1
|
Kitaoka Y, Sase K. Molecular aspects of optic nerve autophagy in glaucoma. Mol Aspects Med 2023; 94:101217. [PMID: 37839231 DOI: 10.1016/j.mam.2023.101217] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/24/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023]
Abstract
The optic nerve consists of the glia, vessels, and axons including myelin and axoplasm. Since axonal degeneration precedes retinal ganglion cell death in glaucoma, the preceding axonal degeneration model may be helpful for understanding the molecular mechanisms of optic nerve degeneration. Optic nerve samples from these models can provide information on several aspects of autophagy. Autophagosomes, the most typical organelles expressing autophagy, are found much more frequently inside axons than around the glia. Thus, immunoblot findings from the optic nerve can reflect the autophagy state in axons. Autophagic flux impairment may occur in degenerating optic nerve axons, as in other central nervous system neurodegenerative diseases. Several molecular candidates are involved in autophagy enhancement, leading to axonal protection. This concept is an attractive approach to the prevention of further retinal ganglion cell death. In this review, we describe the factors affecting autophagy, including nicotinamide riboside, p38, ULK, AMPK, ROCK, and SIRT1, in the optic nerve and propose potential methods of axonal protection via enhancement of autophagy.
Collapse
Affiliation(s)
- Yasushi Kitaoka
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan; Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Kana Sase
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| |
Collapse
|
2
|
Sase K, Tsukahara C, Fujita N, Arizono I, Otsubo M, Kitaoka Y. Inhibition of p38 ameliorates axonal loss with modulation of autophagy in TNF-induced optic nerve damage. Int Ophthalmol 2023:10.1007/s10792-023-02706-1. [PMID: 37062014 PMCID: PMC10400678 DOI: 10.1007/s10792-023-02706-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 04/09/2023] [Indexed: 04/17/2023]
Abstract
PURPOSE A relationship between p38 and autophagy remains debated. The aim of the current study is to investigate whether an inhibitor of p38 prevents axon loss induced by TNF and whether it affects autophagy. METHODS Rats were given intravitreal injection of TNF, TNF plus SB203580, a p38 inhibitor, or SB203580 alone. Immunoblot analysis was performed to examine p62 expression which is a marker of autophagic flux and LC3-II expression which is an autophagy marker in optic nerves 1 week after intravitreal injection. Morphometric analysis of axons was performed to evaluate the effects of SB203580 against TNF-induced optic nerve damage 2 weeks after intravitreal injection. Immunohistochemical analysis was performed to evaluate the expressions of LC3, neurofilament, phosphorylated p38 and p62 in the optic nerve. RESULTS Quantification of axon number showed that TNF-induced axon loss was significantly protected by SB203580. Immunoblot analysis showed that the increase of p62 induced by TNF was totally eliminated by SB203580, and the SB203580 alone injection decreased the expression of p62. The level of LC3-II was significantly upregulated in the TNF plus SB203580 group compared with the TNF alone group, and the SB203580 alone injection increased the expression of LC3-II. Immunohistochemical analysis showed that LC3 immunoreactivity was found in the neurofilament positive fibers and that these immunoreactivities were enhanced by SB203580. Some colocalizations of p-p38 and p62 were observed in the TNF-treated optic nerve. CONCLUSION These results suggest that inhibition of p38 exerts axonal protection with upregulated autophagy in TNF-induced optic nerve damage.
Collapse
Affiliation(s)
- Kana Sase
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Chihiro Tsukahara
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Naoki Fujita
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
| | - Ibuki Arizono
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Mizuki Otsubo
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | - Yasushi Kitaoka
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| |
Collapse
|
3
|
MAPK Pathways in Ocular Pathophysiology: Potential Therapeutic Drugs and Challenges. Cells 2023; 12:cells12040617. [PMID: 36831285 PMCID: PMC9954064 DOI: 10.3390/cells12040617] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Mitogen-activated protein kinase (MAPK) pathways represent ubiquitous cellular signal transduction pathways that regulate all aspects of life and are frequently altered in disease. Once activated through phosphorylation, these MAPKs in turn phosphorylate and activate transcription factors present either in the cytoplasm or in the nucleus, leading to the expression of target genes and, as a consequence, they elicit various biological responses. The aim of this work is to provide a comprehensive review focusing on the roles of MAPK signaling pathways in ocular pathophysiology and the potential to influence these for the treatment of eye diseases. We summarize the current knowledge of identified MAPK-targeting compounds in the context of ocular diseases such as macular degeneration, cataract, glaucoma and keratopathy, but also in rare ocular diseases where the cell differentiation, proliferation or migration are defective. Potential therapeutic interventions are also discussed. Additionally, we discuss challenges in overcoming the reported eye toxicity of some MAPK inhibitors.
Collapse
|
4
|
Boccuni I, Fairless R. Retinal Glutamate Neurotransmission: From Physiology to Pathophysiological Mechanisms of Retinal Ganglion Cell Degeneration. Life (Basel) 2022; 12:638. [PMID: 35629305 PMCID: PMC9147752 DOI: 10.3390/life12050638] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Glutamate neurotransmission and metabolism are finely modulated by the retinal network, where the efficient processing of visual information is shaped by the differential distribution and composition of glutamate receptors and transporters. However, disturbances in glutamate homeostasis can result in glutamate excitotoxicity, a major initiating factor of common neurodegenerative diseases. Within the retina, glutamate excitotoxicity can impair visual transmission by initiating degeneration of neuronal populations, including retinal ganglion cells (RGCs). The vulnerability of RGCs is observed not just as a result of retinal diseases but has also been ascribed to other common neurodegenerative and peripheral diseases. In this review, we describe the vulnerability of RGCs to glutamate excitotoxicity and the contribution of different glutamate receptors and transporters to this. In particular, we focus on the N-methyl-d-aspartate (NMDA) receptor as the major effector of glutamate-induced mechanisms of neurodegeneration, including impairment of calcium homeostasis, changes in gene expression and signalling, and mitochondrial dysfunction, as well as the role of endoplasmic reticular stress. Due to recent developments in the search for modulators of NMDA receptor signalling, novel neuroprotective strategies may be on the horizon.
Collapse
Affiliation(s)
- Isabella Boccuni
- Institute for Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
5
|
Lambuk L, Iezhitsa I, Agarwal R, Agarwal P, Peresypkina A, Pobeda A, Ismail NM. Magnesium acetyltaurate prevents retinal damage and visual impairment in rats through suppression of NMDA-induced upregulation of NF-κB, p53 and AP-1 (c-Jun/c-Fos). Neural Regen Res 2021; 16:2330-2344. [PMID: 33818520 PMCID: PMC8354133 DOI: 10.4103/1673-5374.310691] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/01/2020] [Accepted: 12/02/2020] [Indexed: 12/22/2022] Open
Abstract
Magnesium acetyltaurate (MgAT) has been shown to have a protective effect against N-methyl-D-aspartate (NMDA)-induced retinal cell apoptosis. The current study investigated the involvement of nuclear factor kappa-B (NF-κB), p53 and AP-1 family members (c-Jun/c-Fos) in neuroprotection by MgAT against NMDA-induced retinal damage. In this study, Sprague-Dawley rats were randomized to undergo intravitreal injection of vehicle, NMDA or MgAT as pre-treatment to NMDA. Seven days after injections, retinal ganglion cells survival was detected using retrograde labelling with fluorogold and BRN3A immunostaining. Functional outcome of retinal damage was assessed using electroretinography, and the mechanisms underlying antiapoptotic effect of MgAT were investigated through assessment of retinal gene expression of NF-κB, p53 and AP-1 family members (c-Jun/c-Fos) using reverse transcription-polymerase chain reaction. Retinal phospho-NF-κB, phospho-p53 and AP-1 levels were evaluated using western blot assay. Rat visual functions were evaluated using visual object recognition tests. Both retrograde labelling and BRN3A immunostaining revealed a significant increase in the number of retinal ganglion cells in rats receiving intravitreal injection of MgAT compared with the rats receiving intravitreal injection of NMDA. Electroretinography indicated that pre-treatment with MgAT partially preserved the functional activity of NMDA-exposed retinas. MgAT abolished NMDA-induced increase of retinal phospho-NF-κB, phospho-p53 and AP-1 expression and suppressed NMDA-induced transcriptional activity of NF-κB, p53 and AP-1 family members (c-Jun/c-Fos). Visual object recognition tests showed that MgAT reduced difficulties in recognizing the visual cues (i.e. objects with different shapes) after NMDA exposure, suggesting that visual functions of rats were relatively preserved by pre-treatment with MgAT. In conclusion, pre-treatment with MgAT prevents NMDA induced retinal injury by inhibiting NMDA-induced neuronal apoptosis via downregulation of transcriptional activity of NF-κB, p53 and AP-1-mediated c-Jun/c-Fos. The experiments were approved by the Animal Ethics Committee of Universiti Teknologi MARA (UiTM), Malaysia, UiTM CARE No 118/2015 on December 4, 2015 and UiTM CARE No 220/7/2017 on December 8, 2017 and Ethics Committee of Belgorod State National Research University, Russia, No 02/20 on January 10, 2020.
Collapse
Affiliation(s)
- Lidawani Lambuk
- Center for Neuroscience Research (NeuRon), Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Igor Iezhitsa
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Renu Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Puneet Agarwal
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Anna Peresypkina
- Department of Pharmacology and Clinical Pharmacology, Institute of Medicine, Belgorod State National Research University, Belgorod, Russia
| | - Anna Pobeda
- Department of Pharmacology and Clinical Pharmacology, Institute of Medicine, Belgorod State National Research University, Belgorod, Russia
| | - Nafeeza Mohd Ismail
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Fujita N, Sase K, Tsukahara C, Arizono I, Takagi H, Kitaoka Y. Pemafibrate prevents retinal neuronal cell death in NMDA-induced excitotoxicity via inhibition of p-c-Jun expression. Mol Biol Rep 2020; 48:195-202. [PMID: 33278012 PMCID: PMC7884588 DOI: 10.1007/s11033-020-06032-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023]
Abstract
Excitotoxicity is involved in the retinal neuronal cell death in diabetic retinopathy. Although fenofibrate has been shown to ameliorate the progression of diabetic retinopathy, the effect of pemafibrate, which is highly selective for peroxisome proliferator-activated receptor α on retinal neuronal cell death has not been documented. Here, we investigated whether pemafibrate exerts a beneficial effect against retinal ganglion cell (RGC) death induced by N-methyl-D-aspartate (NMDA) in rats. Experiments were performed on adult male Wistar rats that received an intravitreal injection of 20 nmol NMDA. Fluoro-Gold labeled RGC morphometry showed that oral intake of pemafibrate once a day for 7 days resulted in significant protection on RGC death induced by NMDA. Phosphorylated c-Jun protein, which is involved in apoptosis, was upregulated after NMDA exposure, and this increase was significantly lessened by the systemic pemafibrate treatment. Phosphorylated c-Jun immunopositive cells were colocalized with Thy-1 immunopositive cells, and the increased these cells were ameliorated by the pemafibrate treatment. An increase in TUNEL-positive cells was significantly suppressed by the pemafibrate treatment. Phosphorylated c-Jun immunopositive cells were colocalized with TUNEL-positive cells, and they were decreased by pemafibrate treatment. These results suggest that the RGC protection achieved with pemafibrate appears to be associated with inhibition of phosphorylated c-Jun and its anti-apoptotic effect.
Collapse
Affiliation(s)
- Naoki Fujita
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan.,Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan
| | - Kana Sase
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan
| | - Chihiro Tsukahara
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan
| | - Ibuki Arizono
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan.,Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan
| | - Hitoshi Takagi
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan
| | - Yasushi Kitaoka
- Department of Molecular Neuroscience, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kaswasaki, Kanagawa, 216-8511, Japan.
| |
Collapse
|
7
|
Akebia Saponin D prevents axonal loss against TNF-induced optic nerve damage with autophagy modulation. Mol Biol Rep 2020; 47:9733-9738. [PMID: 33249542 PMCID: PMC7723935 DOI: 10.1007/s11033-020-06008-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/16/2020] [Indexed: 10/30/2022]
Abstract
Akebia Saponin D (ASD), a triterpenoid saponin, was shown to have protective effects in certain neuronal cells. The purpose of the present study was to investigate the possibility of ASD to prevent tumor necrosis factor (TNF)-induced axonal loss and the ASD modulation of the biologic process of autophagy in optic nerves. Rats were given intravitreal administration of TNF, simultaneous administration of 2, 20, or 200 pmol ASD and TNF, or ASD alone. LC3-II and p62 expression, which is a marker of autophagic flux, and phosphorylated p38 (p-p38) expression in optic nerves were examined by immunoblot analysis. Morphometric analysis revealed a significant ameliorated effect of ASD against TNF-induced optic nerve damage. p62 was significantly increased in the optic nerve in TNF-treated eyes, but this increase was totally prevented by ASD. The ASD alone injection showed significant reduction of p62 levels compared with the PBS-treated control eyes. LC3-II was significantly increased by ASD treatment in the TNF-injected eyes. p-p38 was significantly increased in the optic nerve in TNF-treated eyes, but this increase was completely prevented by ASD. The protective effects of ASD may be associated with enhanced autophagy activation and inhibition of p-p38.
Collapse
|
8
|
Chen W, Li Z, Wang Q, Wang Y, Zhang Y. The Role of C-Jun N-terminal Kinase-1 in Controlling Aquaporin-1 and Choroidal Thickness during Recovery from Form-deprivation Myopia in Guinea Pigs. Curr Eye Res 2020; 46:885-894. [PMID: 33115288 DOI: 10.1080/02713683.2020.1839107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE The development and recovery(REC) of myopia is associated with changing of choroidal thickness(CT) in model of guinea pigs. Aquaporin-1 (AQP-1) is related to the changes of CT during the recovery from myopia, but the corresponding signaling pathway has not been clarified. This study aimed to investigate the effect of JNK1 on CT/AQP-1 and the recovery from myopia. MATERIALS AND METHODS According to the different single intravitreal injections in eyes that underwent form deprivation for 21 days, guinea pigs were divided into four groups: the REC group, the REC+anisomycin (REC-AN, agonist for JNK1, 0.2 nmol) group, the REC+SP600125 (REC-SP, inhibitor for JNK1, 0.2 nmol) group, and the REC+dimethyl sulfoxide (REC-DM) group. Each group was divided into three subgroups based on the duration of the form deprivation: 3 days (d), 7 d and 10 d. All animals underwent biometric measurements (refractive error, axial length (AL), and CT), and the protein expression of AQP-1 and p-JNK1 in the choroid was also measured. RESULTS In REC and REC-DM groups, significant differences in CT/refractive error/AL/p-JNK1 or AQP-1 were only found in the 3d group compared with normal control (NC) group (all p < .05). In REC-AN group, CT/p-JNK1 or AQP-1 in 3d group was significantly higher than that in other 3d groups (all p < .05), but no significant difference in refractive error or AL was found compared with NC group at three time points (all p > .05). In REC-SP group, a significant difference in refractive error/CT/p-JNK1 or AQP1 was found in 3d/7d group compared with NC group (all p < .05), but AL was only found in 3d groups (p = .001). CONCLUSIONS Changes in JNK1 phosphorylation can regulate AQP-1 and CT during the recovery from myopia and the recovery time. Thus, JNK1 may be a potential therapeutic target for preventing/treating myopia.
Collapse
Affiliation(s)
- Wei Chen
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, China
| | - Zhiwei Li
- Department of Ophthalmology, Shandong Provincial Hospital Affilliated to Shandong University, Jinan, China
| | - Qimiao Wang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, China
| | - Yan Wang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, China
| | - Yue Zhang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Clinical College of Ophthalmology, Tianjin Medical University, Tianjin Eye Institute, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin, China
| |
Collapse
|
9
|
Munemasa Y. Histone H2B induces retinal ganglion cell death through toll-like receptor 4 in the vitreous of acute primary angle closure patients. J Transl Med 2020; 100:1080-1089. [PMID: 32321985 PMCID: PMC7374083 DOI: 10.1038/s41374-020-0427-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 03/29/2020] [Accepted: 03/29/2020] [Indexed: 11/14/2022] Open
Abstract
Acute primary angle closure (APAC) is a disease of ophthalmic urgency; lack of treatment can lead to blindness. Even after adequate treatment for APAC, subsequent elevated acute intraocular pressure induces severe neuronal damage which can result in secondary glaucomatous optic neuropathy (GON). Damage-associated molecular patterns (DAMPs) are released from damaged and dead neuronal cells, which induce secondary inflammatory changes and further tissue damage. Our hypothesis is that histone H2B (H2B), which is one of the DAMPs, is released from damaged cells in the development of GON after APAC treatment. Intravitreal injection of H2B induces neuronal cell death through toll-like receptor 4 (TLR4) expression, following the upregulation of inflammatory cytokine mRNAs and phosphorylation of mitogen activated protein kinases (MAPKs). Knockdown of TLR4 caused a reduction of H2B neurotoxicity in damaged cells through TLR4 signaling. Significantly increased H2B was observed in the vitreous cells of APAC patients. In addition, enhanced H2B protein correlated with decreased ganglion cell analysis and retinal ganglion cell (RGC) layer thinning, which indicates the effect of H2B on RGCs. Our data from clinical and animal studies show the involvement of H2B-TLR4 pathways in the development of GON after APAC treatment providing new insight for the mechanism of RGC degeneration.
Collapse
Affiliation(s)
- Yasunari Munemasa
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao Miyamae, Kawasaki, Kanagawa, 2168511, Japan.
| |
Collapse
|
10
|
Pérez de Lara MJ, Avilés-Trigueros M, Guzmán-Aránguez A, Valiente-Soriano FJ, de la Villa P, Vidal-Sanz M, Pintor J. Potential role of P2X7 receptor in neurodegenerative processes in a murine model of glaucoma. Brain Res Bull 2019; 150:61-74. [PMID: 31102752 DOI: 10.1016/j.brainresbull.2019.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 04/23/2019] [Accepted: 05/10/2019] [Indexed: 12/20/2022]
Abstract
Glaucoma is a common cause of visual impairment and blindness, characterized by retinal ganglion cell (RGC) death. The mechanisms that trigger the development of glaucoma remain unknown and have gained significant relevance in the study of this neurodegenerative disease. P2X7 purinergic receptors (P2X7R) could be involved in the regulation of the synaptic transmission and neuronal death in the retina through different pathways. The aim of this study was to characterize the molecular signals underlying glaucomatous retinal injury. The time-course of functional, morphological, and molecular changes in the glaucomatous retina of the DBA/2J mice were investigated. The expression and localization of P2X7R was analysed in relation with retinal markers. Caspase-3, JNK, and p38 were evaluated in control and glaucomatous mice by immunohistochemical and western-blot analysis. Furthermore, electroretinogram recordings (ERG) were performed to assess inner retina dysfunction. Glaucomatous mice exhibited changes in P2X7R expression as long as the pathology progressed. There was P2X7R overexpression in RGCs, the primary injured neurons, which correlated with the loss of function through ERG measurements. All analyzed MAPK and caspase-3 proteins were upregulated in the DBA/2J retinas suggesting a pro-apoptotic cell death. The increase in P2X7Rs presence may contribute, together with other factors, to the changes in retinal functionality and the concomitant death of RGCs. These findings provide evidence of possible intracellular pathways responsible for apoptosis regulation during glaucomatous degeneration.
Collapse
Affiliation(s)
- María J Pérez de Lara
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037, Madrid, Spain
| | - Marcelino Avilés-Trigueros
- Laboratory of Experimental Ophthalmology, Dept. of Ophthalmology, Faculty of Medicine, University of Murcia and Murcia Institute of Bio-Health Research (IMIB), E-30120, El Palmar, Murcia, Spain
| | - Ana Guzmán-Aránguez
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037, Madrid, Spain
| | - F Javier Valiente-Soriano
- Laboratory of Experimental Ophthalmology, Dept. of Ophthalmology, Faculty of Medicine, University of Murcia and Murcia Institute of Bio-Health Research (IMIB), E-30120, El Palmar, Murcia, Spain
| | - Pedro de la Villa
- Systems Biology Department, Faculty of Medicine, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Manuel Vidal-Sanz
- Laboratory of Experimental Ophthalmology, Dept. of Ophthalmology, Faculty of Medicine, University of Murcia and Murcia Institute of Bio-Health Research (IMIB), E-30120, El Palmar, Murcia, Spain.
| | - Jesús Pintor
- Department of Biochemistry and Molecular Biology IV, Faculty of Optics and Optometry, Complutense University of Madrid, c/Arcos de Jalón 118, E-28037, Madrid, Spain
| |
Collapse
|
11
|
N-Methyl- N-Nitrosourea-Induced Photoreceptor Degeneration Is Inhibited by Nicotinamide via the Blockade of Upstream Events before the Phosphorylation of Signalling Proteins. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3238719. [PMID: 31179317 PMCID: PMC6507250 DOI: 10.1155/2019/3238719] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 01/12/2023]
Abstract
N-methyl-N-nitrosourea (MNU), a known carcinogen, is generally used in animal models to chemically induce photoreceptor degeneration. It has been reported that nicotinamide (NAM) exerts a protective effect on MNU-induced photoreceptor degeneration. We investigated the molecular mechanisms on MNU-induced photoreceptor degeneration. Intraperitoneal MNU injection (75 mg/kg) in rats induced selective photoreceptor degeneration in 7 days. NAM administration completely inhibited photoreceptor degeneration. Photoreceptor layer abnormality was observed within 6 hours after MNU injection, whereas it was restored in the NAM-treated retina, as detected by optical coherence tomography. One day following MNU administration, phosphorylation of the cell death-associated signalling proteins c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (p38) increased, while the apoptosis-related proteins, full-length poly(ADP-ribose) polymerase (PARP) and apoptosis-inducing factor (AIF), were depleted. These changes were not observed in the NAM-treated retinas. Cell survival signalling, such as extracellular signal-regulated kinase (ERK), Akt, and cAMP response element binding protein (CREB) phosphorylation, increased in the MNU- but not in the NAM-treated rat retinas. Increased phosphorylated ERK (p-ERK) levels were observed within 6 hours after MNU administration, suggestive of cell survival signalling activation. This did not occur in NAM-treated retinas. These results indicate that NAM regulates upstream cellular events prior to the activation of cell death-related signalling events, such as JNK and p38 phosphorylation.
Collapse
|
12
|
Mammone T, Chidlow G, Casson RJ, Wood JPM. Expression and activation of mitogen-activated protein kinases in the optic nerve head in a rat model of ocular hypertension. Mol Cell Neurosci 2018; 88:270-291. [PMID: 29408550 DOI: 10.1016/j.mcn.2018.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 01/03/2018] [Accepted: 01/11/2018] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Glaucoma is a leading cause of irreversible blindness manifesting as an age-related, progressive optic neuropathy with associated retinal ganglion cell (RGC) loss. Mitogen-activated protein kinases (MAPKs: p42/44 MAPK, SAPK/JNK, p38 MAPK) are activated in various retinal disease models and likely contribute to the mechanisms of RGC death. Although MAPKs play roles in the development of retinal pathology, their action in the optic nerve head (ONH), where the initial insult to RGC axons likely resides in glaucoma, remains unexplored. METHODS An experimental paradigm representing glaucoma was established by induction of chronic ocular hypertension (OHT) via laser-induced coagulation of the trabecular meshwork in Sprague-Dawley rats. MAPKs were subsequently investigated over the following days for expression and activity alterations, using RT-PCR, immunohistochemistry and Western immunoblot. RESULTS p42/44 MAPK expression was unaltered after intraocular pressure (IOP) elevation, but there was a significant activation of this enzyme in ONH astrocytes after 6-24 h. Activated SAPK/JNK isoforms were present throughout healthy RGC axons but after IOP elevation or optic nerve crush, they both accumulated at the ONH, likely due to RGC axon transport disruption, and were subject to additional activation. p38 MAPK was expressed by a population of microglia which were significantly more populous following IOP elevation. However it was only significantly activated in microglia after 3 days, and then only in the ONH and optic nerve; in the retina it was solely activated in RGC perikarya. CONCLUSIONS In conclusion, each of the MAPKs showed a specific spatio-temporal expression and activation pattern in the retina, ONH and optic nerve as a result of IOP elevation. These findings likely reflect the roles of the individual enzymes, and the cells in which they reside, in the developing pathology following IOP elevation. These data have implications for understanding the mechanisms of ocular pathology in diseases such as glaucoma.
Collapse
Affiliation(s)
- Teresa Mammone
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - Glyn Chidlow
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - Robert J Casson
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| | - John P M Wood
- Ophthalmic Research Laboratories, Central Adelaide Local Health Network, Level 7 Adelaide Health & Medical Sciences Building, University of Adelaide, Adelaide, South Australia, Australia; Department of Ophthalmology and Visual Sciences, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
13
|
Fahrenthold BK, Fernandes KA, Libby RT. Assessment of intrinsic and extrinsic signaling pathway in excitotoxic retinal ganglion cell death. Sci Rep 2018; 8:4641. [PMID: 29545615 PMCID: PMC5854579 DOI: 10.1038/s41598-018-22848-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/01/2018] [Indexed: 01/15/2023] Open
Abstract
Excitotoxicity leads to the activation of a cytotoxic cascade that causes neuronal death. In the retina, retinal ganglion cells (RGCs) die after an excitotoxic insult. Multiple pathways have been proposed to contribute to RGC death after an excitotoxic insult, including TNF signaling, JNK activation, and ER stress. To test the importance of these pathways in RGC death after excitotoxic injury, the excitotoxin N-methyl-D-aspartate (NMDA) was intravitreally injected into mice deficient in components of these pathways. Absence of Tnf or its canonical downstream mediator, Bid, did not confer short- or long-term protection to RGCs. Despite known activation in RGCs and a prominent role in mediating RGC death after other insults, attenuating JNK signaling did not prevent RGC death after excitotoxic insult. Additionally, deficiency of the ER stress protein DDIT3 (CHOP), which has been shown to be involved in RGC death, did not lessen NMDA induced RGC death. Furthermore, absence of both Jun (JNK’s canonical target) and Ddit3, which together provide robust, long-term protection to RGC somas after axonal insult, did not lessen RGC death. Collectively, these results indicate that the drivers of excitotoxic injury remain to be identified and/or multiple cell death pathways are activated in response to injury.
Collapse
Affiliation(s)
- Berkeley K Fahrenthold
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, 14642, USA.,Neuroscience Graduate Program, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Kimberly A Fernandes
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Richard T Libby
- Department of Ophthalmology, University of Rochester Medical Center, Rochester, NY, 14642, USA. .,Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, 14642, USA. .,The Center for Visual Sciences, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| |
Collapse
|
14
|
Kim BJ, Silverman SM, Liu Y, Wordinger RJ, Pang IH, Clark AF. In vitro and in vivo neuroprotective effects of cJun N-terminal kinase inhibitors on retinal ganglion cells. Mol Neurodegener 2016; 11:30. [PMID: 27098079 PMCID: PMC4839164 DOI: 10.1186/s13024-016-0093-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 04/08/2016] [Indexed: 01/24/2023] Open
Abstract
Background The c-Jun N-terminal kinase (JNK) signaling pathway plays an important role in neuronal pathophysiology. Using JNK inhibitors, we examined involvement of the JNK pathway in cultured rat retinal ganglion cell (RGC) death and in mouse retinal ischemia/reperfusion (I/R) injury of the visual axis. The in vitro effects of JNK inhibitors were evaluated in cultured adult rat retinal cells enriched in RGCs. Retinal I/R was induced in C57BL/6J mice through elevation of intraocular pressure to 120 mmHg for 60 min followed by reperfusion. SP600125 was administered intraperitoneally once daily for 28 days. Phosphorylation of JNK and c-Jun in the retina was examined by immunoblotting and immunohistochemistry. The thickness of retinal layers and cell numbers in the ganglion cell layer (GCL) were examined using H&E stained retinal cross sections and spectral domain optical coherence tomography (SD-OCT). Retinal function was measured by scotopic flash electroretinography (ERG). Volumetric measurement of the superior colliculus (SC) as well as VGLUT2 and PSD95 expression were studied. Results JNK inhibitors SP600125 and TAT-JNK-III, dose-dependently and significantly (p < 0.05) protected against glutamate excitotoxicity and trophic factor withdrawal induced RGC death in culture. In the I/R model, phosphorylation of JNK (pJNK) in the retina was significantly (p < 0.05) increased after injury. I/R injury significantly (p < 0.05) decreased the thickness of retinal layers, including the whole retina, inner plexiform layer, and inner nuclear layer and cell numbers in the GCL. Administration of SP600125 for 28 days protected against all these degenerative morphological changes (p < 0.05). In addition, SP600125 significantly (p < 0.05) protected against I/R-induced reduction in scotopic ERG b-wave amplitude at 3, 7, 14, 21 and 28 days after injury. SP600125 also protected against the I/R-induced losses in volume and levels of synaptic markers in the SC. Moreover, the protective effects of SP600125 in the retina and SC were also detected even with only 7 days (Days 1–7 after I/R) of SP600125 treatment. Conclusions Our results demonstrate the important role the JNK pathway plays in retinal degeneration in both in vitro and in vivo models and suggest that JNK inhibitors may be a useful therapeutic strategy for neuroprotection of RGCs in the retina. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0093-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Byung-Jin Kim
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.,Present Address: Department of Ophthalmology, Johns Hopkins University School of Medicine, 400 N. Broadway, Baltimore, MD, 21231, USA
| | - Sean M Silverman
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Yang Liu
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.,Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Robert J Wordinger
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Iok-Hou Pang
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA.,Department of Pharmaceutical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | - Abbot F Clark
- North Texas Eye Research Institute, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76109, USA. .,Department of Cell Biology & Immunology, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA.
| |
Collapse
|
15
|
Levkovitch-Verbin H. Retinal ganglion cell apoptotic pathway in glaucoma: Initiating and downstream mechanisms. PROGRESS IN BRAIN RESEARCH 2015; 220:37-57. [PMID: 26497784 DOI: 10.1016/bs.pbr.2015.05.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Apoptosis of retinal ganglion cells (RGCs) in glaucoma causes progressive visual field loss, making it the primary cause of irreversible blindness worldwide. Elevated intraocular pressure and aging, the main risk factors for glaucoma, accelerate RGC apoptosis. Numerous pathways and mechanisms were found to be involved in RGC death in glaucoma. Neurotrophic factors deprivation is an early event. Oxidative stress, mitochondrial dysfunction, inflammation, glial cell dysfunction, and activation of apoptotic pathways and prosurvival pathways play a significant role in RGC death in glaucoma. The most important among the involved pathways are the MAP-kinase pathway, PI-3 kinase/Akt pathway, Bcl-2 family, caspase family, and IAP family.
Collapse
Affiliation(s)
- Hani Levkovitch-Verbin
- Glaucoma Service, Goldschleger Eye Institute, Sheba Medical Center, Tel-Hashomer, Israel; Sackler Faculty of Medicine, Tel-Aviv University, Tel-Hashomer, Israel.
| |
Collapse
|
16
|
Han N, Yu L, Song Z, Luo L, Wu Y. Agmatine protects Müller cells from high-concentration glucose-induced cell damage via N-methyl-D-aspartic acid receptor inhibition. Mol Med Rep 2015; 12:1098-106. [PMID: 25816073 PMCID: PMC4438955 DOI: 10.3892/mmr.2015.3540] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 03/09/2015] [Indexed: 01/14/2023] Open
Abstract
Neural injury is associated with the development of diabetic retinopathy. Müller cells provide structural and metabolic support for retinal neurons. High glucose concentrations are known to induce Müller cell activity. Agmatine is an endogenous polyamine, which is enzymatically formed in the mammalian brain and has exhibited neuroprotective effects in a number of experimental models. The aims of the present study were to investigate whether agmatine protects Müller cells from glucose-induced damage and to explore the mechanisms underlying this process. Lactate dehydrogenase activity and tumor necrosis factor-α mRNA expression were significantly reduced in Müller cells exposed to a high glucose concentration, following agmatine treatment, compared with cells not treated with agmatine. In addition, agmatine treatment inhibited glucose-induced Müller cell apoptosis, which was associated with the regulation of Bax and Bcl-2 expression. Agmatine treatment suppressed glucose-induced phosphorylation of mitogen-activated protein kinase (MAPK) protein in Müller cells. The present study demonstrated that the protective effects of agmatine on Müller cells were inhibited by N-methyl-D-aspartic acid (NMDA). The results of the present study suggested that agmatine treatment protects Müller cells from high-concentration glucose-induced cell damage. The underlying mechanisms may relate to the anti-inflammatory and antiapoptotic effects of agmatine, as well as to the inhibition of the MAPK pathway, via NMDA receptor suppression. Agmatine may be of use in the development of novel therapeutic approaches for patients with diabetic retinopathy.
Collapse
Affiliation(s)
- Ning Han
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Li Yu
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhidu Song
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Lifu Luo
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - Yazhen Wu
- Department of Ocular Fundus Disease, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
17
|
Yang L, Wu L, Wang D, Li Y, Dou H, Tso MOM, Ma Z. Role of endoplasmic reticulum stress in the loss of retinal ganglion cells in diabetic retinopathy. Neural Regen Res 2014; 8:3148-58. [PMID: 25206636 PMCID: PMC4158709 DOI: 10.3969/j.issn.1673-5374.2013.33.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/21/2013] [Indexed: 01/26/2023] Open
Abstract
Endoplasmic reticulum stress is closely involved in the early stage of diabetic retinopathy. In the present study, a streptozotocin-induced diabetic animal model was given an intraperitoneal injection of tauroursodeoxycholic acid. Results from immunofluorescent co-localization experiments showed that both caspase-12 protein and c-Jun N-terminal kinase 1 phosphorylation levels significantly in-creased, which was associated with retinal ganglion cell death in diabetic retinas. The C/ERB mologous protein pathway directly contributed to glial reactivity, and was subsequently responsible for neuronal loss and vascular abnormalities in diabetic retinopathy. Our experimental findings dicate that endoplasmic reticulum stress plays an important role in diabetes-induced retinal neu-ronal loss and vascular abnormalities, and that inhibiting the activation of the endoplasmic reticulum stress pathway provides effective protection against diabetic retinopathy.
Collapse
Affiliation(s)
- Liping Yang
- Department of Ophthalmology, Peking University Third Hospital, Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| | - Lemeng Wu
- Department of Ophthalmology, Peking University Third Hospital, Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| | - Dongmei Wang
- Department of Ophthalmology, Peking University Third Hospital, Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| | - Ying Li
- Department of Ophthalmology, Peking University Third Hospital, Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| | - Hongliang Dou
- Department of Ophthalmology, Peking University Third Hospital, Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| | - Mark O M Tso
- Department of Ophthalmology, Peking University Third Hospital, Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China ; Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Zhizhong Ma
- Department of Ophthalmology, Peking University Third Hospital, Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing 100191, China
| |
Collapse
|
18
|
Ichikawa A, Nakahara T, Kurauchi Y, Mori A, Sakamoto K, Ishii K. Rapamycin prevents N-methyl-D-aspartate-induced retinal damage through an ERK-dependent mechanism in rats. J Neurosci Res 2014; 92:692-702. [PMID: 24510654 DOI: 10.1002/jnr.23358] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 12/04/2013] [Accepted: 12/04/2013] [Indexed: 12/30/2022]
Abstract
Recent studies have demonstrated that inhibition of the mammalian target of rapamycin (mTOR) protects against neuronal injury, but the mechanisms underlying this protection are not fully understood. The present study investigates whether rapamycin, an inhibitor of the mTOR pathway, protects against N-methyl-D-aspartate (NMDA)-induced retinal neurotoxicity and whether the extracellular signal-regulated kinase (ERK) pathway contributes to this protective effect in rats. Significant cell loss in the ganglion cell layer and a reduction in thickness of the inner plexiform layer were observed 7 days after a single intravitreal injection of NMDA (200 nmol/eye). These NMDA-induced morphological changes were significantly reduced by rapamycin (20 nmol/eye). The number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive apoptotic cells had increased 6 hr after NMDA injection, an effect that was significantly attenuated by rapamycin. The ERK inhibitor U0126 (1 nmol/eye) almost completely abolished rapamycin's inhibition of NMDA-induced apoptosis. Immunohistochemical studies showed that NMDA caused a time-dependent increase in levels of the phosphorylated form of the ribosomal protein S6 (pS6), a downstream indicator of mTOR activity. The increased pS6 levels were markedly decreased by rapamycin. Both NMDA and rapamycin increased the level of phosphorylated ERK (pERK) in Müller cells, and coinjection of both agents further increased pERK levels. These results suggest that rapamycin has a neuroprotective effect against NMDA-induced retinal neurotoxicity and that this effect could be patially mediated by activation of the ERK pathway in retinal Müller cells.
Collapse
Affiliation(s)
- Atsuko Ichikawa
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
19
|
Dapper JD, Crish SD, Pang IH, Calkins DJ. Proximal inhibition of p38 MAPK stress signaling prevents distal axonopathy. Neurobiol Dis 2013; 59:26-37. [PMID: 23859799 DOI: 10.1016/j.nbd.2013.07.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/04/2013] [Indexed: 12/21/2022] Open
Abstract
The p38 mitogen-activated protein kinase (MAPK) isoforms are phosphorylated by a variety of stress stimuli in neurodegenerative disease and act as upstream activators of myriad pathogenic processes. Thus, p38 MAPK inhibitors are of growing interest as possible therapeutic interventions. Axonal dysfunction is an early component of most neurodegenerative disorders, including the most prevalent optic neuropathy, glaucoma. Sensitivity to intraocular pressure at an early stage disrupts anterograde transport along retinal ganglion cell (RGC) axons to projection targets in the brain with subsequent degeneration of the axons themselves; RGC body loss is much later. Here we show that elevated ocular pressure in rats increases p38 MAPK activation in retina, especially in RGC bodies. Topical eye-drop application of a potent and selective inhibitor of the p38 MAPK catalytic domain (Ro3206145) prevented both the degradation of anterograde transport to the brain and degeneration of axons in the optic nerve. Ro3206145 reduced in the retina phosphorylation of tau and heat-shock protein 27, both down-stream targets of p38 MAPK activation implicated in glaucoma, as well as expression of two inflammatory responses. We also observed increased p38 MAPK activation in mouse models. Thus, inhibition of p38 MAPK signaling in the retina may represent a therapeutic target for preventing early pathogenesis in optic neuropathies.
Collapse
Affiliation(s)
- Jason D Dapper
- The Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | | | | | |
Collapse
|
20
|
Tian Z, Liu SB, Wang YC, Li XQ, Zheng LH, Zhao MG. Neuroprotective Effects of Formononetin Against NMDA-Induced Apoptosis in Cortical Neurons. Phytother Res 2013; 27:1770-5. [PMID: 23362211 DOI: 10.1002/ptr.4928] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Revised: 11/21/2012] [Accepted: 12/19/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Zhen Tian
- Department of Pharmacology, School of Pharmacy; Fourth Military Medical University; Xi'an 710032 China
| | - Shui-bing Liu
- Department of Pharmacology, School of Pharmacy; Fourth Military Medical University; Xi'an 710032 China
| | - Yu-cai Wang
- Department of Orthopaedics, Tangdu Hospital; Fourth Military Medical University; Xi'an 710032 China
| | - Xiao-qiang Li
- Department of Pharmacology, School of Pharmacy; Fourth Military Medical University; Xi'an 710032 China
| | - Lian-he Zheng
- Department of Orthopaedics, Tangdu Hospital; Fourth Military Medical University; Xi'an 710032 China
| | - Ming-gao Zhao
- Department of Pharmacology, School of Pharmacy; Fourth Military Medical University; Xi'an 710032 China
| |
Collapse
|
21
|
Munemasa Y, Kitaoka Y. Molecular mechanisms of retinal ganglion cell degeneration in glaucoma and future prospects for cell body and axonal protection. Front Cell Neurosci 2013; 6:60. [PMID: 23316132 PMCID: PMC3540394 DOI: 10.3389/fncel.2012.00060] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 12/06/2012] [Indexed: 12/20/2022] Open
Abstract
Glaucoma, which affects more than 70 million people worldwide, is a heterogeneous group of disorders with a resultant common denominator; optic neuropathy, eventually leading to irreversible blindness. The clinical manifestations of primary open-angle glaucoma (POAG), the most common subtype of glaucoma, include excavation of the optic disc and progressive loss of visual field. Axonal degeneration of retinal ganglion cells (RGCs) and apoptotic death of their cell bodies are observed in glaucoma, in which the reduction of intraocular pressure (IOP) is known to slow progression of the disease. A pattern of localized retinal nerve fiber layer (RNFL) defects in glaucoma patients indicates that axonal degeneration may precede RGC body death in this condition. The mechanisms of degeneration of neuronal cell bodies and their axons may differ. In this review, we addressed the molecular mechanisms of cell body death and axonal degeneration in glaucoma and proposed axonal protection in addition to cell body protection. The concept of axonal protection may become a new therapeutic strategy to prevent further axonal degeneration or revive dying axons in patients with preperimetric glaucoma. Further study will be needed to clarify whether the combination therapy of axonal protection and cell body protection will have greater protective effects in early or progressive glaucomatous optic neuropathy (GON).
Collapse
Affiliation(s)
- Yasunari Munemasa
- Department of Ophthalmology, St. Marianna University School of Medicine Kawasaki, Kanagawa, Japan
| | | |
Collapse
|
22
|
Ribas VT, Gonçalves BS, Linden R, Chiarini LB. Activation of c-Jun N-terminal kinase (JNK) during mitosis in retinal progenitor cells. PLoS One 2012; 7:e34483. [PMID: 22496813 PMCID: PMC3319587 DOI: 10.1371/journal.pone.0034483] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 03/05/2012] [Indexed: 01/19/2023] Open
Abstract
Most studies of c-Jun N-terminal Kinase (JNK) activation in retinal tissue were done in the context of neurodegeneration. In this study, we investigated the behavior of JNK during mitosis of progenitor cells in the retina of newborn rats. Retinal explants from newborn rats were kept in vitro for 3 hours and under distinct treatments. Sections of retinal explants or freshly fixed retinal tissue were used to detect JNK phosphorylation by immunohistochemistry, and were examined through both fluorescence and confocal microscopy. Mitotic cells were identified by chromatin morphology, histone-H3 phosphorylation, and location in the retinal tissue. The subcellular localization of proteins was analyzed by double staining with both a DNA marker and an antibody to each protein. Phosphorylation of JNK was also examined by western blot. The results showed that in the retina of newborn rats (P1), JNK is phosphorylated during mitosis of progenitor cells, mainly during the early stages of mitosis. JNK1 and/or JNK2 were preferentially phosphorylated in mitotic cells. Inhibition of JNK induced cell cycle arrest, specifically in mitosis. Treatment with the JNK inhibitor decreased the number of cells in anaphase, but did not alter the number of cells in either prophase/prometaphase or metaphase. Moreover, cells with aberrant chromatin morphology were found after treatment with the JNK inhibitor. The data show, for the first time, that JNK is activated in mitotic progenitor cells of developing retinal tissue, suggesting a new role of JNK in the control of progenitor cell proliferation in the retina.
Collapse
Affiliation(s)
| | | | - Rafael Linden
- Instituto de Biofísica Carlos Chagas Filho, UFRJ, Rio de Janeiro, Brasil
| | | |
Collapse
|
23
|
Takada K, Munemasa Y, Kuribayashi J, Fujino H, Kitaoka Y. Protective effect of thalidomide against N-methyl-D-aspartate-induced retinal neurotoxicity. J Neurosci Res 2011; 89:1596-604. [PMID: 21702058 DOI: 10.1002/jnr.22698] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 04/19/2011] [Accepted: 04/20/2011] [Indexed: 12/23/2022]
Abstract
Thalidomide, an inhibitor of tumor necrosis factor-α (TNF-α) production, has been indicated to be useful for many inflammatory and oncogenic diseases. In the present study, we examined whether thalidomide (50 mg/kg/day, p.o.) has a protective effect against N-methyl-D-aspartate (NMDA)-induced retinal neurotoxicity in rats. A morphometric analysis showed that systemic administration of thalidomide protects neural cells in the ganglion cell layer (GCL) in a dose-dependent manner and significantly decreases the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells in GCL and in the inner nuclear layer (INL). ELISA showed that thalidomide significantly suppressed the elevation of TNF-α 6 and 24 hr after an NMDA injection. Western blot analysis revealed a significant increase in nuclear factor-κB (NF-κB) p65 level in the retinas treated with NMDA at 24 hr after the injection, but not at 6 or 72 hr. Furthermore, an increase in p-JNK and p-p38 levels was also observed in the retina after NMDA injection. Thalidomide suppressed the increased expressions of NF-κB p65, p-JNK, and p-p38 after NMDA injection. Immunohistochemical analysis showed that thalidomide attenuated NF-κB p65 immunoreactivity in the GCL induced by NMDA treatment. In the NMDA-treated group, translocation of NF-κB p65 from the cytoplasm to the nucleus was detected in TUNEL-positive cells exposed to NMDA treatment. These results suggest new indications for thalidomide against neurodegenerative diseases.
Collapse
Affiliation(s)
- Kazuhide Takada
- 5th Year, St. Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
| | | | | | | | | |
Collapse
|
24
|
Kuribayashi J, Kitaoka Y, Munemasa Y, Ueno S. Kinesin-1 and degenerative changes in optic nerve axons in NMDA-induced neurotoxicity. Brain Res 2010; 1362:133-40. [PMID: 20863816 DOI: 10.1016/j.brainres.2010.09.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 09/14/2010] [Accepted: 09/14/2010] [Indexed: 12/29/2022]
Abstract
We examined the histologic findings of optic nerve axons and changes in kinesin-1, which is involved in axonal flow, in N-methyl-d-aspartate (NMDA)-induced neurotoxicity in rats. Substantial degenerative changes visualized as black profiles and pale large axons were observed 72h after NMDA injection, but those degenerative changes were not apparent in axons 12 and 24h after injection. Morphometric analysis showed a significant, approximately 40% reduction in the number of axons 72h after NMDA injection. Immunohistochemical study showed that there was a recognizable loss of neurofilament-immunopositive dots, but myelin basic protein immunostaining was unchanged 72h after NMDA injection. Western blot analysis showed early elevation of kinesin-1 (KIF5B) protein levels in the retina 24 and 72h after NMDA injection. Conversely, significant decreases in KIF5B protein levels in the optic nerve were seen during the same time course. Immunohistochemical study also showed that there was a reduction in KIF5B immunoreactivity in axons, but neurofilament immunostaining was unchanged 24h after NMDA injection. These findings suggest that the intravitreal injection of NMDA causes neurofilament loss without myelin alteration in the early stage. The depletion of kinesin-1 precedes axonal degeneration of the optic nerve in NMDA-induced neurotoxicity.
Collapse
Affiliation(s)
- Junko Kuribayashi
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki-shi,Kanagawa 216-8511, Japan
| | | | | | | |
Collapse
|
25
|
Bessero AC, Chiodini F, Rungger-Brändle E, Bonny C, Clarke PGH. Role of the c-Jun N-terminal kinase pathway in retinal excitotoxicity, and neuroprotection by its inhibition. J Neurochem 2010; 113:1307-18. [PMID: 20345748 DOI: 10.1111/j.1471-4159.2010.06705.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Retinal excitotoxicity is associated with retinal ischemia, and with glaucomatous and traumatic optic neuropathy. The present study investigates the role of c-Jun N-terminal kinase (JNK) activation in NMDA-mediated retinal excitotoxicity and determines whether neuroprotection can be obtained with the JNK pathway inhibitor, D-form of JNK-inhibitor 1 (D-JNKI-1). Young adult rats received intravitreal injections of 20 nmol NMDA, which caused extensive neuronal death in the inner nuclear and ganglion cell layers. This excitotoxicity was associated with strong activation of calpain, as revealed by fodrin cleavage, and of JNK. The cell-permeable peptide D-JNKI-1 was used to inhibit JNK. Within 40 min of its intravitreal injection, FITC-labeled D-JNKI-1 spread through the retinal ganglion cell layer into the inner nuclear layer and interfered with the NMDA-induced phosphorylation of JNK. Injections of unlabeled D-JNKI-1 gave unprecedentedly strong neuroprotection against cell death in both layers, lasting for at least 10 days. The NMDA-induced calpain-specific fodrin cleavage was likewise strongly inhibited by D-JNKI-1. Moreover the electroretinogram was partially preserved by D-JNKI-1. Thus, the JNK pathway is involved in NMDA-mediated retinal excitotoxicity and JNK inhibition by D-JNKI-1 provides strong neuroprotection as shown morphologically, biochemically and physiologically.
Collapse
Affiliation(s)
- Anne-Caroline Bessero
- Département de Biologie Cellulaire et de Morphologie (DBCM), University of Lausanne, Lausanne, Switzerland.
| | | | | | | | | |
Collapse
|
26
|
Secretoneurin and the tachykinins substance P and neurokinin-A/B in NMDA-induced excitotoxicity in the rat retina. ACTA ACUST UNITED AC 2010; 165:123-7. [PMID: 20138192 DOI: 10.1016/j.regpep.2010.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2009] [Revised: 01/08/2010] [Accepted: 01/27/2010] [Indexed: 11/24/2022]
Abstract
In a recent investigation using the NMDA-excitotoxicity model in the rat retina, we found that, whereas, following intravitreal injection of NMDA, a time-dependent decrease of the levels of a neuropeptide, namely vasoactive intestinal polypeptide (VIP), was fully counteracted by topical treatment with flunarizine eye drops, the levels of pituitary adenylate-cyclase activating peptide-38 (PACAP-38), another neuropeptide, remained unchanged. The aim of the present study was to find out if NMDA causes reduction in the levels of other neuropeptides such as secretoneurin (SN), neurokinin-A/B (NKA/NKB) and substance P (SP), and if so, whether flunarizine has the ability to counteract this effect or prevent such reduction. The reduction of the levels of SN and NKA/NKB 14 days after intravitreal injection of 2 μl of 100 nmol NMDA into one eye was more pronounced than after 7 days; topical flunarizine had a slight counteracting effect, but could not prevent the decrease in the levels of these peptides. Reduction in SP levels after 28 and 56 days was fully counteracted by flunarizine. By enabling a pronounced influx of Ca²+ ions into peptide-expressing cells, NMDA leads to cell death. Since each of these peptides exerts neuroprotective properties in the central nervous system, the drop in their levels caused by acute insult (e.g. NMDA excitotoxicity) or chronic insult (e.g. glaucoma) may cause a breakdown of endogenous neuroprotection in the retina given that these peptides feature neuroprotective properties in the retina as well.
Collapse
|
27
|
Inokuchi Y, Imai S, Nakajima Y, Shimazawa M, Aihara M, Araie M, Hara H. Edaravone, a free radical scavenger, protects against retinal damage in vitro and in vivo. J Pharmacol Exp Ther 2009; 329:687-98. [PMID: 19201991 DOI: 10.1124/jpet.108.148676] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one), a free radical scavenger, is used for the treatment of acute cerebral infarction. In this study, we investigated whether edaravone is neuroprotective against retinal damage. In vitro, we used a radical-scavenging capacity assay using reactive oxygen species-sensitive probes to investigate the effects of edaravone on H(2)O(2), superoxide anion (O(2)*), and hydroxyl radical (*OH) production in a rat retinal ganglion cell line (RGC-5). The effect of edaravone on oxygen-glucose deprivation (OGD)-induced RGC-5 damage was evaluated using a 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazolium, monosodium salt assay of cell viability. Edaravone (3-methyl-1-phenyl-2-pyrazolin-5-one) significantly decreased radical generation and reduced the cell death induced by OGD stress. In vivo, retinal damage was induced by intravitreous injection of N-methyl-D-aspartate (NMDA; 5 nmol) and was evaluated by examining ganglion cell layer cell loss, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining, and the expressions of two oxidant-stress markers [4-hydroxy-2-nonenal (4-HNE) and 8-hydroxy-2-deoxyguanosine (8-OHdG)]. In addition, activations of mitogen-activated protein kinases (MAPKs) [extracellular signal-regulated protein kinases (ERK), c-Jun NH(2)-terminal kinases (JNK), and p38 MAPK], as downstream signal pathways after NMDA receptor activation, were measured using immunoblotting and immunostaining. Edaravone at 5 and 50 nmol intravitreous injection or at 1 and 3 mg/kg i.v. significantly protected against NMDA-induced retinal cell death. At 50 nmol intravitreous injection, it 1) decreased the retinal expressions of TUNEL-positive cells, 4-HNE, and 8-OHdG and 2) reduced the retinal expressions of NMDA-induced phosphorylated JNK and phosphorylated p38 but not that of phosphorylated ERK. These findings suggest that oxidative stress plays a pivotal role in retinal damage and that edaravone may be a candidate for the effective treatment of retinal diseases.
Collapse
Affiliation(s)
- Yuta Inokuchi
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | | | | | | | | | | | | |
Collapse
|
28
|
Protection against chronic hypoperfusion-induced retinal neurodegeneration by PARP inhibition via activation of PI-3-kinase Akt pathway and suppression of JNK and p38 MAP kinases. Neurotox Res 2009; 16:68-76. [PMID: 19526300 DOI: 10.1007/s12640-009-9049-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Revised: 03/25/2009] [Accepted: 03/27/2009] [Indexed: 12/11/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP) activation is considered as a major regulator of cell death in various pathophysiological conditions, however, no direct information is available about its role in chronic hypoperfusion-induced neuronal death. Here, we provide evidence for the protective effect of PARP inhibition on degenerative retinal damage induced by bilateral common carotid artery occlusion (BCCAO), an adequate chronic hypoperfusion murine model. We found that BCCAO in adult male Wistar rats led to severe degeneration of all retinal layers that was attenuated by a carboxaminobenzimidazol-derivative PARP inhibitor (HO3089) administered unilaterally into the vitreous body immediately following carotid occlusion and then 4 times in a 2-week-period. Normal morphological structure of the retina was preserved and the thickness of the retinal layers was increased in HO3089-treated eyes compared to the BCCAO eyes. For Western blot studies, HO3089 was administered immediately after BCCAO and retinas were removed 4 h later. According to Western blot analysis utilizing phosphorylation-specific primary antibodies, besides activating poly-ADP-ribose (PAR) synthesis, BCCAO induced phosphorylation of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). HO3089 inhibited PAR synthesis, and decreased the phosphorylation of these proapoptotic MAPKs. In addition, HO3089 treatment induced phosphorylation, that is activation, of the protective Akt/glycogen synthase kinase (GSK)-3beta and extracellular signal-regulated kinase (ERK1/2) signaling pathways. These data indicate that PARP activation has a major role in mediating chronic hypoperfusion-induced neuronal death, and inhibition of the enzyme prevents the pathological changes both in the morphology and the kinase signaling cascades involved. These results identify PARP inhibition as a possible molecular target in the clinical management of chronic hypoperfusion-induced neurodegenerative diseases including ocular ischemic syndrome.
Collapse
|
29
|
Inokuchi Y, Shimazawa M, Nakajima Y, Komuro I, Matsuda T, Baba A, Araie M, Kita S, Iwamoto T, Hara H. A Na+/Ca2+ exchanger isoform, NCX1, is involved in retinal cell death after N-methyl-D-aspartate injection and ischemia-reperfusion. J Neurosci Res 2009; 87:906-17. [PMID: 18855935 DOI: 10.1002/jnr.21906] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
We investigated the expression of Na(+)/Ca(2+) exchanger (NCX) and the functional role of NCX in retinal damage by using NCX1-heterozygous deficient mice (NCX1(+/-)) and SEA0400 (2-[4-[(2,5-difluorophenyl)methoxy] phenoxy]-5-ethoxyaniline), a selective NCX inhibitor in vivo. We also examined the role of NCX in oxygen-glucose deprivation (OGD) stress with a retinal ganglion cell line (RGC-5) cell culture in vitro. The expression of NCX1 was confirmed and entirely localized in retina by immunoblotting and immunohistochemistry, respectively. NCX1(+/-) mice possessed significant protection against retinal damage induced by intravitreal injection of N-methyl-D-aspartate (NMDA). SEA0400 at 3 and 10 mg/kg significantly reduced NMDA- or high intraocular pressure-induced retinal cell damage in mice. Furthermore, SEA0400 reduced the number of TUNEL (terminal deoxynucleotidyl transferase dUTP nick-end labeling)-positive cells and the expression of phosphorylated mitogen-activated protein kinases (ERK1/2, JNK, p38) induced by NMDA injection. In RGC-5, SEA0400 at 0.3 and 1 microM significantly inhibited OGD-induced cell damage. OGD-induced cell damage was aggravated by ouabain (a Na(+),K(+)-ATPase inhibitor) at 100 microM, and this increased damage was significantly reduced by SEA0400 at 1 microM. In conclusion, these results suggest that NCX1 may play a role in retinal cell death induced by NMDA and ischemia-reperfusion.
Collapse
Affiliation(s)
- Y Inokuchi
- Department of Biofunctional Evaluation, Molecular Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Effects of unoprostone on phosphorylated extracellular signal-regulated kinase expression in endothelin-1-induced retinal and optic nerve damage. Vis Neurosci 2008; 25:197-208. [PMID: 18442442 DOI: 10.1017/s095252380808053x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Endothelin-1 (ET-1), a potent vasoconstrictor peptide, has been implicated in the development of normal- and high-tension glaucoma. We investigated the effects of unoprostone on extracellular signal-regulated kinase (ERK) in ET-1-induced retinal ganglion cell (RGC) death and optic nerve injury. Our morphometric study showed that intravitreal injection of ET-1 led to cell loss in the RGC layer (RGCL) in 28 days. Western blot analysis showed decreased neurofilament (NF) protein in the optic nerve 28 days after ET-1 injection. In this in vivo model, increased phosphorylated ERK (p-ERK) was observed in the retina on 1 day and subsequently in the optic nerve from 7 days after ET-1 injection. Simultaneous injection of M1, as a metabolite of unoprostone, showed further increased p-ERK levels compared with ET-1 injection alone. Our morphometric study of flat-mount preparations stained with cresyl violet or retrograde labeling with a neuro-tracer and Western blot analysis of NF showed that inhibition of ERK phosphorylation led to acceleration of ET-1-induced RGC death and optic nerve damage. In addition, M1 significantly attenuated both RGC loss and the decrease in NF protein induced by ET-1. The protective effects of M1 were significantly inhibited by U0126, an ERK inhibitor. These results suggest that unoprostone has neuroprotective effects against ET-1-induced neuronal injury through ERK phosphorylation.
Collapse
|
31
|
Oliveira CS, Rigon AP, Leal RB, Rossi FM. The activation of ERK1/2 and p38 mitogen‐activated protein kinases is dynamically regulated in the developing rat visual system. Int J Dev Neurosci 2008; 26:355-62. [DOI: 10.1016/j.ijdevneu.2007.12.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 12/11/2007] [Accepted: 12/12/2007] [Indexed: 10/22/2022] Open
Affiliation(s)
- Camila Salum Oliveira
- Laboratório de Neuroquímica‐3, Departamento de BioquímicaCentro de Ciências Biológicas, Universidade Federal de Santa CatarinaFlorianópolisSC88040‐900Brazil
| | - Ana Paula Rigon
- Laboratório de Neuroquímica‐3, Departamento de BioquímicaCentro de Ciências Biológicas, Universidade Federal de Santa CatarinaFlorianópolisSC88040‐900Brazil
| | - Rodrigo Bainy Leal
- Laboratório de Neuroquímica‐3, Departamento de BioquímicaCentro de Ciências Biológicas, Universidade Federal de Santa CatarinaFlorianópolisSC88040‐900Brazil
| | - Francesco Mattia Rossi
- Laboratório de Neuroquímica‐3, Departamento de BioquímicaCentro de Ciências Biológicas, Universidade Federal de Santa CatarinaFlorianópolisSC88040‐900Brazil
| |
Collapse
|
32
|
Jiao H, Zhang L, Gao F, Lou D, Zhang J, Xu M. Dopamine D(1) and D(3) receptors oppositely regulate NMDA- and cocaine-induced MAPK signaling via NMDA receptor phosphorylation. J Neurochem 2007; 103:840-8. [PMID: 17897358 DOI: 10.1111/j.1471-4159.2007.04840.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Development of drug addiction involves complex molecular changes in the CNS. The mitogen-activated protein kinase (MAPK) signaling pathway plays a key role in mediating neuronal activation induced by dopamine, glutamate, and drugs of abuse. We previously showed that dopamine D(1) and D(3) receptors play different roles in regulating cocaine-induced MAPK activation. Although there are functional and physical interactions between dopamine and glutamate receptors, little is known regarding the involvement of D(1) and D(3) receptors in modulating glutamate-induced MAPK activation and underlying mechanisms. In this study, we show that D(1) and D(3) receptors play opposite roles in regulating N-methyl-d-aspartate (NMDA) -induced activation of extracellular signal-regulated kinase (ERK) in the caudate putamen (CPu). D(3) receptors also inhibit NMDA-induced activation of the c-Jun N-terminal kinase and p38 kinase in the CPu. NMDA-induced activation of the NMDA-receptor R1 subunit (NR1), Ca(2+)/calmodulin-dependent protein kinase II and the cAMP-response element binding protein (CREB), and cocaine-induced CREB activation in the CPu are also oppositely regulated by dopamine D(1) and D(3) receptors. Finally, the blockade of NMDA-receptor reduces cocaine-induced ERK activation, and inhibits phosphorylation of NR1, Ca(2+)/calmodulin-dependent protein kinase II, and CREB, while inhibiting ERK activation attenuates cocaine-induced CREB phosphorylation in the CPu. These results suggest that dopamine D(1) and D(3) receptors oppositely regulate NMDA- and cocaine-induced MAPK signaling via phosphorylation of NR1.
Collapse
Affiliation(s)
- Hongyuan Jiao
- Department of Anesthesia and Critical Care, University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | |
Collapse
|
33
|
Abstract
Neuronal death due to excessive activation of N-methyl-d-aspartate (NMDA) receptors is a hallmark of neurodegenerative diseases. The polyamines: putrescine, spermine, and spermidine, bind to specific sites on the NMDA receptor and promote its activation, but their role in NMDA-induced neuronal death is ill defined. In this study, we characterized the role of polyamines in excitotoxic death of retinal ganglion cells (RGCs), a population of central neurons susceptible to NMDA-induced damage. Our data show that endogenous arginase I, the rate limiting enzyme for polyamine biosynthesis, is expressed in the intact, adult retina. Intraocular injection of NMDA visibly increased arginase I expression in Müller cells, the predominant glial cell-type in the mammalian retina. Inhibition of polyamine synthesis using di-fluoro-methyl-ornithine (DFMO) was markedly neuroprotective, while injection of exogenous polyamines in conjunction with NMDA exacerbated RGC death. Blockade of the polyamine binding sites on NMDA receptors using the non-competitive antagonist ifenprodil was neuroprotective, suggesting that polyamines contribute to excitotoxic death, at least partly, by binding to NMDA receptors. Importantly, we also demonstrate that NMDA leads to activation of both the Erk1/2 and PI3 K/Akt pathways, but only the PI3 K/Akt kinase was required for di-fluoro-methyl-ornithine-induced RGC survival. In summary, our study reveals that polyamines modulate neuronal death in the retina via different mechanisms that potentiate NMDA-triggered excitotoxicity.
Collapse
Affiliation(s)
- Vincent Pernet
- Department of Pathology and Cell Biology, Université de Montréal, Boulevard Edouard-Montpetit, Montreal, Quebec, Canada
| | | | | |
Collapse
|
34
|
Bevilaqua LRM, Rossato JI, Clarke JHR, Medina JH, Izquierdo I, Cammarota M. Inhibition of c-Jun N-terminal kinase in the CA1 region of the dorsal hippocampus blocks extinction of inhibitory avoidance memory. Behav Pharmacol 2007; 18:483-9. [PMID: 17762516 DOI: 10.1097/fbp.0b013e3282ee7436] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Step-down inhibitory avoidance (IA) memory formation involves association of stepping-down from a platform present in a training box (conditioned stimulus) with a footshock (unconditioned stimulus). A single short training session is enough to induce a lasting and strong memory trace expressed as an increase in step-down latency. Repeated nonreinforced retrieval, however, induces extinction of the IA response, a process involving a new learning that overrules the original one to indicate that the conditioned stimulus no longer predicts the unconditioned stimulus. Although the molecular requirements of IA memory consolidation are well understood, comparatively less is known about the signaling pathways involved in its extinction. Here we report that, when given into dorsal CA1 immediately but not 180 min after daily nonreinforced retrieval sessions, SP60015, a specific inhibitor of the mitogen-activated protein kinase, c-Jun N-terminal kinase, impaired IA memory extinction in a dose-dependent manner without producing any motor or perceptual impairment or damaging the hippocampal formation. Our results suggest that, as happens during consolidation, extinction of IA long-term memory also requires c-Jun N-terminal kinase activity in the CA1 region of the dorsal hippocampus.
Collapse
Affiliation(s)
- Lia R M Bevilaqua
- Center for Memory Research, Biomedical Research Institute, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | | | | | | | | |
Collapse
|
35
|
Molz S, Decker H, Dal-Cim T, Cremonez C, Cordova FM, Leal RB, Tasca CI. Glutamate-induced toxicity in hippocampal slices involves apoptotic features and p38 MAPK signaling. Neurochem Res 2007; 33:27-36. [PMID: 17616814 DOI: 10.1007/s11064-007-9402-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Accepted: 06/05/2007] [Indexed: 02/02/2023]
Abstract
Glutamate excitotoxicity may culminate with neuronal and glial cell death. Glutamate induces apoptosis in vivo and in cell cultures. However, glutamate-induced apoptosis and the signaling pathways related to glutamate-induced cell death in acute hippocampal slices remain elusive. Hippocampal slices exposed to 1 or 10 mM glutamate for 1 h and evaluated after 6 h, showed reduced cell viability, without altering membrane permeability. This action of glutamate was accompanied by cytochrome c release, caspase-3 activation and DNA fragmentation. Glutamate at low concentration (10 microM) induced caspase-3 activation and DNA fragmentation, but it did not cause cytochrome c release and, it did not alter the viability of slices. Glutamate-induced impairment of hippocampal cell viability was completely blocked by MK-801 (non-competitive antagonist of NMDA receptors) and GAMS (antagonist of KA/AMPA glutamate receptors). Regarding intracellular signaling pathways, glutamate-induced cell death was not altered by a MEK1 inhibitor, PD98059. However, the p38 MAPK inhibitor, SB203580, prevented glutamate-induced cell damage. In the present study we have shown that glutamate induces apoptosis in hippocampal slices and it causes an impairment of cell viability that was dependent of ionotropic and metabotropic receptors activation and, may involve the activation of p38 MAPK pathway.
Collapse
Affiliation(s)
- Simone Molz
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Trindade, 88040-900 Florianopolis, SC, Brazil
| | | | | | | | | | | | | |
Collapse
|
36
|
Effects of pituitary adenylate cyclase activating polypeptide (PACAP) on the PKA-bad-14-3-3 signaling pathway in glutamate-induced retinal injury in neonatal rats. Neurotox Res 2007; 12:95-104. [DOI: 10.1007/bf03033918] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
37
|
Kitaoka Y, Munemasa Y, Nakazawa T, Ueno S. NMDA-induced interleukin-1β expression is mediated by nuclear factor-kappa B p65 in the retina. Brain Res 2007; 1142:247-55. [DOI: 10.1016/j.brainres.2007.01.097] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2006] [Revised: 01/12/2007] [Accepted: 01/12/2007] [Indexed: 12/16/2022]
|
38
|
Hayashi Y, Kitaoka Y, Munemasa Y, Ohtani-Kaneko R, Kikusui T, Uematsu A, Takeda H, Hirata K, Mori Y, Ueno S. Neuroprotective effect of 17beta-estradiol against N-methyl-D-aspartate-induced retinal neurotoxicity via p-ERK induction. J Neurosci Res 2007; 85:386-94. [PMID: 17131424 DOI: 10.1002/jnr.21127] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We investigated whether the neuroprotective effect of estrogen is mediated by the estrogen receptor (ER) and whether extracellular signal-regulated kinase (ERK) is involved in the protective effect of estrogen against N-methyl-D-aspartate (NMDA)-induced retinal neurotoxicity. Retrograde labeling of retinal ganglion cells (RGCs) showed that pretreatment with 17beta-estradiol (E2) using a silastic implant significantly attenuated the loss of RGCs induced by intravitreal injection of NMDA. Simultaneous administration of U0126, an ERK inhibitor, with NMDA completely abolished the protective effect of E2. Moreover, ICI182,780, an ER antagonist, also significantly diminished the protective effect of E2. Pretreatment with E2 significantly reduced the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive cells in the retinal ganglion cell layer (RGCL) and the inner nuclear layer (INL) 12 hr after NMDA injection. Moreover, ICI182,780 inhibited the ameliorative effect of E2 on TUNEL-positive cells in a dose-dependent manner. Immunostaining of anti-ERalpha monoclonal antibody was observed mainly in the RGCL and the INL. Western blot analysis showed a significant increase in the level of phosphorylated ERK (p-ERK) 6 hr after NMDA injection. However, NMDA did not increase the level of p-ERK protein 7 days after injection. Pretreatment of E2 induced further increases of p-ERK expression 6 hr and 7 days after NMDA injection, and U0126 and ICI182,780 significantly inhibited E2-induced p-ERK expression after 6 hr. These results suggest that E2 has an ER-mediated neuroprotective effect against NMDA-induced retinal neurotoxicity and that this effect may be associated with induction of p-ERK in the retina.
Collapse
Affiliation(s)
- Yasuhiro Hayashi
- Department of Ophthalmology, St. Marianna University School of Medicine, Miyamae-ku, Kawasaki-shi, Kanagawa, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Rácz B, Gallyas F, Kiss P, Tóth G, Hegyi O, Gasz B, Borsiczky B, Ferencz A, Roth E, Tamás A, Lengvári I, Lubics A, Reglodi D. The neuroprotective effects of PACAP in monosodium glutamate-induced retinal lesion involve inhibition of proapoptotic signaling pathways. ACTA ACUST UNITED AC 2006; 137:20-6. [PMID: 16945433 DOI: 10.1016/j.regpep.2006.02.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2005] [Revised: 02/10/2006] [Accepted: 02/27/2006] [Indexed: 11/20/2022]
Abstract
Pituitary adenylate cyclase activating polypeptide (PACAP) and its receptors are present in the retina and exert several distinct functions. PACAP has well-known neuroprotective effects in neuronal cultures in vitro and against different insults in vivo. Recently we have shown that PACAP is neuroprotective against monosodium glutamate (MSG)-induced retinal degeneration. In the present study we investigated the possible signal transduction pathways involved in the protective effect of intravitreal PACAP administration against apoptotic retinal degeneration induced by neonatal MSG treatment. MSG induced activation of proapoptotic signaling proteins and reduced the levels of antiapoptotic molecules in neonatal retinas. Co-treatment with PACAP attenuated the MSG-induced activation of caspase-3 and JNK, inhibited the MSG-induced cytosolic translocation of apoptosis inducing factor (AIF) and cytochrome c, and increased the level of phospho-Bad. Furthermore, PACAP treatment alone decreased cytosolic AIF and cytochrome c levels, while PACAP6-38 increased cytochrome c release, caspase-3 and JNK activity and decreased phospho-Bad activity. In summary, our results show that PACAP treatment attenuated the MSG-induced changes in apoptotic signaling molecules in vivo and suggest that also endogenously present PACAP has neuroprotective effects. These results may have further clinical implications in reducing glutamate-induced excitotoxicity in several ophthalmic diseases.
Collapse
Affiliation(s)
- Boglárka Rácz
- Department of Surgical Research and Techniques, University of Pecs, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Inomata Y, Nakamura H, Tanito M, Teratani A, Kawaji T, Kondo N, Yodoi J, Tanihara H. Thioredoxin inhibits NMDA-induced neurotoxicity in the rat retina. J Neurochem 2006; 98:372-85. [PMID: 16805832 DOI: 10.1111/j.1471-4159.2006.03871.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Thioredoxin (TRX) plays a variety of redox-related roles in organisms. To investigate its function as an endogenous redox regulator in NMDA-induced retinal neurotoxicity, we injected NMDA with TRX, mutant TRX or saline into the vitreous cavity of rat eyes. Retinal ganglion cells were rescued by TRX, compared with saline, when evaluated by retrograde labeling analysis at 7 days after NMDA injection. TRX, but not its mutant form, prevented NMDA-induced apoptosis in the retina, as measured by terminal deoxynucleotidyl transferase-mediated UTP nick-end labeling. The induction of caspase 3 and 9, but not caspase 8, by NMDA was significantly lower in TRX-treated eyes than in saline-treated eyes. NMDA-induced activation of the MAPKs, p38 kinase and c-Jun N-terminal kinase after 6 h and of the MAPK kinases (MKKs) MKK3/6 and MKK4 after 3 h was markedly suppressed in retinal ganglion cells by TRX but not by the mutant form. NMDA-induced increases in protein carbonylation, nitrosylation and lipid peroxidation were also suppressed in TRX-treated eyes. We concluded that the intravitreous injection of TRX effectively attenuated NMDA-induced retinal cell damage and that suppression of oxidative stress and inhibition of apoptotic signaling pathways were involved in this neuroprotection.
Collapse
Affiliation(s)
- Yasuya Inomata
- Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Munemasa Y, Ohtani-Kaneko R, Kitaoka Y, Kumai T, Kitaoka Y, Hayashi Y, Watanabe M, Takeda H, Hirata K, Ueno S. Pro-apoptotic role of c-Jun in NMDA-induced neurotoxicity in the rat retina. J Neurosci Res 2006; 83:907-18. [PMID: 16477618 DOI: 10.1002/jnr.20786] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
We examined the role of c-Jun on N-methyl-D-aspartate (NMDA)-induced neurotoxicity in the rat retina. An increase in c-Jun mRNA, c-Jun protein and phosphorylated c-Jun (p-c-Jun) levels in the retina was detected 3 hr after intravitreal injection of NMDA (20 nmol). These levels peaked after 12 hr, and then returned to their control levels by 24 hr. c-Jun and p-c-Jun immunoreactivities were observed in the retinal ganglion cell layer (RGCL), especially in retinal ganglion cells (RGCs), and in the inner nuclear layer (INL) 12 hr after NMDA injection, and terminal deoxynucleotidyl transferase-mediated nick-end labeling (TUNEL)-positive cells were immunopositive for c-Jun and p-c-Jun. A c-Jun antisense oligodeoxynucleotide (AS ODN), which was simultaneously injected with NMDA, penetrated the cells in the RGCL and the INL, suppressed the NMDA-induced increase in c-Jun and p-c-Jun protein levels and reduced the number of TUNEL-positive cells in the RGCL 12 hr after the injection. The protective effect of c-Jun AS ODN on the NMDA-treated retina was also shown by the RGCL cell count and measurement of the IPL thickness, as well as by quantitative real-time PCR analysis of Thy-1 mRNA 7 days after the injection. These results suggest that c-Jun synthesis and phosphorylation participate in NMDA-induced neuronal cell death.
Collapse
Affiliation(s)
- Yasunari Munemasa
- Department of Ophthalmology, St. Marianna University School of Medicine, Kawasaki-shi, Kanagawa, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kuribayashi K, Kitaoka Y, Kumai T, Munemasa Y, Kitaoka Y, Isenoumi K, Motoki M, Kogo J, Hayashi Y, Kobayashi S, Ueno S. Neuroprotective effect of atrial natriuretic peptide against NMDA-induced neurotoxicity in the rat retina. Brain Res 2006; 1071:34-41. [PMID: 16443199 DOI: 10.1016/j.brainres.2005.11.068] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2005] [Revised: 11/18/2005] [Accepted: 11/22/2005] [Indexed: 12/30/2022]
Abstract
Atrial natriuretic peptide (ANP) can regulate aqueous humor production in the eye and has recently been suggested to play some functional roles in the retina. It has also been reported that ANP increases tyrosine hydroxylase (TH) mRNA levels and intracellular dopamine levels in PC12 cells. The effect of ANP on TH levels and the role of ANP in retinal excitotoxicity remain unknown. In this study, we investigated the effects of ANP on TH expression and dopamine levels in rat retina after intravitreal injection of NMDA. Immunohistochemistry localized natriuretic peptide receptor-A (NPRA) in the ganglion cell layer (GCL), the inner nuclear layer (INL) and the outer nuclear layer (ONL) in the rat retina. Quantitative real-time PCR and Western blot analysis showed a dramatic reduction in retinal TH levels 5 days after NMDA injection, while ANP, at a concentration of 10(-4) M, ameliorated this reduction in TH mRNA and TH protein levels. High-performance liquid chromatography (HPLC) analysis showed that NMDA reduced dopamine levels in the retina, and that ANP attenuated this reduction. Moreover, morphological analysis showed that ANP ameliorated NMDA-induced neurotoxicity through NPRA. The ameliorative effect of ANP was inhibited by a dopamine D(1) receptor antagonist. These results suggest that ANP may have a neuroprotective effect through possible involvement of dopamine induction.
Collapse
Affiliation(s)
- Kohei Kuribayashi
- Department of Ophthalmology, St. Marianna University School of Medicine, 2-16-1 Sugao, Kawasaki-shi, Kanagawa 216-8511, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|