting dihydroxyacetone phosphate into glyceraldehyde-3-phosphate. This may serve as a functional adaptation because dihydroxyacetone phosphate is a major precursor in phospholipid biosynthesis, a process particularly active in photoreceptors because of the constant renewal of their light-sensitive membrane disc stacks. Overall, our approach for proteomic profiling of very small tissue amounts at a resolution of a few microns, combining cryosectioning and liquid chromatography-tandem MS, can be applied for quantitative investigation of proteomes where spatial resolution is paramount.
Collapse
Affiliation(s)
- Boris Reidel
- Albert Eye Research Institute, 2310 Erwin Road, Durham NC 27710, USA
| | | | | | | | | | | |
Collapse
|