1
|
Yamagishi H, Kirai N, Morita A, Kashihara T, Nakahara T. Role of monocarboxylate transporters in AMPK-mediated protection against excitotoxic injury in the rat retina. Eur J Pharmacol 2024; 970:176510. [PMID: 38493917 DOI: 10.1016/j.ejphar.2024.176510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Activation of adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway protects against N-methyl-D-aspartic acid (NMDA)-induced excitotoxic retinal injury. AMPK activation enhances fatty acid metabolism and ketone body synthesis. Ketone bodies are transported into neurons by monocarboxylate transporters (MCTs) and exert neuroprotective effects. In this study, we examined the distribution and expression levels of MCT1 and MCT2 in the retina and analyzed the effects of pharmacological inhibition of MCTs on the protective effects of metformin and 5-aminoimidazole-4-carboxamide (AICAR), activators of AMPK, against NMDA-induced retinal injury in rats. MCT1 was expressed in the blood vessels, processes of astrocytes and Müller cells, and inner segments of photoreceptors in the rat retina, whereas MCT2 was expressed in neuronal cells in the ganglion cell layer (GCL) and in astrocyte processes. The expression levels of MCT2, but not MCT1, decreased one day after intravitreal injection of NMDA (200 nmol). Intravitreal injection of NMDA decreased the number of cells in the GCL compared to the vehicle seven days after injection. Simultaneous injection of metformin (20 nmol) or AICAR (50 nmol) with NMDA attenuated NMDA-induced cell loss in the GCL, and these protective effects were attenuated by AR-C155858 (1 pmol), an inhibitor of MCTs. AR-C155858 alone had no significant effect on the retinal structure. These results suggest that AMPK-activating compounds protect against NMDA-induced excitotoxic retinal injury via mechanisms involving MCTs in rats. NMDA-induced neurotoxicity may be associated with retinal neurodegenerative changes in glaucoma and diabetic retinopathy. Therefore, AMPK-activating compounds may be effective in managing these retinal diseases.
Collapse
Affiliation(s)
- Honoka Yamagishi
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Nozomu Kirai
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Akane Morita
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Toshihide Kashihara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan.
| |
Collapse
|
2
|
Calbiague García V, Chen Y, Cádiz B, Wang L, Paquet-Durand F, Schmachtenberg O. Imaging of lactate metabolism in retinal Müller cells with a FRET nanosensor. Exp Eye Res 2023; 226:109352. [PMID: 36528083 DOI: 10.1016/j.exer.2022.109352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/30/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Müller cells, the glial cells of the retina, provide metabolic support for photoreceptors and inner retinal neurons, and have been proposed as source of the significant lactate production of this tissue. To better understand the role of lactate in retinal metabolism, we expressed a lactate and a glucose nanosensor in organotypic mouse retinal explants cultured for 14 days, and used FRET imaging in acute vibratome sections of the explants to study metabolite flux in real time. Pharmacological manipulation with specific monocarboxylate transporter (MCT) inhibitors and immunohistochemistry revealed the functional expression of MCT1, MCT2 and MCT4 in Müller cells of retinal explants. The introduction of FRET nanosensors to measure key metabolites at the cellular level may contribute to a better understanding of heretofore poorly understood issues in retinal metabolism.
Collapse
Affiliation(s)
- Víctor Calbiague García
- PhD Program in Neuroscience, Universidad de Valparaíso, Valparaíso, Chile; CINV, Instituto de Biología, Universidad de Valparaíso, Chile
| | - Yiyi Chen
- Institute for Ophthalmic Research, University of Tübingen, Germany
| | - Bárbara Cádiz
- CINV, Instituto de Biología, Universidad de Valparaíso, Chile
| | - Lan Wang
- Institute for Ophthalmic Research, University of Tübingen, Germany
| | | | | |
Collapse
|
3
|
Torres-Vergara P, Rivera R, Escudero C, Penny J. Maternal and Fetal Expression of ATP-Binding Cassette and Solute Carrier Transporters Involved in the Brain Disposition of Drugs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:149-177. [PMID: 37466773 DOI: 10.1007/978-3-031-32554-0_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Evidence from preclinical and clinical studies demonstrate that pregnancy is a physiological state capable of modifying drug disposition. Factors including increased hepatic metabolism and renal excretion are responsible for impacting disposition, and the role of membrane transporters expressed in biological barriers, including the placental- and blood-brain barriers, has received considerable attention. In this regard, the brain disposition of drugs in the mother and fetus has been the subject of studies attempting to characterize the mechanisms by which pregnancy could alter the expression of ATP-binding cassette (ABC) and solute carrier (SLC) transporters. This chapter will summarize findings of the influence of pregnancy on the maternal and fetal expression of ABC and SLC transporters in the brain and the consequences of such changes on the disposition of therapeutic drugs.
Collapse
Affiliation(s)
- Pablo Torres-Vergara
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile.
- Grupo de Investigación Vascular (GRIVAS), Universidad del Bio-Bio, Chillán, Chile.
| | - Robin Rivera
- Departamento de Farmacia, Facultad de Farmacia, Universidad de Concepción, Concepción, Chile
| | - Carlos Escudero
- Grupo de Investigación Vascular (GRIVAS), Universidad del Bio-Bio, Chillán, Chile
- Laboratorio de Fisiología Vascular, Facultad de Ciencias Básicas, Universidad del Bio Bio, Chillán, Chile
| | - Jeffrey Penny
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Health and Medicine, The University of Manchester, Manchester, UK
| |
Collapse
|
4
|
Chiang PP, Kuo SP, Newman EA. Cellular mechanisms mediating activity-dependent extracellular space shrinkage in the retina. Glia 2022; 70:1927-1937. [PMID: 35678626 PMCID: PMC9378592 DOI: 10.1002/glia.24228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022]
Abstract
Volume transmission plays an essential role in CNS function, with neurotransmitters released from synapses diffusing through the extracellular space (ECS) to distant sites. Changes in the ECS volume fraction (α) will influence the diffusion and the concentration of transmitters within the ECS. We have recently shown that neuronal activity evoked by physiological photic stimuli results in rapid decreases in ECS α as large as 10% in the retina. We now characterize the cellular mechanisms responsible for this ECS shrinkage. We find that block of inwardly rectifying K+ channels with Ba2+, inhibition of the Na+/K+/2Cl− cotransporter with bumetanide, or block of AQP4 water channels with TGN‐020 do not diminish the light‐evoked ECS decrease. Inhibition of the Na+/HCO3− cotransporter by removing HCO3− from the superfusate, in contrast, reduces the light‐evoked ECS decrease by 95.6%. Inhibition of the monocarboxylate transporter with alpha‐cyano‐4‐hydroxycinnamate (4‐CIN) also reduces the ECS shrinkage, but only by 32.5%. We tested whether the swelling of Müller cells, the principal glial cells of the retina, is responsible for the light‐evoked ECS shrinkage. Light stimulation evoked a 6.3% increase in the volume of the fine processes of Müller cells. This volume increase was reduced by 97.1% when HCO3− was removed from the superfusate. We conclude that a large fraction of the activity‐dependent decrease in ECS α is generated by the activation of the Na+/HCO3− cotransporter in Müller cells. The monocarboxylate transporter may also contribute to the response.
Collapse
Affiliation(s)
- Pei-Pei Chiang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sidney P Kuo
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eric A Newman
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
5
|
Bodnar CN, Watson JB, Higgins EK, Quan N, Bachstetter AD. Inflammatory Regulation of CNS Barriers After Traumatic Brain Injury: A Tale Directed by Interleukin-1. Front Immunol 2021; 12:688254. [PMID: 34093593 PMCID: PMC8176952 DOI: 10.3389/fimmu.2021.688254] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/05/2021] [Indexed: 01/13/2023] Open
Abstract
Several barriers separate the central nervous system (CNS) from the rest of the body. These barriers are essential for regulating the movement of fluid, ions, molecules, and immune cells into and out of the brain parenchyma. Each CNS barrier is unique and highly dynamic. Endothelial cells, epithelial cells, pericytes, astrocytes, and other cellular constituents each have intricate functions that are essential to sustain the brain's health. Along with damaging neurons, a traumatic brain injury (TBI) also directly insults the CNS barrier-forming cells. Disruption to the barriers first occurs by physical damage to the cells, called the primary injury. Subsequently, during the secondary injury cascade, a further array of molecular and biochemical changes occurs at the barriers. These changes are focused on rebuilding and remodeling, as well as movement of immune cells and waste into and out of the brain. Secondary injury cascades further damage the CNS barriers. Inflammation is central to healthy remodeling of CNS barriers. However, inflammation, as a secondary pathology, also plays a role in the chronic disruption of the barriers' functions after TBI. The goal of this paper is to review the different barriers of the brain, including (1) the blood-brain barrier, (2) the blood-cerebrospinal fluid barrier, (3) the meningeal barrier, (4) the blood-retina barrier, and (5) the brain-lesion border. We then detail the changes at these barriers due to both primary and secondary injury following TBI and indicate areas open for future research and discoveries. Finally, we describe the unique function of the pro-inflammatory cytokine interleukin-1 as a central actor in the inflammatory regulation of CNS barrier function and dysfunction after a TBI.
Collapse
Affiliation(s)
- Colleen N. Bodnar
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - James B. Watson
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Emma K. Higgins
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| | - Adam D. Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
6
|
Liu L, Chai L, Ran J, Yang Y, Zhang L. BAI1 acts as a tumor suppressor in lung cancer A549 cells by inducing metabolic reprogramming via the SCD1/HMGCR module. Carcinogenesis 2021; 41:1724-1734. [PMID: 32255478 DOI: 10.1093/carcin/bgaa036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 03/02/2020] [Accepted: 04/06/2020] [Indexed: 02/05/2023] Open
Abstract
Brain-specific angiogenesis inhibitor 1 (BAI1) is an important tumor suppressor in multiple cancers. However, the mechanisms behind its anti-tumor activity, particularly the relationship between BAI1 and metabolic aberrant of a tumor, remained unveiled. This study aimed to investigate whether BAI1 could inhibit biological functions in lung cancer A549 cells and the critical regulating molecules that induce metabolic reprogramming. Immunohistochemistry staining was performed to analyze whether variations in the expression of BAI1 in tumor tissues contributes to poor prognosis of lung cancer. Overexpressed BAI1 (BAI1-OE-A549) and control (Vector-NC-A549) were generated by lentiviral transfection. Biological function assays (proliferation, apoptosis, colony formation, invasion and in vivo metastasis), as well as metabolic reprogramming (by the Warburg effect and the glycolytic rate), were performed in both groups. Our results indicated that lower levels of BAI1 contributed to poor prognosis of lung cancer patients. Furthermore, overexpressed of BAI1 dramatically inhibited proliferation, migration, invasion, colony formation and in vivo metastasis of A549 cells. The Warburg effect and the Seahorse assay revealed that BAI1-OE induced metabolism reprogramming by inhibiting the Warburg effect and glycolysis. Further exploration indicated that BAI1 induced metabolic reprogramming by upregulating stearoyl-CoA desaturase 1 (SCD1) and inhibited 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR). Our study revealed a novel mechanism through which BAI1 acted as tumor suppressor by inducing metabolic reprogramming via the SCD1 and HMGCR module.
Collapse
Affiliation(s)
- Lei Liu
- Lab of Pathology, Key Lab of Transplantation Engineering and Immunology, Ministry of Health, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.,Department of Laboratory Medicine, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province, China
| | - Li Chai
- Research Core Facility, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jingjing Ran
- Lab of Pathology, Key Lab of Transplantation Engineering and Immunology, Ministry of Health, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ying Yang
- Center of Precision Medicine, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Zhang
- Lab of Pathology, Key Lab of Transplantation Engineering and Immunology, Ministry of Health, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
7
|
Fairweather SJ, Shah N, Brӧer S. Heteromeric Solute Carriers: Function, Structure, Pathology and Pharmacology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 21:13-127. [PMID: 33052588 DOI: 10.1007/5584_2020_584] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Solute carriers form one of three major superfamilies of membrane transporters in humans, and include uniporters, exchangers and symporters. Following several decades of molecular characterisation, multiple solute carriers that form obligatory heteromers with unrelated subunits are emerging as a distinctive principle of membrane transporter assembly. Here we comprehensively review experimentally established heteromeric solute carriers: SLC3-SLC7 amino acid exchangers, SLC16 monocarboxylate/H+ symporters and basigin/embigin, SLC4A1 (AE1) and glycophorin A exchanger, SLC51 heteromer Ost α-Ost β uniporter, and SLC6 heteromeric symporters. The review covers the history of the heteromer discovery, transporter physiology, structure, disease associations and pharmacology - all with a focus on the heteromeric assembly. The cellular locations, requirements for complex formation, and the functional role of dimerization are extensively detailed, including analysis of the first complete heteromer structures, the SLC7-SLC3 family transporters LAT1-4F2hc, b0,+AT-rBAT and the SLC6 family heteromer B0AT1-ACE2. We present a systematic analysis of the structural and functional aspects of heteromeric solute carriers and conclude with common principles of their functional roles and structural architecture.
Collapse
Affiliation(s)
- Stephen J Fairweather
- Research School of Biology, Australian National University, Canberra, ACT, Australia. .,Resarch School of Chemistry, Australian National University, Canberra, ACT, Australia.
| | - Nishank Shah
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Stefan Brӧer
- Research School of Biology, Australian National University, Canberra, ACT, Australia.
| |
Collapse
|
8
|
Yumnamcha T, Guerra M, Singh LP, Ibrahim AS. Metabolic Dysregulation and Neurovascular Dysfunction in Diabetic Retinopathy. Antioxidants (Basel) 2020; 9:E1244. [PMID: 33302369 PMCID: PMC7762582 DOI: 10.3390/antiox9121244] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/11/2022] Open
Abstract
Diabetic retinopathy is a major cause of ocular complications in patients with type 1 and type 2 diabetes in developed countries. Due to the continued increase in the number of people with obesity and diabetes in the United States of America and globally, the incidence of diabetic retinopathy is expected to increase significantly in the coming years. Diabetic retinopathy is widely accepted as a combination of neurodegenerative and microvascular changes; however, which change occurs first is not yet understood. Although the pathogenesis of diabetic retinopathy is very complex, regulated by numerous signaling pathways and cellular processes, maintaining glucose homeostasis is still an essential component for normal physiological functioning of retinal cells. The maintenance of glucose homeostasis is finely regulated by coordinated interplay between glycolysis, Krebs cycle, and oxidative phosphorylation. Glycolysis is the most conserved metabolic pathway in biology and is tightly regulated to maintain a steady-state concentration of glycolytic intermediates; this regulation is called scheduled or regulated glycolysis. However, an abnormal increase in glycolytic flux generates large amounts of intermediate metabolites that can be shunted into different damaging pathways including the polyol pathway, hexosamine pathway, diacylglycerol-dependent activation of the protein kinase C pathway, and Amadori/advanced glycation end products (AGEs) pathway. In addition, disrupting the balance between glycolysis and oxidative phosphorylation leads to other biochemical and molecular changes observed in diabetic retinopathy including endoplasmic reticulum-mitochondria miscommunication and mitophagy dysregulation. This review will focus on how dysregulation of glycolysis contributes to diabetic retinopathy.
Collapse
Affiliation(s)
- Thangal Yumnamcha
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Michael Guerra
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Lalit Pukhrambam Singh
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
| | - Ahmed S. Ibrahim
- Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, MI 48201, USA; (M.G.); (L.P.S.)
- Department of Pharmacology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
9
|
Synthesis and anticancer activity of new coumarin-3-carboxylic acid derivatives as potential lactatetransportinhibitors. Bioorg Med Chem 2020; 29:115870. [PMID: 33221062 DOI: 10.1016/j.bmc.2020.115870] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/01/2020] [Accepted: 11/07/2020] [Indexed: 12/11/2022]
Abstract
As an oncometabolite, lactate plays a very important role in tumor proliferation, metastasis, angiogenesis, immune escape and other tumor biological functions. Pharmacological inhibition oflactate transport has been viewed as a promising therapeutic strategy to target a range of human cancers. In this study, a series of new coumarin-3-carboxylic acid derivatives 5a-t and 9a-b were synthesized and evaluated as lactate transport inhibitors. Their cytotoxic activity has been tested against three cell lines high-expressing and low-expressing monocarboxylate transporter 1 (MCT1) which acts as the main carrier for lactate. Compound 5c-e, 5g-i and 5m-o showed significant cytotoxicity and good selectivity against Hela and HCT116 cell lines with high MCT1 expression. Notably, coumarin-3-hydrazide 5o, the lead molecule with the most potent cytotoxic activity, exhibitedsignificant anti-proliferationandapoptosisinductioneffects. Further studies revealed that compound 5o decreased the expression level of target MCT1, and suppressed the energetic metabolism of Hela and HCT116 cells byremarkably reducing glucoseconsumptionandlactate production. Additionally, compound 5o induced intracellular lactate accumulation and inhibited lactate uptake, which implied that it blocked lactate transport via MCT1. These results indicate a good start point for the development of lactate transport inhibitors as new anticancer agents.
Collapse
|
10
|
Allen AE, Martin EA, Greenwood K, Grant C, Vince P, Lucas RJ, Redfern WS. Effects of a monocarboxylate transport 1 inhibitor, AZD3965, on retinal and visual function in the rat. Br J Pharmacol 2020; 177:4734-4749. [PMID: 32833237 DOI: 10.1111/bph.15239] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 07/21/2020] [Accepted: 07/24/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Inhibition of monocarboxylate transport 1 (MCT1) is of interest in targeting highly glycolytic tumours. However, MCT1 is expressed in retina, and so inhibition of MCT1 could affect retinal function. EXPERIMENTAL APPROACH AZD3965, an MCT1 inhibitor selected for clinical development, and two additional MCT1 inhibitors were evaluated for effects on visual acuity in albino (Han Wistar) rats. The effects of AZD3965 on visual acuity and electroretinography (ERG) were further investigated in pigmented (Long-Evans) rats, with dosing for up to 7 days. KEY RESULTS All three MCT1 inhibitors reduced visual acuity within 2 h of dosing, suggesting a class effect. The deficit caused by AZD3965 (1,000 mg·kg-1 p.o. per day for 4 days) in Long Evans rats recovered to pre-dose levels 7 days after cessation of dosing. AZD3965 (50 to 1,000 mg·kg-1 p.o.) reduced the amplitude of scotopic a- and b-waves, and photopic b-wave of the ERG in a dose-related fashion, within 2 h of dosing. The effects on the scotopic ERG had diminished by Day 7 of dosing, demonstrating partial restoration of function despite continued treatment. Seven days after cessation of dosing at the highest dose tested (1,000 mg·kg-1 ), there was recovery of both scotopic a- and b- waves and, to a lesser extent, photopic b-wave. ERG was affected at lower plasma exposures than was visual function. CONCLUSIONS AND IMPLICATIONS This study clarifies the role of the MCT1 transporter in retinal function. The monitorability of the functional effects on the retina enabled safe clinical use of AZD3965.
Collapse
Affiliation(s)
- Annette E Allen
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Elizabeth A Martin
- Regulatory Safety Centre of Excellence, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Katherine Greenwood
- Regulatory Safety Centre of Excellence, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.,Gentronix Limited, Cheshire, UK
| | - Claire Grant
- Regulatory Safety Centre of Excellence, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Peter Vince
- Regulatory Safety Centre of Excellence, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Robert J Lucas
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - William S Redfern
- Regulatory Safety Centre of Excellence, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.,Certara UK Limited, Sheffield, UK
| |
Collapse
|
11
|
Khan AZ, Utheim TP, Moe MC, Aass HCD, Sapkota D, Vallenari EM, Eidet JR. The Silk Protein Sericin Promotes Viability of ARPE-19 and Induced Pluripotent Stem Cell-Derived Retinal Pigment Epithelial Cells in vitro. Curr Eye Res 2020; 46:504-514. [PMID: 32777180 DOI: 10.1080/02713683.2020.1809001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE Maintaining mature and viable retinal pigment epithelial cells (RPE) in vitro has proven challenging. Investigating compounds that can promote RPE-viability and maturation is motivated by RPE transplantation research, the quest to understand RPE physiology, and a desire to modulate RPE in pathological states. We have previously reported that the silk protein sericin promotes viability, maturation, and pigmentation of human fetal RPE. In the present study, our aim was to uncover whether these effects can be seen in adult retinal pigment epithelial cell line-19 (ARPE-19) and induced pluripotent stem cell-derived RPE (iPSC-RPE). METHODS ARPE-19 and iPSC-RPE were cultured with or without 10 mg/mL sericin. After 7 days, viability was assessed with calcein-acetoxymethyl ester (CAM) and ethidium homodimer-1 (EH-1) assays, flow cytometry, and morphometric analysis. Expression levels of RPE65, tyrosinase, and Pmel17 were quantified to compare maturation between the sericin-treated and control cultures. Light microscopy and staining of the tight junction protein zonula occludens protein 1 (ZO-1) were employed to study sericin's effects on RPE morphology. We also measured culture medium pH, glucose, lactate, and extracellular ion content. RESULTS Sericin-supplemented RPE cultures demonstrated significantly better viability compared to control cultures. Sericin appeared to improve ARPE-19 maturation and morphology in vitro. No effects were seen on RPE pigmentation with the concentration of sericin and duration of cell culture herein reported. CONCLUSIONS This is the first study to demonstrate that supplementing the culture media with sericin promotes the viability of iPSC-RPE and ARPE-19. Sericin's viability-promoting effects may have important implications for retinal therapeutics and regenerative medicine research.
Collapse
Affiliation(s)
- Ayyad Zartasht Khan
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Tor Paaske Utheim
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway.,Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway.,Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway.,Department of Ophthalmology, Oslo University Hospital, Oslo, Norway.,Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Bergen, Norway.,Department of Ophthalmology, Stavanger University Hospital, Stavanger, Norway
| | - Morten Carstens Moe
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
| | | | - Dipak Sapkota
- Department of Oral Biology, Faculty of Dentistry, University of Oslo, Oslo, Norway
| | | | - Jon Roger Eidet
- Department of Ophthalmology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
12
|
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
|
13
|
Liu X, Pan G. Roles of Drug Transporters in Blood-Retinal Barrier. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1141:467-504. [PMID: 31571172 PMCID: PMC7120327 DOI: 10.1007/978-981-13-7647-4_10] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Blood-retinal barrier (BRB) includes inner BRB (iBRB) and outer BRB (oBRB), which are formed by retinal capillary endothelial (RCEC) cells and by retinal pigment epithelial (RPE) cells in collaboration with Bruch's membrane and the choriocapillaris, respectively. Functions of the BRB are to regulate fluids and molecular movement between the ocular vascular beds and retinal tissues and to prevent leakage of macromolecules and other potentially harmful agents into the retina, keeping the microenvironment of the retina and retinal neurons. These functions are mainly attributed to absent fenestrations of RCECs, tight junctions, expression of a great diversity of transporters, and coverage of pericytes and glial cells. BRB existence also becomes a reason that systemic administration for some drugs is not suitable for the treatment of retinal diseases. Some diseases (such as diabetes and ischemia-reperfusion) impair BRB function via altering tight junctions, RCEC death, and transporter expression. This chapter will illustrate function of BRB, expressions and functions of these transporters, and their clinical significances.
Collapse
Affiliation(s)
- Xiaodong Liu
- grid.254147.10000 0000 9776 7793School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu China
| | - Guoyu Pan
- grid.9227.e0000000119573309Shanghai Institute of Materia Medica, Chinese Academy of Science, Shanghai, Shanghai China
| |
Collapse
|
14
|
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: 74] [Impact Index Per Article: 12.3] [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
|
15
|
Vellonen KS, Hellinen L, Mannermaa E, Ruponen M, Urtti A, Kidron H. Expression, activity and pharmacokinetic impact of ocular transporters. Adv Drug Deliv Rev 2018; 126:3-22. [PMID: 29248478 DOI: 10.1016/j.addr.2017.12.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/24/2017] [Accepted: 12/13/2017] [Indexed: 12/13/2022]
Abstract
The eye is protected by several tissues that limit the permeability and entry of potentially harmful substances, but also hamper the delivery of drugs in the treatment of ocular diseases. Active transport across the ocular barriers may affect drug distribution, but the impact of drug transporters on ocular drug delivery is not well known. We have collected and critically reviewed the literature for ocular expression and activity of known drug transporters. The review concentrates on drug transporters that have been functionally characterized in ocular tissues or primary cells and on transporters for which there is available expression data at the protein level. Species differences are highlighted, since these may explain observed inconsistencies in the influence of specific transporters on drug disposition. There is variable evidence about the pharmacokinetic role of transporters in ocular tissues. The strongest evidence for the role of active transport is available for the blood-retinal barrier. We explored the role of active transport in the cornea and blood retinal barrier with pharmacokinetic simulations. The simulations show that the active transport is important only in the case of specific parameter combinations.
Collapse
|
16
|
Li X, Yu X, Dai D, Song X, Xu W. The altered glucose metabolism in tumor and a tumor acidic microenvironment associated with extracellular matrix metalloproteinase inducer and monocarboxylate transporters. Oncotarget 2018; 7:23141-55. [PMID: 27009812 PMCID: PMC5029616 DOI: 10.18632/oncotarget.8153] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 01/13/2016] [Indexed: 02/06/2023] Open
Abstract
Extracellular matrix metalloproteinase inducer, also knowns as cluster of differentiation 147 (CD147) or basigin, is a widely distributed cell surface glycoprotein that is involved in numerous physiological and pathological functions, especially in tumor invasion and metastasis. Monocarboxylate transporters (MCTs) catalyze the proton-linked transport of monocarboxylates such as L-lactate across the plasma membrane to preserve the intracellular pH and maintain cell homeostasis. As a chaperone to some MCT isoforms, CD147 overexpression significantly contributes to the metabolic transformation of tumor. This overexpression is characterized by accelerated aerobic glycolysis and lactate efflux, and it eventually provides the tumor cells with a metabolic advantage and an invasive phenotype in the acidic tumor microenvironment. This review highlights the roles of CD147 and MCTs in tumor cell metabolism and the associated molecular mechanisms. The regulation of CD147 and MCTs may prove to be with a therapeutic potential for tumors through the metabolic modification of the tumor microenvironment.
Collapse
Affiliation(s)
- Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiaozhou Yu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Dong Dai
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Xiuyu Song
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,National Clinical Research Center for Cancer, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
17
|
Ferguson BS, Rogatzki MJ, Goodwin ML, Kane DA, Rightmire Z, Gladden LB. Lactate metabolism: historical context, prior misinterpretations, and current understanding. Eur J Appl Physiol 2018; 118:691-728. [PMID: 29322250 DOI: 10.1007/s00421-017-3795-6] [Citation(s) in RCA: 245] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 12/22/2017] [Indexed: 02/07/2023]
Abstract
Lactate (La-) has long been at the center of controversy in research, clinical, and athletic settings. Since its discovery in 1780, La- has often been erroneously viewed as simply a hypoxic waste product with multiple deleterious effects. Not until the 1980s, with the introduction of the cell-to-cell lactate shuttle did a paradigm shift in our understanding of the role of La- in metabolism begin. The evidence for La- as a major player in the coordination of whole-body metabolism has since grown rapidly. La- is a readily combusted fuel that is shuttled throughout the body, and it is a potent signal for angiogenesis irrespective of oxygen tension. Despite this, many fundamental discoveries about La- are still working their way into mainstream research, clinical care, and practice. The purpose of this review is to synthesize current understanding of La- metabolism via an appraisal of its robust experimental history, particularly in exercise physiology. That La- production increases during dysoxia is beyond debate, but this condition is the exception rather than the rule. Fluctuations in blood [La-] in health and disease are not typically due to low oxygen tension, a principle first demonstrated with exercise and now understood to varying degrees across disciplines. From its role in coordinating whole-body metabolism as a fuel to its role as a signaling molecule in tumors, the study of La- metabolism continues to expand and holds potential for multiple clinical applications. This review highlights La-'s central role in metabolism and amplifies our understanding of past research.
Collapse
Affiliation(s)
- Brian S Ferguson
- College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthew J Rogatzki
- Department of Health and Exercise Science, Appalachian State University, Boone, NC, USA
| | - Matthew L Goodwin
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, Salt Lake City, UT, USA
| | - Daniel A Kane
- Department of Human Kinetics, St. Francis Xavier University, Antigonish, Canada
| | - Zachary Rightmire
- School of Kinesiology, Auburn University, 301 Wire Road, Auburn, AL, 36849, USA
| | - L Bruce Gladden
- School of Kinesiology, Auburn University, 301 Wire Road, Auburn, AL, 36849, USA.
| |
Collapse
|
18
|
Toft-Kehler AK, Skytt DM, Kolko M. A Perspective on the Müller Cell-Neuron Metabolic Partnership in the Inner Retina. Mol Neurobiol 2017; 55:5353-5361. [PMID: 28929338 DOI: 10.1007/s12035-017-0760-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/31/2017] [Indexed: 10/18/2022]
Abstract
The Müller cells represent the predominant macroglial cell in the retina. In recent decades, Müller cells have been acknowledged to be far more influential on neuronal homeostasis in the retina than previously assumed. With its unique localization, spanning the entire retina being interposed between the vessels and neurons, Müller cells are responsible for the functional and metabolic support of the surrounding neurons. As a consequence of major energy demands in the retina, high levels of glucose are consumed and processed by Müller cells. The present review provides a perspective on the symbiotic relationship between Müller cells and inner retinal neurons on a cellular level by emphasizing the essential role of energy metabolism within Müller cells in relation to retinal neuron survival.
Collapse
Affiliation(s)
- A K Toft-Kehler
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - D M Skytt
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Miriam Kolko
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark. .,Department of Ophthalmology, Zealand University Hospital, Vestermarksvej 23, 4000, Roskilde, Denmark. .,Department of Ophthalmology, Copenhagen University Hospital, Rigshospitalet, Nordre Ringvej 57, 2600, Glostrup, Denmark.
| |
Collapse
|
19
|
Morris ME, Rodriguez-Cruz V, Felmlee MA. SLC and ABC Transporters: Expression, Localization, and Species Differences at the Blood-Brain and the Blood-Cerebrospinal Fluid Barriers. AAPS JOURNAL 2017; 19:1317-1331. [PMID: 28664465 DOI: 10.1208/s12248-017-0110-8] [Citation(s) in RCA: 124] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/05/2017] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB) and the blood-cerebrospinal fluid barrier (BCSFB) separate the brain and cerebrospinal fluid (CSF) from the systemic circulation and represent a barrier to the uptake of both endogenous compounds and xenobiotics into the brain. For compounds whose passive diffusion is limited due to their ionization or hydrophilicity, membrane transporters can facilitate their uptake across the BBB or BCSFB. Members of the solute carrier (SLC) and ATP-binding case (ABC) families are present on these barriers. Differences exist in the localization and expression of transport proteins between the BBB and BCSFB, resulting in functional differences in transport properties. This review focuses on the expression, membrane localization, and different isoforms present at each barrier. Diseases that affect the central nervous system including brain tumors, HIV, Alzheimer's disease, Parkinson's disease, and stroke affect the integrity and expression of transporters at the BBB and BCSFB and will be briefly reviewed.
Collapse
Affiliation(s)
- Marilyn E Morris
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA.
| | - Vivian Rodriguez-Cruz
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York at Buffalo, Buffalo, New York, 14214-8033, USA
| | - Melanie A Felmlee
- Department of Pharmaceutics and Medicinal Chemistry, Thomas J Long School of Pharmacy and Health Sciences, University of the Pacific, 3601 Pacific Ave, Stockton, California, 95211, USA
| |
Collapse
|
20
|
Toft-Kehler AK, Skytt DM, Svare A, Lefevere E, Van Hove I, Moons L, Waagepetersen HS, Kolko M. Mitochondrial function in Müller cells - Does it matter? Mitochondrion 2017; 36:43-51. [PMID: 28179130 DOI: 10.1016/j.mito.2017.02.002] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 01/26/2017] [Accepted: 02/03/2017] [Indexed: 11/17/2022]
Abstract
Growing evidence suggests that mitochondrial dysfunction might play a key role in the pathogenesis of age-related neurodegenerative inner retinal diseases such as diabetic retinopathy and glaucoma. Therefore, the present review provides a perspective on the impact of functional mitochondria in the most predominant glial cells of the retina, the Müller cells. Müller cells span the entire thickness of the neuroretina and are in close proximity to retinal cells including the retinal neurons that provides visual signaling to the brain. Among multiple functions, Müller cells are responsible for the removal of neurotransmitters, buffering potassium, and providing neurons with essential metabolites. Thus, Müller cells are responsible for a stable metabolic dialogue in the inner retina and their crucial role in supporting retinal neurons is indisputable. Müller cell functions require considerable energy production and previous literature has primarily emphasized glycolysis as the main energy provider. However, recent studies highlight the need of mitochondrial ATP production to upheld Müller cell functions. Therefore, the present review aims to provide an overview of the current evidence on the impact of mitochondrial functions in Müller cells.
Collapse
Affiliation(s)
- Anne Katrine Toft-Kehler
- Eye Translational Research Unit, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen O, Denmark; Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark.
| | - Dorte Marie Skytt
- Eye Translational Research Unit, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen O, Denmark; Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark
| | - Alicia Svare
- Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark
| | - Evy Lefevere
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Inge Van Hove
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Helle S Waagepetersen
- Neuromet, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen O, Denmark
| | - Miriam Kolko
- Eye Translational Research Unit, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen O, Denmark; Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark; Zealand University Hospital, Department of Ophthalmology, Vestermarksvej 23, 4000 Roskilde, Denmark.
| |
Collapse
|
21
|
Elizondo-Vega R, García-Robles MA. Molecular Characteristics, Regulation, and Function of Monocarboxylate Transporters. ADVANCES IN NEUROBIOLOGY 2017; 16:255-267. [PMID: 28828614 DOI: 10.1007/978-3-319-55769-4_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Lactate transporters play an important role in the glutamate recycling. Here their kinetics and tissue distribution with emphasis on the brain are addressed. Recent evidence shows their participation in important brain functions that involve intercellular communication, such as hypothalamic glucose sensing. Furthermore, we describe the regulation of their expression and some animal models that have allowed clarification of their functions.
Collapse
|
22
|
Zieger M, Punzo C. Improved cell metabolism prolongs photoreceptor survival upon retinal-pigmented epithelium loss in the sodium iodate induced model of geographic atrophy. Oncotarget 2016; 7:9620-33. [PMID: 26883199 PMCID: PMC4891071 DOI: 10.18632/oncotarget.7330] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 01/23/2016] [Indexed: 01/06/2023] Open
Abstract
Age-related macular degeneration (AMD) is characterized by malfunction and loss of retinal-pigmented epithelium (RPE) cells. Because the RPE transfers nutrients from the choriocapillaris to photoreceptor (PR), PRs are affected as well. Geographic atrophy (GA) is an advanced form of AMD characterized by severe vision impairment due to RPE loss over large areas. Currently there is no treatment to delay the degeneration of nutrient deprived PRs once RPE cells die. Here we show that cell-autonomous activation of the key regulator of cell metabolism, the kinase mammalian target of rapamycin complex 1 (mTORC1), delays PR death in the sodium iodate induced model of RPE atrophy. Consistent with this finding loss of mTORC1 in cones accelerates cone death as cones fail to balance demand with supply. Interestingly, promoting rod survival does not promote cone survival in this model of RPE atrophy as both, rods and cones suffer from a sick and dying RPE. The findings suggest that activation of metabolic genes downstream of mTORC1 can serve as a strategy to prolong PR survival when RPE cells malfunction or die.
Collapse
Affiliation(s)
- Marina Zieger
- Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Claudio Punzo
- Department of Ophthalmology and Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
23
|
Kishimoto A, Takahashi-Iwanaga H, Watanabe M M, Iwanaga T. Differential expression of endothelial nutrient transporters (MCT1 and GLUT1) in the developing eyes of mice. Exp Eye Res 2016; 153:170-177. [PMID: 27793618 DOI: 10.1016/j.exer.2016.10.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/05/2016] [Accepted: 10/24/2016] [Indexed: 12/31/2022]
Abstract
The blood-brain barrier in the neonatal brain expresses the monocarboxylate transporter (MCT)-1 rather than the glucose transporter (GLUT)-1, due to the special energy supply during the suckling period. The hyaloid vascular system, consisting of the vasa hyaloidea propria and tunica vasculosa lentis, is a temporary vasculature present only during the early development of mammalian eyes and later regresses. Although the ocular vasculature manifests such a unique developmental process, no information is available concerning the expression of endothelial nutrient transporters in the developing eye. The present immunohistochemical study using whole mount preparations of murine eyes found that the hyaloid vascular system predominantly expressed GLUT1 in the endothelium, in contrast to the brain endothelium. Characteristically, the endothelium in peripheral regions of the neonatal hyaloid vessels displayed a mosaic pattern of MCT1-immunoreactive cells scattered within the GLUT1-expressing endothelium. The proper retinal vessels first developed by sprouting angiogenesis endowed with filopodia, which were absolutely free from the immunoreactivities of GLUT1 and MCT1. The remodeling retinal capillary networks and veins in the surface layer of the retina mainly expressed MCT1 until the weaning period. Immunostaining of MCT1 in the retina revealed fine radicular processes projecting from the endothelium, differing from the MCT1-immunonegative filopodia. These findings suggest that the expression of nutrient transporters in the ocular blood vessels is differentially regulated at a cellular level and that the neonatal eyes provide an interesting model for research on nutrient transporters in the endothelium.
Collapse
Affiliation(s)
- Ayuko Kishimoto
- Laboratory of Histology and Cytology, Department of Anatomy, Hokkaido University, Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Hiromi Takahashi-Iwanaga
- Laboratory of Histology and Cytology, Department of Anatomy, Hokkaido University, Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Masahiko Watanabe M
- Laboratory of Histology and Cytology, Department of Anatomy, Hokkaido University, Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Department of Anatomy, Hokkaido University, Graduate School of Medicine, Sapporo 060-8638, Japan.
| |
Collapse
|
24
|
Pannicke T, Ivo Chao T, Reisenhofer M, Francke M, Reichenbach A. Comparative electrophysiology of retinal Müller glial cells-A survey on vertebrate species. Glia 2016; 65:533-568. [PMID: 27767232 DOI: 10.1002/glia.23082] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 09/15/2016] [Accepted: 09/28/2016] [Indexed: 12/13/2022]
Abstract
Müller cells are the dominant macroglial cells in the retina of all vertebrates. They fulfill a variety of functions important for retinal physiology, among them spatial buffering of K+ ions and uptake of glutamate and other neurotransmitters. To this end, Müller cells express inwardly rectifying K+ channels and electrogenic glutamate transporters. Moreover, a lot of voltage- and ligand-gated ion channels, aquaporin water channels, and electrogenic transporters are expressed in Müller cells, some of them in a species-specific manner. For example, voltage-dependent Na+ channels are found exclusively in some but not all mammalian species. Whereas a lot of data exist from amphibians and mammals, the results from other vertebrates are sparse. It is the aim of this review to present a survey on Müller cell electrophysiology covering all classes of vertebrates. The focus is on functional studies, mainly performed using the whole-cell patch-clamp technique. However, data about the expression of membrane channels and transporters from immunohistochemistry are also included. Possible functional roles of membrane channels and transporters are discussed. Obviously, electrophysiological properties involved in the main functions of Müller cells developed early in vertebrate evolution. GLIA 2017;65:533-568.
Collapse
Affiliation(s)
- Thomas Pannicke
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
| | - T Ivo Chao
- Institute of Anatomy and Cell Biology, Medical School Göttingen, Germany
| | - Miriam Reisenhofer
- Department of Chemistry, University of Zürich, Switzerland
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Mike Francke
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
- Sächsischer Inkubator für klinische Translation (SIKT), Universität Leipzig, Germany
| | - Andreas Reichenbach
- Paul-Flechsig-Institut für Hirnforschung, Abteilung Pathophysiologie der Neuroglia, Universität Leipzig, Germany
| |
Collapse
|
25
|
Lactate Transport and Receptor Actions in Retina: Potential Roles in Retinal Function and Disease. Neurochem Res 2015; 41:1229-36. [PMID: 26677077 DOI: 10.1007/s11064-015-1792-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 11/06/2015] [Accepted: 11/08/2015] [Indexed: 01/01/2023]
Abstract
In retina, like in brain, lactate equilibrates across cell membranes via monocarboxylate transporters and in the extracellular space by diffusion, forming a basis for the action of lactate as a transmitter of metabolic signals. In the present paper, we argue that the lactate receptor GPR81, also known as HCAR1, may contribute importantly to the control of retinal cell functions in health and disease. GPR81, a G-protein coupled receptor, is known to downregulate cAMP both in adipose and nervous tissue. The receptor also acts through other down-stream mechanisms to control functions, such as excitability, metabolism and inflammation. Recent publications predict effects of the lactate receptor on neurodegeneration. Neurodegenerative diseases in retina, where the retinal ganglion cells die, notably glaucoma and diabetic retinopathy, may be linked to disturbed lactate homeostasis. Pilot studies reveal high GPR81 mRNA in retina and indicate GPR81 localization in Müller cells and retinal ganglion cells. Moreover, monocarboxylate transporters are expressed in retinal cells. We envision that lactate receptors and transporters could be useful future targets of novel therapeutic strategies to protect neurons and prevent or counteract glaucoma as well as other retinal diseases.
Collapse
|
26
|
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: 186] [Impact Index Per Article: 18.6] [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
|
27
|
Morland C, Lauritzen KH, Puchades M, Holm-Hansen S, Andersson K, Gjedde A, Attramadal H, Storm-Mathisen J, Bergersen LH. The lactate receptor, G-protein-coupled receptor 81/hydroxycarboxylic acid receptor 1: Expression and action in brain. J Neurosci Res 2015; 93:1045-55. [DOI: 10.1002/jnr.23593] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/21/2015] [Accepted: 03/23/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Cecilie Morland
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
- The Brain and Muscle Energy Group; Department of Oral Biology; University of Oslo; Oslo Norway
| | - Knut Husø Lauritzen
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
| | - Maja Puchades
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
| | - Signe Holm-Hansen
- Department of Neuroscience and Pharmacology; University of Copenhagen; Copenhagen Denmark
| | - Krister Andersson
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
| | - Albert Gjedde
- Department of Neuroscience and Pharmacology; University of Copenhagen; Copenhagen Denmark
| | - Håvard Attramadal
- Institute for Surgical Research, Oslo University Hospital; Oslo Norway
- Center for Heart Failure Research, University of Oslo; Oslo Norway
| | - Jon Storm-Mathisen
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
| | - Linda Hildegard Bergersen
- The Brain and Muscle Energy Group; Department of Anatomy; Institute of Basic Medical Sciences, University of Oslo; Oslo Norway
- Department of Neuroscience and Pharmacology; University of Copenhagen; Copenhagen Denmark
- Center for Healthy Aging; Faculty of Health Sciences; University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
28
|
Attenuation of choroidal neovascularization by histone deacetylase inhibitor. PLoS One 2015; 10:e0120587. [PMID: 25807249 PMCID: PMC4373846 DOI: 10.1371/journal.pone.0120587] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 01/24/2015] [Indexed: 01/13/2023] Open
Abstract
Choroidal neovascularization (CNV) is a blinding complication of age-related macular degeneration that manifests as the growth of immature choroidal blood vessels through Bruch’s membrane, where they can leak fluid or hemorrhage under the retina. Here, we demonstrate that the histone deacetylase inhibitor (HDACi) trichostatin A (TSA) can down-regulate the pro-angiogenic hypoxia-inducible factor-1α and vascular endothelial growth factor (VEGF), and up-regulate the anti-angiogenic and neuro-protective pigment epithelium derived factor in human retinal pigment epithelial (RPE) cells. Most strikingly, TSA markedly down-regulates the expression of VEGF receptor-2 in human vascular endothelial cells and, thus, can knock down pro-angiogenic cell signaling. Additionally, TSA suppresses CNV-associated wound healing response and RPE epithelial-mesenchymal transdifferentiation. In the laser-induced model of CNV using C57Bl/6 mice, systemic administration of TSA significantly reduces fluorescein leakage and the size of CNV lesions at post—laser days 7 and 14 as well as the immunohistochemical expression of VEGF, VEGFR2, and smooth muscle actin in CNV lesions at post-laser day 7. This report suggests that TSA, and possibly HDACi’s in general, should be further evaluated for their therapeutic potential for the treatment of CNV.
Collapse
|
29
|
|
30
|
IWANAGA T, KISHIMOTO A. Cellular distributions of monocarboxylate transporters: a review . Biomed Res 2015; 36:279-301. [DOI: 10.2220/biomedres.36.279] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Affiliation(s)
- Toshihiko IWANAGA
- Laboratory of Histology and Cytology, Graduate School of Medicine, Hokkaido University
| | - Ayuko KISHIMOTO
- Laboratory of Histology and Cytology, Graduate School of Medicine, Hokkaido University
| |
Collapse
|
31
|
Baek G, Tse YF, Hu Z, Cox D, Buboltz N, McCue P, Yeo CJ, White MA, DeBerardinis RJ, Knudsen ES, Witkiewicz AK. MCT4 defines a glycolytic subtype of pancreatic cancer with poor prognosis and unique metabolic dependencies. Cell Rep 2014; 9:2233-49. [PMID: 25497091 DOI: 10.1016/j.celrep.2014.11.025] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 11/04/2014] [Accepted: 11/17/2014] [Indexed: 12/11/2022] Open
Abstract
KRAS mutation, which occurs in ∼ 95% of pancreatic ductal adenocarcinoma (PDA), has been shown to program tumor metabolism. MCT4 is highly upregulated in a subset of PDA with a glycolytic gene expression program and poor survival. Models with high levels of MCT4 preferentially employ glycolytic metabolism. Selectively in such "addicted" models, MCT4 attenuation compromised glycolytic flux with compensatory induction of oxidative phosphorylation and scavenging of metabolites by macropinocytosis and autophagy. In spite of these adaptations, MCT4 depletion induced cell death characterized by elevated reactive oxygen species and metabolic crisis. Cell death induced by MCT4-depletion was augmented by inhibition of compensatory pathways. In xenograft models, MCT4 had a significant impact on tumor metabolism and was required for rapid tumor growth. Together, these findings illustrate the metabolic diversity of PDA described by MCT4, delineate pathways through which this lactate transporter supports cancer growth, and demonstrate that PDA can be rationally targeted based on metabolic addictions.
Collapse
Affiliation(s)
- GuemHee Baek
- Department of Pathology, UT Southwestern, Dallas, TX 75390, USA
| | - Yan F Tse
- Department of Pathology, UT Southwestern, Dallas, TX 75390, USA
| | - Zeping Hu
- Children's Medical Center Research Institute at UT Southwestern, Dallas, TX 75390, USA
| | - Derek Cox
- Department of Pathology, UT Southwestern, Dallas, TX 75390, USA
| | - Noah Buboltz
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Peter McCue
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Charles J Yeo
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Michael A White
- Department of Cell Biology, UT Southwestern, Dallas, TX 75390, USA
| | - Ralph J DeBerardinis
- Simmons Cancer Center, UT Southwestern, Dallas, TX 75390, USA; Children's Medical Center Research Institute at UT Southwestern, Dallas, TX 75390, USA
| | - Erik S Knudsen
- Department of Pathology, UT Southwestern, Dallas, TX 75390, USA; Simmons Cancer Center, UT Southwestern, Dallas, TX 75390, USA
| | - Agnieszka K Witkiewicz
- Department of Pathology, UT Southwestern, Dallas, TX 75390, USA; Simmons Cancer Center, UT Southwestern, Dallas, TX 75390, USA.
| |
Collapse
|
32
|
Li S, Nguyen TT, Bonanno JA. CD147 required for corneal endothelial lactate transport. Invest Ophthalmol Vis Sci 2014; 55:4673-81. [PMID: 24970254 DOI: 10.1167/iovs.14-14386] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE CD147/basigin is a chaperone for lactate:H(+) cotransporters (monocarboxylate transporters) MCT1 and MCT4. We tested the hypothesis that MCT1 and -4 in corneal endothelium contribute to lactate efflux from stroma to anterior chamber and that silencing CD147 expression would cause corneal edema. METHODS CD147 was silenced via small interfering ribonucleic acid (siRNA) transfection of rabbit corneas ex vivo and anterior chamber lenti-small hairpin RNA (shRNA) pseudovirus in vivo. CD147 and MCT expression was examined by Western blot, RT-PCR, and immunofluorescence. Functional effects were examined by measuring lactate-induced cell acidification, corneal lactate efflux, [lactate], central cornea thickness (CCT), and Azopt (a carbonic anhydrase inhibitor) sensitivity. RESULTS In ex vivo corneas, 100 nM CD147 siRNA reduced CD147, MCT1, and MCT4 expression by 85%, 79%, and 73%, respectively, while MCT2 expression was unaffected. CD147 siRNA decreased lactate efflux from 3.9 ± 0.81 to 1.5 ± 0.37 nmol/min, increased corneal [lactate] from 19.28 ± 7.15 to 56.73 ± 8.97 nmol/mg, acidified endothelial cells (pHi = 6.83 ± 0.07 vs. 7.19 ± 0.09 in control), and slowed basolateral lactate-induced acidification from 0.0034 ± 0.0005 to 0.0012 ± 0.0005 pH/s, whereas apical acidification was unchanged. In vivo, CD147 shRNA increased CCT by 28.1 ± 0.9 μm at 28 days; Azopt increased CCT to 24.4 ± 3.12 vs. 12.0 ± 0.48 μm in control, and corneal [lactate] was 47.63 ± 6.29 nmol/mg in shCD147 corneas and 17.82 ± 4.93 nmol/mg in paired controls. CONCLUSIONS CD147 is required for the expression of MCT1 and MCT4 in the corneal endothelium. Silencing CD147 slows lactate efflux, resulting in stromal lactate accumulation and corneal edema, consistent with lactate efflux as a significant component of the corneal endothelial pump.
Collapse
Affiliation(s)
- Shimin Li
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Tracy T Nguyen
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| | - Joseph A Bonanno
- School of Optometry, Indiana University, Bloomington, Indiana, United States
| |
Collapse
|
33
|
Mariga ST, Kolko M, Gjedde A, Bergersen LH. Lactate transport and receptor actions in cerebral malaria. Front Neurosci 2014; 8:125. [PMID: 24904266 PMCID: PMC4032900 DOI: 10.3389/fnins.2014.00125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 04/22/2014] [Indexed: 01/09/2023] Open
Abstract
Cerebral malaria (CM), caused by Plasmodium falciparum infection, is a prevalent neurological disorder in the tropics. Most of the patients are children, typically with intractable seizures and high mortality. Current treatment is unsatisfactory. Understanding the pathogenesis of CM is required in order to identify therapeutic targets. Here, we argue that cerebral energy metabolic defects are probable etiological factors in CM pathogenesis, because malaria parasites consume large amounts of glucose metabolized mostly to lactate. Monocarboxylate transporters (MCTs) mediate facilitated transfer, which serves to equalize lactate concentrations across cell membranes in the direction of the concentration gradient. The equalizing action of MCTs is the basis for lactate’s role as a volume transmitter of metabolic signals in the brain. Lactate binds to the lactate receptor GPR81, recently discovered on brain cells and cerebral blood vessels, causing inhibition of adenylyl cyclase. High levels of lactate delivered by the parasite at the vascular endothelium may damage the blood–brain barrier, disrupt lactate homeostasis in the brain, and imply MCTs and the lactate receptor as novel therapeutic targets in CM.
Collapse
Affiliation(s)
- Shelton T Mariga
- Department of Neuroscience and Pharmacology, University of Copenhagen Copenhagen, Denmark
| | - Miriam Kolko
- Department of Neuroscience and Pharmacology, University of Copenhagen Copenhagen, Denmark ; Department of Ophthalmology, Roskilde Hospital Roskilde, Denmark
| | - Albert Gjedde
- Department of Neuroscience and Pharmacology, University of Copenhagen Copenhagen, Denmark
| | - Linda H Bergersen
- Department of Neuroscience and Pharmacology, University of Copenhagen Copenhagen, Denmark ; The Brain and Muscle Energy Group and SN-Lab, Department of Anatomy and Department of Oral Biology, Institute of Basic Medical Sciences and Centre for Molecular Biology and Neuroscience/SERTA Healthy Brain Aging Centre, University of Oslo Oslo, Norway
| |
Collapse
|
34
|
Adijanto J, Philp NJ. Cultured primary human fetal retinal pigment epithelium (hfRPE) as a model for evaluating RPE metabolism. Exp Eye Res 2014; 126:77-84. [PMID: 24485945 DOI: 10.1016/j.exer.2014.01.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 01/15/2014] [Accepted: 01/19/2014] [Indexed: 12/20/2022]
Abstract
Mitochondrial dysfunction has been shown to contribute to age-related and proliferative retinal diseases. Over the past decade, the primary human fetal RPE (hfRPE) culture model has emerged as an effective tool for studying RPE function and mechanisms of retinal diseases. This model system has been rigorously characterized and shown to closely resemble native RPE cells at the genomic and protein level, and that they are capable of accomplishing the characteristic functions of a healthy native RPE (e.g., rod phagocytosis, ion and fluid transport, and retinoid cycle). In this review, we demonstrated that the metabolic activity of the RPE is an indicator of its health and state of differentiation, and present the hfRPE culture model as a valuable in vitro system for evaluating RPE metabolism in the context of RPE differentiation and retinal disease.
Collapse
Affiliation(s)
- Jeffrey Adijanto
- Thomas Jefferson University, Dept. of Pathology, Anatomy, & Cell Biology, 1020 Locust Street, Rm315, Philadelphia, PA 19107, USA.
| | - Nancy J Philp
- Thomas Jefferson University, Dept. of Pathology, Anatomy, & Cell Biology, 1020 Locust Street, Rm315, Philadelphia, PA 19107, USA.
| |
Collapse
|
35
|
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: 41] [Impact Index Per Article: 3.4] [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
|
36
|
Rinholm JE, Bergersen LH. White matter lactate--does it matter? Neuroscience 2013; 276:109-16. [PMID: 24125892 DOI: 10.1016/j.neuroscience.2013.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/12/2013] [Accepted: 10/02/2013] [Indexed: 10/26/2022]
Abstract
About half of the human brain is white matter, characterized by axons covered in myelin, which facilitates the high speed of nerve signals from one brain area to another. At the time of myelination, the oligodendrocytes that synthesize myelin require a large amount of energy for this task. Conditions that deprive the tissue of energy can kill the oligodendrocytes. During brain development, the oligodendrocytes may use lactate as an alternative source of energy and material for myelin formation. Mature oligodendrocytes, however, can release lactate through the myelin sheath as nutrient for axons. In addition, lactate carries signals as a volume transmitter. Myelin thus seems to serve as a provider of substrates and signals for axons, and not as a mere insulator. We review the fluxes of lactate in white matter and their significance in brain function.
Collapse
Affiliation(s)
- J E Rinholm
- The Brain and Muscle Energy Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, PB1105 Blindern, N-0317 Oslo, Norway; Janelia Farm Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - L H Bergersen
- The Brain and Muscle Energy Group, Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, PB1105 Blindern, N-0317 Oslo, Norway; Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark; Center for Healthy Aging, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Oral Biology, University of Oslo, Norway.
| |
Collapse
|
37
|
Vijay N, Morris ME. Role of monocarboxylate transporters in drug delivery to the brain. Curr Pharm Des 2013; 20:1487-98. [PMID: 23789956 DOI: 10.2174/13816128113199990462] [Citation(s) in RCA: 286] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/18/2013] [Indexed: 02/08/2023]
Abstract
Monocarboxylate transporters (MCTs) are known to mediate the transport of short chain monocarboxylates such as lactate, pyruvate and butyrate. Currently, fourteen members of this transporter family have been identified by sequence homology, of which only the first four members (MCT1- MCT4) have been shown to mediate the proton-linked transport of monocarboxylates. Another transporter family involved in the transport of endogenous monocarboxylates is the sodium coupled MCTs (SMCTs). These act as a symporter and are dependent on a sodium gradient for their functional activity. MCT1 is the predominant transporter among the MCT isoforms and is present in almost all tissues including kidney, intestine, liver, heart, skeletal muscle and brain. The various isoforms differ in terms of their substrate specificity and tissue localization. Due to the expression of these transporters in the kidney, intestine, and brain, they may play an important role in influencing drug disposition. Apart from endogenous short chain monocarboxylates, they also mediate the transport of exogenous drugs such as salicylic acid, valproic acid, and simvastatin acid. The influence of MCTs on drug pharmacokinetics has been extensively studied for γ-hydroxybutyrate (GHB) including distribution of this drug of abuse into the brain and the results will be summarized in this review. The physiological role of these transporters in the brain and their specific cellular localization within the brain will also be discussed. This review will also focus on utilization of MCTs as potential targets for drug delivery into the brain including their role in the treatment of malignant brain tumors.
Collapse
|
38
|
|
39
|
Lapan SW, Reddien PW. Transcriptome analysis of the planarian eye identifies ovo as a specific regulator of eye regeneration. Cell Rep 2012; 2:294-307. [PMID: 22884275 DOI: 10.1016/j.celrep.2012.06.018] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/12/2012] [Accepted: 06/22/2012] [Indexed: 11/24/2022] Open
Abstract
Among the millions of invertebrate species with visual systems, the genetic basis of eye development and function is well understood only in Drosophila melanogaster. We describe an eye transcriptome for the planarian Schmidtea mediterranea. Planarian photoreceptors expressed orthologs of genes required for phototransduction and microvillus structure in Drosophila and vertebrates, and optic pigment cells expressed solute transporters and melanin synthesis enzymes similar to those active in the vertebrate retinal pigment epithelium. Orthologs of several planarian eye genes, such as bestrophin-1 and Usher syndrome genes, cause eye defects in mammals when perturbed and were not previously described to have roles in invertebrate eyes. Five previously undescribed planarian eye transcription factors were required for normal eye formation during head regeneration. In particular, a conserved, transcription-factor-encoding ovo gene was expressed from the earliest stages of eye regeneration and was required for regeneration of all cell types of the eye.
Collapse
Affiliation(s)
- Sylvain W Lapan
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | | |
Collapse
|
40
|
Pinheiro C, Longatto-Filho A, Azevedo-Silva J, Casal M, Schmitt FC, Baltazar F. Role of monocarboxylate transporters in human cancers: state of the art. J Bioenerg Biomembr 2012; 44:127-39. [PMID: 22407107 DOI: 10.1007/s10863-012-9428-1] [Citation(s) in RCA: 294] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Monocarboxylate transporters (MCTs) belong to the SLC16 gene family, presently composed by 14 members. MCT1-MCT4 are proton symporters, which mediate the transmembrane transport of pyruvate, lactate and ketone bodies. The role of MCTs in cell homeostasis has been characterized in detail in normal tissues, however, their role in cancer is still far from understood. Most solid tumors are known to rely on glycolysis for energy production and this activity leads to production of important amounts of lactate, which are exported into the extracellular milieu, contributing to the acidic microenvironment. In this context, MCTs will play a dual role in the maintenance of the hyper-glycolytic acid-resistant phenotype of cancer, allowing the maintenance of the high glycolytic rates by performing lactate efflux, and pH regulation by the co-transport of protons. Thus, they constitute attractive targets for cancer therapy, which have been little explored. Here we review the literature on the role of MCTs in solid tumors in different locations, such as colon, central nervous system, breast, lung, gynecologic tract, prostate, stomach, however, there are many conflicting results and in most cases there are no functional studies showing the dependence of the tumors on MCT expression and activity. Additional studies on MCT expression in other tumor types, confirmation of the results already published as well as additional functional studies are needed to deeply understand the role of MCTs in cancer maintenance and aggressiveness.
Collapse
Affiliation(s)
- Céline Pinheiro
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | | | | | | | | | | |
Collapse
|
41
|
Halestrap AP, Wilson MC. The monocarboxylate transporter family--role and regulation. IUBMB Life 2011; 64:109-19. [PMID: 22162139 DOI: 10.1002/iub.572] [Citation(s) in RCA: 530] [Impact Index Per Article: 37.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 08/08/2011] [Indexed: 11/07/2022]
Abstract
Monocarboxylate transporter (MCT) isoforms 1-4 catalyze the proton-linked transport of monocarboxylates such as L-lactate across the plasma membrane, whereas MCT8 and MCT10 are thyroid hormone and aromatic amino acid transporters, respectively. The importance of MCTs is becoming increasingly evident as their extensive physiological and pathological roles are revealed. MCTs 1-4 play essential metabolic roles in most tissues with their distinct properties, expression profile, and subcellular localization matching the particular metabolic needs of a tissue. Important metabolic roles include energy metabolism in the brain, skeletal muscle, heart, tumor cells, and T-lymphocyte activation, gluconeogenesis in the liver and kidney, spermatogenesis, bowel metabolism of short-chain fatty acids, and drug transport. MCT8 is essential for thyroid hormone transport across the blood-brain barrier. Genetic perturbation of MCT function may be involved in disease states such as pancreatic β-cell malfunction (inappropriate MCT1 expression), chronic fatigue syndromes (impairment of muscle MCT function), and psychomotor retardation (MCT8 mutation). MCT expression can be regulated at both the transcriptional and post-transcriptional levels. Of particular importance is the upregulation of muscle MCT1 expression in response to training and MCT4 expression in response to hypoxia. The latter is mediated by hypoxia inducible factor 1α and often observed in tumor cells that rely almost entirely on glycolysis for their energy provision. The recent discovery of potent and specific MCT1 inhibitors that prevent proliferation of T-lymphocytes confirms that MCTs may be promising pharmacological targets including for cancer chemotherapy.
Collapse
Affiliation(s)
- Andrew P Halestrap
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol, UK.
| | | |
Collapse
|
42
|
Halestrap AP. The monocarboxylate transporter family--Structure and functional characterization. IUBMB Life 2011; 64:1-9. [PMID: 22131303 DOI: 10.1002/iub.573] [Citation(s) in RCA: 509] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 08/08/2011] [Indexed: 11/11/2022]
Abstract
Monocarboxylate transporters (MCTs) catalyze the proton-linked transport of monocarboxylates such as L-lactate, pyruvate, and the ketone bodies across the plasma membrane. There are four isoforms, MCTs 1-4, which are known to perform this function in mammals, each with distinct substrate and inhibitor affinities. They are part of the larger SLC16 family of solute carriers, also known as the MCT family, which has 14 members in total, all sharing conserved sequence motifs. The family includes a high-affinity thyroid hormone transporter (MCT8), an aromatic amino acid transporter (T-type amino acid transporter 1/MCT10), and eight orphan members yet to be characterized. MCTs were predicted to have 12 transmembrane helices (TMs) with intracellular C- and N-termini and a large intracellular loop between TMs 6 and 7, and this was confirmed by labeling studies and proteolytic digestion. Site-directed mutagenesis has identified key residues required for catalysis and inhibitor binding and enabled the development of a molecular model of MCT1 in both inward and outward facing conformations. This suggests a likely mechanism for the translocation cycle. Although MCT family members are not themselves glycosylated, MCTs1-4 require association with a glycosylated ancillary protein, either basigin or embigin, for their correct translocation to the plasma membrane. These ancillary proteins have a single transmembrane domain and two to three extracellular immunoglobulin domains. They must remain closely associated with MCTs1-4 to maintain transporter activity. MCT1, MCT3, and MCT4 bind preferentially to basigin and MCT2 to embigin. The choice of binding partner does not affect substrate specificity or kinetics but can influence inhibitor specificity.
Collapse
Affiliation(s)
- Andrew P Halestrap
- School of Biochemistry, Medical Sciences Building, University of Bristol, Bristol, UK.
| |
Collapse
|
43
|
Lauritzen F, Perez EL, Melillo ER, Roh JM, Zaveri HP, Lee TSW, Wang Y, Bergersen LH, Eid T. Altered expression of brain monocarboxylate transporter 1 in models of temporal lobe epilepsy. Neurobiol Dis 2011; 45:165-76. [PMID: 21856423 DOI: 10.1016/j.nbd.2011.08.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 07/15/2011] [Accepted: 08/03/2011] [Indexed: 12/22/2022] Open
Abstract
Monocarboxylate transporter 1 (MCT1) facilitates the transport of monocarboxylate fuels (lactate, pyruvate and ketone bodies) and acidic drugs, such as valproic acid, across cell membranes. We recently reported that MCT1 is deficient on microvessels in the epileptogenic hippocampal formation in patients with medication-refractory temporal lobe epilepsy (TLE). To further define the role of MCT1 in the pathophysiology of TLE, we used immunohistochemistry and stereological analysis to localize and quantify the transporter in the hippocampal formation in three novel and highly relevant rat models of TLE and in nonepileptic control animals. One model utilizes methionine sulfoximine to induce brain glutamine synthetase deficiency and recurrent limbic seizures, while two models employ an episode of perforant pathway stimulation to cause epilepsy. MCT1 was lost on microvessels and upregulated on astrocytes in the hippocampal formation in all models of TLE. Notably, the loss of MCT1 on microvessels was not due to a reduction in microvessel density. The similarities in MCT1 expression among human subjects with TLE and several animal models of the disease strongly suggest a critical role of this molecule in the pathogenesis of TLE. We hypothesize that the downregulation of MCT1 may promote seizures via impaired uptake of ketone bodies and antiepileptic drugs by the epileptogenic brain. We also propose that the overexpression of MCT1 on astrocytes may lead to increased uptake or release of monocarboxylates by these cells, with important implications for brain metabolism and excitability. These hypotheses can now be rigorously tested in several animal models that replicate key features of human TLE.
Collapse
Affiliation(s)
- Fredrik Lauritzen
- Department of Laboratory Medicine, Yale University School of Medicine, P.O. Box 208035, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Monocarboxylate transporter 1 is deficient on microvessels in the human epileptogenic hippocampus. Neurobiol Dis 2010; 41:577-84. [PMID: 21081165 DOI: 10.1016/j.nbd.2010.11.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Revised: 11/01/2010] [Accepted: 11/09/2010] [Indexed: 11/20/2022] Open
Abstract
Monocarboxylate transporter 1 (MCT1) facilitates the transport of important metabolic fuels (lactate, pyruvate and ketone bodies) and possibly also acidic drugs such as valproic acid across the blood-brain barrier. Because an impaired brain energy metabolism and resistance to antiepileptic drugs are common features of temporal lobe epilepsy (TLE), we sought to study the expression of MCT1 in the brain of patients with this disease. Immunohistochemistry and immunogold electron microscopy were used to assess the distribution of MCT1 in brain specimens from patients with TLE and concomitant hippocampal sclerosis (referred to as mesial TLE or MTLE (n=15)), patients with TLE and no hippocampal sclerosis (non-MTLE, n=13) and neurologically normal autopsy subjects (n=8). MCT1 was present on an extensive network of microvessels throughout the hippocampal formation in autopsy controls and to a lesser degree in non-MTLE. Patients with MTLE were markedly deficient in MCT1 on microvessels in several areas of the hippocampal formation, especially CA1, which exhibited a 37% to 48% loss of MCT1 on the plasma membrane of endothelial cells when compared with non-MTLE. These findings suggest that the uptake of blood-derived monocarboxylate fuels and possibly also acidic drugs, such as valproic acid, is perturbed in the epileptogenic hippocampus, particularly in MTLE. We hypothesize that the loss of MCT1 on brain microvessels is mechanistically involved in the pathophysiology of drug-resistant TLE, and propose that re-expression of MCT1 may represent a novel therapeutic approach for this disease.
Collapse
|
45
|
Lange K. Fundamental role of microvilli in the main functions of differentiated cells: Outline of an universal regulating and signaling system at the cell periphery. J Cell Physiol 2010; 226:896-927. [PMID: 20607764 DOI: 10.1002/jcp.22302] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
46
|
The retinal pigment epithelium: something more than a constituent of the blood-retinal barrier--implications for the pathogenesis of diabetic retinopathy. J Biomed Biotechnol 2010; 2010:190724. [PMID: 20182540 PMCID: PMC2825554 DOI: 10.1155/2010/190724] [Citation(s) in RCA: 320] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 09/28/2009] [Accepted: 11/16/2009] [Indexed: 12/27/2022] Open
Abstract
The retinal pigment epithelium (RPE) is an specialized epithelium lying in the interface between the neural retina and the choriocapillaris where it forms the outer blood-retinal barrier (BRB). The main functions of the RPE are the following: (1) transport of nutrients, ions, and water, (2) absorption of light and protection against photooxidation, (3) reisomerization of all-trans-retinal into 11-cis-retinal, which is crucial for the visual cycle, (4) phagocytosis of shed photoreceptor membranes, and (5) secretion of essential factors for the structural integrity of the retina. An overview of these functions will be given. Most of the research on the physiopathology of diabetic retinopathy has been focused on the impairment of the neuroretina and the breakdown of the inner BRB. By contrast, the effects of diabetes on the RPE and in particular on its secretory activity have received less attention. In this regard, new therapeutic strategies addressed to modulating RPE impairment are warranted.
Collapse
|
47
|
Tea M, Fogarty R, Brereton HM, Michael MZ, Van der Hoek MB, Tsykin A, Coster DJ, Williams KA. Gene expression microarray analysis of early oxygen-induced retinopathy in the rat. J Ocul Biol Dis Infor 2009; 2:190-201. [PMID: 20157446 PMCID: PMC2821581 DOI: 10.1007/s12177-009-9041-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2009] [Accepted: 11/06/2009] [Indexed: 12/27/2022] Open
Abstract
Different inbred strains of rat differ in their susceptibility to oxygen-induced retinopathy (OIR), an animal model of human retinopathy of prematurity. We examined gene expression in Sprague-Dawley (susceptible) and Fischer 344 (resistant) neonatal rats after 3 days exposure to cyclic hyperoxia or room air, using Affymetrix rat Genearrays. False discovery rate analysis was used to identify differentially regulated genes. Such genes were then ranked by fold change and submitted to the online database, DAVID. The Sprague-Dawley list returned the term "response to hypoxia," absent from the Fischer 344 output. Manual analysis indicated that many genes known to be upregulated by hypoxia-inducible factor-1alpha were downregulated by cyclic hyperoxia. Quantitative real-time RT-PCR analysis of Egln3, Bnip3, Slc16a3, and Hk2 confirmed the microarray results. We conclude that combined methodologies are required for adequate dissection of the pathophysiology of strain susceptibility to OIR in the rat. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s12177-009-9041-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melinda Tea
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| | - Rhys Fogarty
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| | - Helen M. Brereton
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| | - Michael Z. Michael
- Department of Gastroenterology and Hepatology, Flinders University of South Australia, Adelaide, Australia
| | - Mark B. Van der Hoek
- Adelaide Microarray Centre, University of Adelaide & Hanson Institute, Adelaide, Australia
| | - Anna Tsykin
- Adelaide Microarray Centre, University of Adelaide & Hanson Institute, Adelaide, Australia
| | - Douglas J. Coster
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
| | - Keryn A. Williams
- Department of Ophthalmology, Flinders University of South Australia, Adelaide, Australia
- Department of Ophthalmology, Flinders Medical Centre, Bedford Park, 5042 SA Australia
| |
Collapse
|
48
|
Genetic ablation of retinal pigment epithelial cells reveals the adaptive response of the epithelium and impact on photoreceptors. Proc Natl Acad Sci U S A 2009; 106:18728-33. [PMID: 19850870 DOI: 10.1073/pnas.0902593106] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The retinal pigment epithelium (RPE) plays a critical role in the maintenance of the outer retina. RPE cell death or dysfunction drives the pathophysiology of many retinal diseases, but the physiological response of the retina to RPE cell loss is poorly understood, mainly because of the absence of suitable experimental models. Here, we generated a transgenic mouse in which an inducible Cre recombinase is expressed exclusively in the RPE under the control of the monocarboxylate transporter 3 gene promoter (RPE(CreER)). This was crossed with a transgenic mouse harboring a diphtheria toxin A (DTA) chain gene rendered transcriptionally silent by a floxed stop sequence. We show that activation of DTA in the double transgenic mouse (RPE(CreER)/DTA) led to 60-80% RPE cell death, with surviving cells maintaining the integrity of the monolayer by increasing their size. Despite the apparent morphological normality of the enlarged RPE cells in the RPE(CreER)/DTA mice, functional analysis revealed significant deficits on electroretinography, and retinal histopathology showed regions of photoreceptor rosetting and degeneration although with retention of a normal vascular network. Our study reveals that whilst the RPE monolayer has a remarkable intrinsic capacity to cope with cellular attrition, specific aspects of RPE multifunctionality essential for photoreceptor survival are compromised. The RPE(CreER)/DTA mouse offers advantages over models that employ chemical or mechanical strategies to kill RPE cells, and should be useful for the development and evaluation of RPE-based therapies, such as stem cell transplantation.
Collapse
|
49
|
Schober MS, Chidlow G, Wood JP, Casson RJ. Bioenergetic-based neuroprotection and glaucoma. Clin Exp Ophthalmol 2008; 36:377-85. [PMID: 18700928 DOI: 10.1111/j.1442-9071.2008.01740.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Primary open-angle glaucoma (POAG) is a pressure-sensitive optic neuropathy which results in the death of retinal ganglion cells and causes associated loss of vision. Presently, the only accepted treatment strategy is to lower the intraocular pressure; however, for some patients this is insufficient to prevent progressive disease. Although the pathogenesis of POAG remains unclear, there is considerable evidence that energy failure at the optic nerve head may be involved. Neuroprotection, a strategy which directly enhances the survival of neurons, is desirable, but remains clinically elusive. One particular form of neuroprotection involves the notion of enhancing the energy supply of neurons. These 'bioenergetic' methods of neuroprotection have proven successful in animal models of other neurodegenerative diseases and conditions, including Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and traumatic brain injury, but have been relatively unexplored in glaucoma models. This review focuses on some of the potential approaches for bioenergetic neuroprotection in the retina, including increasing the energy buffering capacity of damaged cells, decreasing the permeability of the mitochondrial membrane pore and free radical scavenging.
Collapse
Affiliation(s)
- Michael S Schober
- South Australian Institute of Ophthalmology, Adelaide, South Australia, Australia.
| | | | | | | |
Collapse
|
50
|
Trimarchi JM, Harpavat S, Billings NA, Cepko CL. Thyroid hormone components are expressed in three sequential waves during development of the chick retina. BMC DEVELOPMENTAL BIOLOGY 2008; 8:101. [PMID: 18854032 PMCID: PMC2579430 DOI: 10.1186/1471-213x-8-101] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 10/14/2008] [Indexed: 11/21/2022]
Abstract
BACKGROUND Thyroid hormone (TH) is an important developmental regulator in many tissues, including the retina. TH is activated locally via deiodinase 2 (Dio2), and it is destroyed by deiodinase 3 (Dio3). The TH receptors, TRa and TRb, mediate TH activity through hormone and DNA binding, and interactions with transcription regulators. RESULTS In the current work, the expression of these TH components was examined in the chick retina over time. Three waves of expression were characterized and found to be correlated with critical developmental events. The first wave occurred as progenitor cells began to make photoreceptors, the second as some cell types adopted a more mature location and differentiation state, and the third as Müller glia were generated. The cell types expressing the components, as well as the kinetics of expression within the cell cycle, were defined. TRb expression initiated during G2 in progenitor cells, concomitant with NeuroD and Otx2, which are expressed in early photoreceptor cells. TRb was expressed in photoreceptor cells for several days and then was reduced in expression level, as the expression of Crx, a later photoreceptor gene, became more evident. Dio3 was expressed throughout the cell cycle in progenitor cells. TRa was in most, if not all, retinal cells. Dio2 appeared transiently in a ventral (high) to dorsal gradient, likely in a subset of photoreceptor cells. CONCLUSION Multiple TH components were expressed in dynamic patterns in cycling progenitor cells and photoreceptors cells across the developing chick retina. These dynamic patterns suggest that TH is playing several roles in retinal development, both within the cycling progenitor cells and possibly with respect to the timing of differentiation of photoreceptor cells.
Collapse
Affiliation(s)
- Jeffrey M Trimarchi
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | | | - Nathan A Billings
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Constance L Cepko
- Department of Genetics and Howard Hughes Medical Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| |
Collapse
|