1
|
Xu W, Liu H. Is CaMKII friend or foe for cell apoptosis in eye?: A narrative review. Medicine (Baltimore) 2023; 102:e36136. [PMID: 38050317 PMCID: PMC10695602 DOI: 10.1097/md.0000000000036136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/25/2023] [Indexed: 12/06/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) controls cell proliferation, differentiation, apoptosis, and other biological processes that have an essential role in eye diseases. However, it seems that previous studies have generated conflicting conclusions about the effect of CaMKII on cell apoptosis. In this review, we explore the positive and potentially deleterious effects of CaMKII on eye cell apoptosis. We can safely conclude that the early elevation of CaMKII could be viewed as a promoter of cell apoptosis. Overexpression of CaMKII by transfection or pretreatment with drugs helped combat cell apoptosis.
Collapse
Affiliation(s)
- Weixing Xu
- School of Graduate, Dalian Medical University, Dalian, China
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Hua Liu
- School of Graduate, Dalian Medical University, Dalian, China
- School of Graduate, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
2
|
Song I, Kuznetsova T, Baidoe-Ansah D, Mirzapourdelavar H, Senkov O, Hayani H, Mironov A, Kaushik R, Druzin M, Johansson S, Dityatev A. Heparan Sulfates Regulate Axonal Excitability and Context Generalization through Ca 2+/Calmodulin-Dependent Protein Kinase II. Cells 2023; 12:cells12050744. [PMID: 36899880 PMCID: PMC10000602 DOI: 10.3390/cells12050744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/03/2023] Open
Abstract
Our previous studies demonstrated that enzymatic removal of highly sulfated heparan sulfates with heparinase 1 impaired axonal excitability and reduced expression of ankyrin G at the axon initial segments in the CA1 region of the hippocampus ex vivo, impaired context discrimination in vivo, and increased Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity in vitro. Here, we show that in vivo delivery of heparinase 1 in the CA1 region of the hippocampus elevated autophosphorylation of CaMKII 24 h after injection in mice. Patch clamp recording in CA1 neurons revealed no significant heparinase effects on the amplitude or frequency of miniature excitatory and inhibitory postsynaptic currents, while the threshold for action potential generation was increased and fewer spikes were generated in response to current injection. Delivery of heparinase on the next day after contextual fear conditioning induced context overgeneralization 24 h after injection. Co-administration of heparinase with the CaMKII inhibitor (autocamtide-2-related inhibitory peptide) rescued neuronal excitability and expression of ankyrin G at the axon initial segment. It also restored context discrimination, suggesting the key role of CaMKII in neuronal signaling downstream of heparan sulfate proteoglycans and highlighting a link between impaired CA1 pyramidal cell excitability and context generalization during recall of contextual memories.
Collapse
Affiliation(s)
- Inseon Song
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Tatiana Kuznetsova
- Department of Integrative Medical Biology, Umeå University, 90187 Umeå, Sweden
| | - David Baidoe-Ansah
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Hadi Mirzapourdelavar
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Oleg Senkov
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Hussam Hayani
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Andrey Mironov
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Rahul Kaushik
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
| | - Michael Druzin
- Department of Integrative Medical Biology, Umeå University, 90187 Umeå, Sweden
| | - Staffan Johansson
- Department of Integrative Medical Biology, Umeå University, 90187 Umeå, Sweden
| | - Alexander Dityatev
- Molecular Neuroplasticity Group, German Center for Neurodegenerative Diseases (DZNE), 39120 Magdeburg, Germany
- Medizinische Fakultät, Otto-von-Güricke-Universität Magdeburg, 39120 Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), 39106 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-67-24526; Fax: +49-391-6724530
| |
Collapse
|
3
|
Guo X, Chen B. Investigating the role of Ca 2+/calmodulin-dependent protein kinase II in the survival of retinal ganglion cells. Neural Regen Res 2021; 17:1001-1002. [PMID: 34558519 PMCID: PMC8552834 DOI: 10.4103/1673-5374.324844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Xinzheng Guo
- Departments of Ophthalmology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bo Chen
- Departments of Ophthalmology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
4
|
Guo X, Zhou J, Starr C, Mohns EJ, Li Y, Chen EP, Yoon Y, Kellner CP, Tanaka K, Wang H, Liu W, Pasquale LR, Demb JB, Crair MC, Chen B. Preservation of vision after CaMKII-mediated protection of retinal ganglion cells. Cell 2021; 184:4299-4314.e12. [PMID: 34297923 PMCID: PMC8530265 DOI: 10.1016/j.cell.2021.06.031] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/22/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022]
Abstract
Retinal ganglion cells (RGCs) are the sole output neurons that transmit visual information from the retina to the brain. Diverse insults and pathological states cause degeneration of RGC somas and axons leading to irreversible vision loss. A fundamental question is whether manipulation of a key regulator of RGC survival can protect RGCs from diverse insults and pathological states, and ultimately preserve vision. Here, we report that CaMKII-CREB signaling is compromised after excitotoxic injury to RGC somas or optic nerve injury to RGC axons, and reactivation of this pathway robustly protects RGCs from both injuries. CaMKII activity also promotes RGC survival in the normal retina. Further, reactivation of CaMKII protects RGCs in two glaucoma models where RGCs degenerate from elevated intraocular pressure or genetic deficiency. Last, CaMKII reactivation protects long-distance RGC axon projections in vivo and preserves visual function, from the retina to the visual cortex, and visually guided behavior.
Collapse
Affiliation(s)
- Xinzheng Guo
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jing Zhou
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christopher Starr
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ethan J Mohns
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Yidong Li
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | - Yonejung Yoon
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christopher P Kellner
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kohichi Tanaka
- Laboratory of Molecular Neuroscience, School of Biomedical Sciences and Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hongbing Wang
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Wei Liu
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jonathan B Demb
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Michael C Crair
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Ophthalmology and Visual Science, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Bo Chen
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
5
|
Rasmussen M, Welinder C, Schwede F, Ekström P. The cGMP system in normal and degenerating mouse neuroretina: New proteins with cGMP interaction potential identified by a proteomics approach. J Neurochem 2020; 157:2173-2186. [PMID: 33230839 PMCID: PMC8359485 DOI: 10.1111/jnc.15251] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/17/2022]
Abstract
The hereditary disease Retinitis pigmentosa results in severe vision loss due to photoreceptor degeneration by unclear mechanisms. In several disease models, the second messenger cGMP accumulates in the degenerating photoreceptors, where it may over‐activate specific cGMP‐interacting proteins, like cGMP‐dependent protein kinase. Moreover, interventions that counteract the activity of these proteins lead to reduced photoreceptor cell death. Yet there is little or no information whether other than such regular cGMP‐interactors are present in the retina, which we, therefore, investigated in wild‐type and retinal degeneration (rd1, rd10, and rd2) mouse models. An affinity chromatography based proteomics approach that utilized immobilized cGMP analogs was applied to enrich and select for regular and potentially new cGMP‐interacting proteins as identified by mass spectrometry. This approach revealed 12 regular and 10 potentially new retinal cGMP‐interacting proteins (e.g., EPAC2 and CaMKIIα). Several of the latter were found to be expressed in the photoreceptors and to have proximity to cGMP and may thus be of interest when defining prospective therapeutic targets or biomarkers for retinal degeneration.
Collapse
Affiliation(s)
- Michel Rasmussen
- Faculty of Medicine, Department of Clinical Sciences Lund, Lund University, Ophthalmology, Lund, Sweden
| | - Charlotte Welinder
- Faculty of Medicine, Department of Clinical Sciences Lund, Oncology, Lund University, Lund, Sweden
| | - Frank Schwede
- BIOLOG Life Science Institute GmbH & Co. KG, Bremen, Germany
| | - Per Ekström
- Faculty of Medicine, Department of Clinical Sciences Lund, Lund University, Ophthalmology, Lund, Sweden
| |
Collapse
|
6
|
Pichavaram P, Palani CD, Patel C, Xu Z, Shosha E, Fouda AY, Caldwell RB, Narayanan SP. Targeting Polyamine Oxidase to Prevent Excitotoxicity-Induced Retinal Neurodegeneration. Front Neurosci 2019; 12:956. [PMID: 30686964 PMCID: PMC6335392 DOI: 10.3389/fnins.2018.00956] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 11/30/2018] [Indexed: 12/21/2022] Open
Abstract
Dysfunction of retinal neurons is a major cause of vision impairment in blinding diseases that affect children and adults worldwide. Cellular damage resulting from polyamine catabolism has been demonstrated to be a major player in many neurodegenerative conditions. We have previously shown that inhibition of polyamine oxidase (PAO) using MDL 72527 significantly reduced retinal neurodegeneration and cell death signaling pathways in hyperoxia-mediated retinopathy. In the present study, we investigated the impact of PAO inhibition in limiting retinal neurodegeneration in a model of NMDA (N-Methyl-D-aspartate)-induced excitotoxicity. Adult mice (8–10 weeks old) were given intravitreal injections (20 nmoles) of NMDA or NMLA (N-Methyl-L-aspartate, control). Intraperitoneal injection of MDL 72527 (40 mg/kg body weight/day) or vehicle (normal saline) was given 24 h before NMDA or NMLA treatment and continued until the animals were sacrificed (varied from 1 to 7 days). Analyses of retinal ganglion cell (RGC) layer cell survival was performed on retinal flatmounts. Retinal cryostat sections were prepared for immunostaining, TUNEL assay and retinal thickness measurements. Fresh frozen retinal samples were used for Western blotting analysis. A marked decrease in the neuronal survival in the RGC layer was observed in NMDA treated retinas compared to their NMLA treated controls, as studied by NeuN immunostaining of retinal flatmounts. Treatment with MDL 72527 significantly improved survival of NeuN positive cells in the NMDA treated retinas. Excitotoxicity induced neurodegeneration was also demonstrated by reduced levels of synaptophysin and degeneration of inner retinal neurons in NMDA treated retinas compared to controls. TUNEL labeling studies showed increased cell death in the NMDA treated retinas. However, treatment with MDL 72527 markedly reduced these changes. Analysis of signaling pathways during excitotoxic injury revealed the downregulation of pro-survival signaling molecules p-ERK and p-Akt, and the upregulation of a pro-apoptotic molecule BID, which were normalized with PAO inhibition. Our data demonstrate that inhibition of polyamine oxidase blocks NMDA-induced retinal neurodegeneration and promotes cell survival, thus offering a new therapeutic target for retinal neurodegenerative disease conditions.
Collapse
Affiliation(s)
- Prahalathan Pichavaram
- Vision Discovery Institute, Augusta University, Augusta, GA, United States.,College of Allied Health Sciences, Augusta University, Augusta, GA, United States
| | - Chithra Devi Palani
- Vision Discovery Institute, Augusta University, Augusta, GA, United States.,Vascular Biology Center, Augusta University, Augusta, GA, United States.,Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States
| | - Chintan Patel
- Vision Discovery Institute, Augusta University, Augusta, GA, United States.,Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Zhimin Xu
- Vision Discovery Institute, Augusta University, Augusta, GA, United States.,Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Esraa Shosha
- Vision Discovery Institute, Augusta University, Augusta, GA, United States.,Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Abdelrahman Y Fouda
- Vision Discovery Institute, Augusta University, Augusta, GA, United States.,Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Ruth B Caldwell
- Vision Discovery Institute, Augusta University, Augusta, GA, United States.,Vascular Biology Center, Augusta University, Augusta, GA, United States.,VA Medical Center, Augusta, GA, United States
| | - Subhadra Priya Narayanan
- Vision Discovery Institute, Augusta University, Augusta, GA, United States.,College of Allied Health Sciences, Augusta University, Augusta, GA, United States.,Vascular Biology Center, Augusta University, Augusta, GA, United States.,Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, United States.,VA Medical Center, Augusta, GA, United States
| |
Collapse
|
7
|
Zhao Q, Qu R, Teng L, Yin C, Yuan Y. HO-1 protects the nerves of rats with cerebral hemorrhage by regulating the PI3K/AKT signaling pathway. Neuropsychiatr Dis Treat 2019; 15:1459-1468. [PMID: 31239681 PMCID: PMC6551621 DOI: 10.2147/ndt.s197030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
Objective: This study aimed to investigate the neuroprotective effect of heme oxygenase-1 (HO-1) on the PI3K/AKT signaling pathway in rats with cerebral hemorrhage. Materials and methods: Adult male Sprague-Dawley rats were randomly divided into: a sham group, a model group and an HO-1 inhibitor group (ZnPP group). Functional defects after surgery were scored according to the Longa5 standard. Hemotoxylin and eosin staining was used to detect whether the model was constructed successfully. Superoxide dismutase (SOD) vitality and malondialdehyde (MDA) content were calculated by the xanthine oxidase method and thiobarbituric acid method, respectively. Blood-brain barrier permeability was measured by Evans Blue. Apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling assay. The expression of Bcl-2 and BAX was evaluated by immunohistochemistry and the expression of PI3K, p-PI3K, AKT and p-AKT was tested by Western blotting. Results: The rat intracerebral hemorrhage model was successfully constructed. Compared with the model group, the bleeding in the ZnPP group was more serious, the cell edema and deformation were aggravated, and the neurological deficit score in the rat was significantly increased. In addition, the content of Evans blue, MDA, the number of apoptotic cells, the water content of brain tissue and the expression of BAX were significantly increased, while the SOD activity and the expressions of Bcl-2, p-PI3K and p-AKT protein were decreased. Conclusion: HO-1 could protect the nerves of rats with cerebral hemorrhage by regulating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Qingping Zhao
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province 264000, People's Republic of China
| | - Rongbo Qu
- Department of Neurosurgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai City, Shandong Province 264100, People's Republic of China
| | - Lu Teng
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province 264000, People's Republic of China
| | - Changyou Yin
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province 264000, People's Republic of China
| | - Yuan Yuan
- Department of Neurosurgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai City, Shandong Province 264000, People's Republic of China
| |
Collapse
|
8
|
Deng G, Orfila JE, Dietz RM, Moreno-Garcia M, Rodgers KM, Coultrap SJ, Quillinan N, Traystman RJ, Bayer KU, Herson PS. Autonomous CaMKII Activity as a Drug Target for Histological and Functional Neuroprotection after Resuscitation from Cardiac Arrest. Cell Rep 2017; 18:1109-1117. [PMID: 28147268 PMCID: PMC5540152 DOI: 10.1016/j.celrep.2017.01.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 12/22/2016] [Accepted: 01/07/2017] [Indexed: 11/21/2022] Open
Abstract
The Ca2+/calmodulin-dependent protein kinase II (CaMKII) is a major mediator of physiological glutamate signaling, but its role in pathological glutamate signaling (excitotoxicity) remains less clear, with indications for both neurotoxic and neuro-protective functions. Here, the role of CaMKII in ischemic injury is assessed utilizing our mouse model of cardiac arrest and cardiopulmonary resuscitation (CA/CPR). CaMKII inhibition (with tatCN21 or tatCN19o) at clinically relevant time points (30 min after resuscitation) greatly reduces neuronal injury. Importantly, CaMKII inhibition also works in combination with mild hypothermia, the current standard of care. The relevant drug target is specifically Ca2+-independent “autonomous” CaMKII activity generated by T286 autophosphorylation, as indicated by substantial reduction in injury in autonomy-incompetent T286A mutant mice. In addition to reducing cell death, tatCN19o also protects the surviving neurons from functional plasticity impairments and prevents behavioral learning deficits, even at extremely low doses (0.01 mg/kg), further highlighting the clinical potential of our findings.
Collapse
Affiliation(s)
- Guiying Deng
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - James E Orfila
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Robert M Dietz
- Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Myriam Moreno-Garcia
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Krista M Rodgers
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Steve J Coultrap
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Nidia Quillinan
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Richard J Traystman
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Paco S Herson
- Department of Anesthesiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Neuronal Injury Program, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| |
Collapse
|
9
|
Xu Q, Deng F, Xing Z, Wu Z, Cen B, Xu S, Zhao Z, Nepomuceno R, Bhuiyan MIH, Sun D, Wang QJ, Ji A. Long non-coding RNA C2dat1 regulates CaMKIIδ expression to promote neuronal survival through the NF-κB signaling pathway following cerebral ischemia. Cell Death Dis 2016; 7:e2173. [PMID: 27031970 PMCID: PMC4823958 DOI: 10.1038/cddis.2016.57] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 02/19/2016] [Accepted: 02/23/2016] [Indexed: 11/08/2022]
Abstract
Increasing evidence has demonstrated a significant role of long non-coding RNAs (lncRNAs) in diverse biological processes. However, their functions in cerebral ischemia remain largely unknown. Through an lncRNA array analysis in a rat model of focal cerebral ischemia/reperfusion (I/R), we have identified CAMK2D-associated transcript 1 (C2dat1) as a novel I/R-induced lncRNA that regulated the expression of CaMKIIδ in murine models of focal cerebral ischemia. C2dat1 mRNA was upregulated in a time-dependent manner in mouse cortical penumbra after focal ischemic brain injury, which was accompanied by increased expression of CaMKIIδ at transcript and protein levels. The expression patterns of C2dat1 and CAMK2D were confirmed in mouse Neuro-2a cells in response to in vitro ischemia (oxygen-glucose deprivation/reoxygenation, OGD/R). Knockdown of C2dat1 resulted in a significant blockade of CaMKIIδ expression, and potentiated OGD/R-induced cell death. Mechanistically, reduced CaMKIIδ expression upon silencing C2dat1 inhibited OGD/R-induced activation of the NF-κB signaling pathway. Further analysis showed that the downregulation of IKKα and IKKβ expression and phosphorylation, and subsequent inhibition of IκBα degradation accounted for the inhibition of the NF-κB signaling activity caused by silencing C2dat1. In summary, we discovered a novel I/R-induced lncRNA C2dat1 that modulates the expression of CaMKIIδ to impact neuronal survival, and may be a potential target for therapeutic intervention of ischemic brain injury.
Collapse
Affiliation(s)
- Q Xu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - F Deng
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Z Xing
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Z Wu
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - B Cen
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - S Xu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Z Zhao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - R Nepomuceno
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - M I H Bhuiyan
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - D Sun
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Q J Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - A Ji
- Department of Pharmacy, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Li J, Wang P, Zhu Y, Chen Z, Shi T, Lei W, Yu S. Curcumin Inhibits Neuronal Loss in the Retina and Elevates Ca²⁺/Calmodulin-Dependent Protein Kinase II Activity in Diabetic Rats. J Ocul Pharmacol Ther 2015. [PMID: 26207889 DOI: 10.1089/jop.2015.0006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
PURPOSE To determine whether curcumin offers neuroprotection to minimize the apoptosis of neural cells in the retina of diabetic rats. METHODS Streptozotocin (STZ)-induced diabetic rats and control rats were used in this study. A subgroup of STZ-induced diabetic rats were treated with curcumin for 12 weeks. Retinal histology, apoptosis of neural cells in the retina, electroretinograms, and retinal glutamate content were evaluated after 12 weeks. Retinal levels of Ca(2+)/calmodulin-dependent protein kinase II (CaMKII), phospho-CaMKII (p-CaMKII), and cleaved caspase-3 were determined by Western blot analysis. RESULTS The amplitudes a-wave, b-wave, and oscillatory potential were reduced by diabetes, but curcumin treatment suppressed this reduction of amplitudes. Curcumin also prevented cell loss from the outer nuclear, inner nuclear, and ganglion cell layers. Apoptosis of retinal neurons was detected in diabetic rats. The concentration of glutamate in the retina was higher in diabetic rats, but was significantly reduced in the curcumin-treated group. Furthermore, p-CaMKII and cleaved caspase-3 expression were upregulated in the diabetic retina, but reduced in curcumin-treated rats. CONCLUSIONS Curcumin attenuated diabetes-induced apoptosis in retinal neurons by reducing the glutamate level and downregulating CaMKII. Thus, curcumin might be used to prevent neuronal damage in the retina of patients with diabetes mellitus.
Collapse
Affiliation(s)
- Jun Li
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Peipei Wang
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Yanxia Zhu
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Zhen Chen
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Tianyan Shi
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Wensheng Lei
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| | - Songping Yu
- Department of Ophthalmology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University , Lishui, Zhejiang Province, People's Republic of China
| |
Collapse
|
11
|
Szabadfi K, Pinter E, Reglodi D, Gabriel R. Neuropeptides, trophic factors, and other substances providing morphofunctional and metabolic protection in experimental models of diabetic retinopathy. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2014; 311:1-121. [PMID: 24952915 DOI: 10.1016/b978-0-12-800179-0.00001-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vision is the most important sensory modality for many species, including humans. Damage to the retina results in vision loss or even blindness. One of the most serious complications of diabetes, a disease that has seen a worldwide increase in prevalence, is diabetic retinopathy. This condition stems from consequences of pathological metabolism and develops in 75% of patients with type 1 and 50% with type 2 diabetes. The development of novel protective drugs is essential. In this review we provide a description of the disease and conclude that type 1 diabetes and type 2 diabetes lead to the same retinopathy. We evaluate existing experimental models and recent developments in finding effective compounds against this disorder. In our opinion, the best models are the long-term streptozotocin-induced diabetes and Otsuka Long-Evans Tokushima Fatty and spontaneously diabetic Torii rats, while the most promising substances are topically administered somatostatin and pigment epithelium-derived factor analogs, antivasculogenic substances, and systemic antioxidants. Future drug development should focus on these.
Collapse
Affiliation(s)
- Krisztina Szabadfi
- Department of Experimental Zoology and Neurobiology, University of Pecs, Pecs, Hungary; Janos Szentagothai Research Center, University of Pecs, Pecs, Hungary.
| | - Erika Pinter
- Janos Szentagothai Research Center, University of Pecs, Pecs, Hungary; Department of Pharmacology and Pharmacotherapy, University of Pecs, Pecs, Hungary
| | - Dora Reglodi
- Department of Anatomy, PTE MTA Lendulet-PACAP Research Team, University of Pecs, Pecs, Hungary
| | - Robert Gabriel
- Department of Experimental Zoology and Neurobiology, University of Pecs, Pecs, Hungary; Janos Szentagothai Research Center, University of Pecs, Pecs, Hungary
| |
Collapse
|
12
|
Receptor interacting protein kinase-mediated necrosis contributes to cone and rod photoreceptor degeneration in the retina lacking interphotoreceptor retinoid-binding protein. J Neurosci 2013; 33:17458-68. [PMID: 24174679 DOI: 10.1523/jneurosci.1380-13.2013] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Interphotoreceptor retinoid-binding protein (IRBP) secreted by photoreceptors plays a pivotal role in photoreceptor survival with an unknown mechanism. A mutation in the human IRBP has been linked to retinitis pigmentosa, a progressive retinal degenerative disease. Mice lacking IRBP display severe early and progressive photoreceptor degeneration. However, the signaling pathway(s) leading to photoreceptor death in IRBP-deficient mice remains poorly understood. Here, we show that amounts of tumor necrosis factor-α (TNF-α) in the interphotoreceptor matrix and retinas of Irbp(-/-) mice were increased more than 10-fold and fivefold, respectively, compared with those in wild-type mice. Moreover, TNF-α receptor 1, an important membrane death receptor that mediates both programmed apoptosis and necrosis, was also significantly increased in Irbp(-/-) retina, and was colocalized with peanut agglutinin to the Irbp(-/-) cone outer segments. Although these death signaling proteins were increased, the caspase-dependent and independent apoptotic pathways were mildly activated in the Irbp(-/-) retinas, suggesting that other cell death mechanism(s) also contributes to the extensive photoreceptor degeneration in Irbp(-/-) retina. We found that receptor interacting protein 1 and 3 (RIP1 and RIP3) kinases, the intracellular key mediators of TNF-induced cellular necrosis, were elevated at least threefold in the Irbp(-/-) retinas. Moreover, pharmacological inhibition of RIP1 kinase significantly prevented cone and rod photoreceptor degeneration in Irbp(-/-) mice. These results reveal that RIP kinase-mediated necrosis strongly contributes to cone and rod degeneration in Irbp(-/-) mice, implicating the TNF-RIP pathway as a potential therapeutic target to prevent or delay photoreceptor degeneration in patients with retinitis pigmentosa caused by IRBP mutation.
Collapse
|
13
|
Ashpole NM, Song W, Brustovetsky T, Engleman EA, Brustovetsky N, Cummins TR, Hudmon A. Calcium/calmodulin-dependent protein kinase II (CaMKII) inhibition induces neurotoxicity via dysregulation of glutamate/calcium signaling and hyperexcitability. J Biol Chem 2012; 287:8495-506. [PMID: 22253441 DOI: 10.1074/jbc.m111.323915] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Aberrant glutamate and calcium signalings are neurotoxic to specific neuronal populations. Calcium/calmodulin-dependent kinase II (CaMKII), a multifunctional serine/threonine protein kinase in neurons, is believed to regulate neurotransmission and synaptic plasticity in response to calcium signaling produced by neuronal activity. Importantly, several CaMKII substrates control neuronal structure, excitability, and plasticity. Here, we demonstrate that CaMKII inhibition for >4 h using small molecule and peptide inhibitors induces apoptosis in cultured cortical neurons. The neuronal death produced by prolonged CaMKII inhibition is associated with an increase in TUNEL staining and caspase-3 cleavage and is blocked with the translation inhibitor cycloheximide. Thus, this neurotoxicity is consistent with apoptotic mechanisms, a conclusion that is further supported by dysregulated calcium signaling with CaMKII inhibition. CaMKII inhibitory peptides also enhance the number of action potentials generated by a ramp depolarization, suggesting increased neuronal excitability with a loss of CaMKII activity. Extracellular glutamate concentrations are augmented with prolonged inhibition of CaMKII. Enzymatic buffering of extracellular glutamate and antagonism of the NMDA subtype of glutamate receptors prevent the calcium dysregulation and neurotoxicity associated with prolonged CaMKII inhibition. However, in the absence of CaMKII inhibition, elevated glutamate levels do not induce neurotoxicity, suggesting that a combination of CaMKII inhibition and elevated extracellular glutamate levels results in neuronal death. In sum, the loss of CaMKII observed with multiple pathological states in the central nervous system, including epilepsy, brain trauma, and ischemia, likely exacerbates programmed cell death by sensitizing vulnerable neuronal populations to excitotoxic glutamate signaling and inducing an excitotoxic insult itself.
Collapse
Affiliation(s)
- Nicole M Ashpole
- Stark Neuroscience Research Institute, Indiana University of School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Hydrogen-rich saline protects retina against glutamate-induced excitotoxic injury in guinea pig. Exp Eye Res 2011; 94:117-27. [PMID: 22154552 DOI: 10.1016/j.exer.2011.11.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 09/30/2011] [Accepted: 11/22/2011] [Indexed: 11/22/2022]
Abstract
Molecular hydrogen (H(2)) is an efficient antioxidant that can selectively reduce hydroxyl radicals and inhibit oxidative stress-induced injuries. We investigated the protective effects and mechanism of hydrogen-rich saline in a glutamate-induced retinal injury model. Retinal excitotoxicity was induced in healthy guinea pigs by injecting glutamate into the vitreous cavity. After 30 min, hydrogen-rich saline was injected into the vitreous cavity, the peritoneal cavity or both. Seven days later, the retinal stress response was evaluated by examining the stress biomarkers, inducible nitric-oxide synthase (iNOS) and glucose-regulated protein 78 (GRP78). The impaired glutamate uptake was assessed by the expression of the excitatory amino acid transporter 1(EAAT-1). The retinal histopathological changes were investigated, focusing on the thicknesses of the entire retina and its inner layer, the number of cells in the retinal ganglion cell layer (GCL) and the ultrastructure of the retinal ganglion cells (RGCs) and glial cells. Compared with the glutamate-induced injury group, the hydrogen-rich saline treatment reduced the loss of cells in the GCL and thinning of the retina and attenuated cellular morphological damage. These improvements were greatest in animals that received H(2) injections into both the vitreous and the peritoneal cavities. The hydrogen-rich saline also inhibited the expression of glial fibrillary acidic protein (GFAP) in Müller cells, CD11b in microglia, and iNOS and GRP78 in glial cells. Moreover, the hydrogen-rich saline increased the expression of EAAT-1. In conclusion, the administration of hydrogen-rich saline through the intravitreal or/and intraperitoneal routes could reduce the retinal excitotoxic injury and promote retinal recovery. This result likely occurs by inhibiting the activation of glial cells, decreasing the production of the iNOS and GRP78 and promoting glutamate clearance.
Collapse
|
15
|
Abstract
Ischemic insults on neurons trigger excessive, pathological glutamate release that causes Ca²⁺ overload resulting in neuronal cell death (excitotoxicity). The Ca²⁺/calmodulin (CaM)-dependent protein kinase II (CaMKII) is a major mediator of physiological excitatory glutamate signals underlying neuronal plasticity and learning. Glutamate stimuli trigger autophosphorylation of CaMKII at T286, a process that makes the kinase "autonomous" (partially active independent from Ca²⁺ stimulation) and that is required for forms of synaptic plasticity. Recent studies suggested autonomous CaMKII activity also as potential drug target for post-insult neuroprotection, both after glutamate insults in neuronal cultures and after focal cerebral ischemia in vivo. However, CaMKII and other members of the CaM kinase family have been implicated in regulation of both neuronal death and survival. Here, we discuss past findings and possible mechanisms of CaM kinase functions in excitotoxicity and cerebral ischemia, with a focus on CaMKII and its regulation.
Collapse
|
16
|
Barber AJ, Gardner TW, Abcouwer SF. The significance of vascular and neural apoptosis to the pathology of diabetic retinopathy. Invest Ophthalmol Vis Sci 2011; 52:1156-63. [PMID: 21357409 DOI: 10.1167/iovs.10-6293] [Citation(s) in RCA: 317] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The most striking features of diabetic retinopathy are the vascular abnormalities that are apparent by fundus examination. There is also strong evidence that diabetes causes apoptosis of neural and vascular cells in the retina. Thus, there is good reason to define diabetic retinopathy as a form of chronic neurovascular degeneration. In keeping with the gradual onset of retinopathy in humans, the rate of cell loss in the animal models is insidious, even in uncontrolled diabetes. This is not surprising given that a sustained high rate of cell loss without regeneration would soon lead to catastrophic tissue destruction. The consequences of ongoing cell death are difficult to detect, and even the quantification of cumulative cell loss requires painstaking histology and microscopy. This subtle cell loss raises the issue of the relevance of the phenomenon to the progression of diabetic retinopathy and the ultimate loss of vision. Neuronal function may be compromised in advance of apoptosis, contributing to an early deterioration of vision. Here we review some of the evidence supporting apoptotic cell death as a contributing mechanism of diabetic retinopathy, explore some of the potential causes, and discuss the potential links between apoptosis and loss of visual function in diabetic retinopathy.
Collapse
Affiliation(s)
- Alistair J Barber
- Department of Ophthalmology, Penn State Hershey Eye Center, Penn State Hershey College of Medicine, Hershey, Pennsylvania 17033, USA.
| | | | | |
Collapse
|
17
|
Schuettauf F, Stein T, Choragiewicz TJ, Rejdak R, Bolz S, Zurakowski D, Varde MA, Laties AM, Thaler S. Caspase inhibitors protect against NMDA-mediated retinal ganglion cell death. Clin Exp Ophthalmol 2011; 39:545-54. [DOI: 10.1111/j.1442-9071.2010.02486.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
18
|
Ashpole NM, Hudmon A. Excitotoxic neuroprotection and vulnerability with CaMKII inhibition. Mol Cell Neurosci 2011; 46:720-30. [PMID: 21316454 DOI: 10.1016/j.mcn.2011.02.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2010] [Revised: 02/02/2011] [Accepted: 02/03/2011] [Indexed: 12/15/2022] Open
Abstract
Aberrant calcium signaling is a common feature of ischemia and multiple neurodegenerative diseases. While activation of calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) is a key event in calcium signaling, its role in excitotoxicity is controversial. Our findings demonstrate neuroprotection in neuronal cultures treated with the small molecule (KN-93) and peptide (tat-AIP and tat-CN21) inhibitors of CaMKII immediately prior to excitotoxic glutamate/glycine insult. Unlike KN-93 which blocks CaMKII activation, but not constitutively active forms of CaMKII, tat-CN21 and tat-AIP significantly reduced excitotoxicity in cultured neurons when applied post-insult. We observed that the neuroprotective effects of tat-CN21 are greatest when applied before the toxic glutamate challenge and diminish with time, with the neuroprotection associated with CaMKII inhibition diminishing back to control 3h post glutamate insult. Mechanistically, tat-CN21 inhibition of CaMKII resulted in an increase in CaMKII activity and the percentage of soluble αCaMKII observed in neuronal lysates 24h following glutamate stimulation. To address the impact of prolonged CaMKII inhibition prior to excitotoxic insult, neuronal cultures were treated with CaMKII inhibitors overnight and then subjected to a sub-maximal excitotoxic insult. In this model, CaMKII inhibition prior to insult exacerbated neuronal death, suggesting that a loss of CaMKII enhances neuronal vulnerability to glutamate. Although changes in αCaMKII or NR2B protein levels are not responsible for this enhanced glutamate vulnerability, this process is blocked by the protein translation inhibitor cycloheximide. In total, the neuroprotection afforded by CaMKII inhibition can be seen as neuroprotective immediately surrounding the excitotoxic insult, whereas sustained CaMKII inhibition produced by excitotoxicity leads to neuronal death by enhancing neuronal vulnerability to glutamate.
Collapse
Affiliation(s)
- Nicole M Ashpole
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
19
|
McMurtrey RJ, Zuo Z. Isoflurane preconditioning and postconditioning in rat hippocampal neurons. Brain Res 2010; 1358:184-190. [PMID: 20709037 PMCID: PMC2949531 DOI: 10.1016/j.brainres.2010.08.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 08/04/2010] [Accepted: 08/06/2010] [Indexed: 01/28/2023]
Abstract
The volatile anesthetic isoflurane is capable of inducing preconditioning and postconditioning effects in the brain. However, the mechanisms for these neuroprotective effects are not fully understood. Here, we showed that rat hippocampal neuronal cultures exposed to 2% isoflurane for 30min at 24h before a 1h oxygen-glucose deprivation (OGD) and a 24h simulated reperfusion had a reduced lactate dehydrogenase release. Similarly, this OGD and simulated reperfusion-induced lactate dehydrogenase release was attenuated by exposing the neuronal cultures to 2% isoflurane for 1h at various times after the onset of the simulated reperfusion (isoflurane postconditioning). The combination of isoflurane preconditioning and postconditioning induced a better neuroprotection than either alone. Inhibition of the calcium/calmodulin-dependent protein kinase II (CaMKII), inhibition of N-methyl d-aspartate (NMDA) receptors, or activation of adenosine A2A receptors resulted in reduction of the OGD and simulated reperfusion-induced cell injury. The combination of CaMKII inhibition and isoflurane preconditioning or postconditioning did not provide better protection than CaMKII inhibition, isoflurane preconditioning, or isoflurane postconditioning alone. The combination of NMDA receptor inhibition and isoflurane postconditioning was not better than NMDA receptor inhibition or isoflurane postconditioning alone for neuroprotection. However, the combination of adenosine A2A receptor activation with either isoflurane preconditioning or isoflurane postconditioning induced a better neuroprotective effect than adenosine A2A receptor activation, isoflurane preconditioning, or isoflurane postconditioning alone. The combination of NMDA receptor inhibition and isoflurane preconditioning caused a better neuroprotective effect than NMDA receptor inhibition or isoflurane preconditioning alone. These results suggest that isoflurane preconditioning- and postconditioning-induced neuroprotection can be additive. Isoflurane preconditioning and isoflurane postconditioning may involve CaMKII inhibition, but may not involve adenosine A2A receptor activation. Inhibition of NMDA receptors may mediate the effects of isoflurane postconditioning, but not isoflurane preconditioning.
Collapse
Affiliation(s)
- Richard J McMurtrey
- Department of Anesthesiology, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
20
|
Vest RS, O'Leary H, Coultrap SJ, Kindy MS, Bayer KU. Effective post-insult neuroprotection by a novel Ca(2+)/ calmodulin-dependent protein kinase II (CaMKII) inhibitor. J Biol Chem 2010; 285:20675-82. [PMID: 20424167 PMCID: PMC2898334 DOI: 10.1074/jbc.m109.088617] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 04/26/2010] [Indexed: 12/31/2022] Open
Abstract
Ca(2+)/calmodulin (CaM)-dependent protein kinase II (CaMKII) is a major mediator of physiological glutamate signaling involved in higher brain functions. Here, we show CaMKII involvement in pathological glutamate signaling relevant in stroke. The novel inhibitor tatCN21 was neuroprotective even when added hours after glutamate insults. By contrast, the "traditional" inhibitor KN93 attenuated excitotoxicity only when present during the insult. Both inhibitors efficiently blocked Ca(2+)/CaM-stimulated CaMKII activity, CaMKII interaction with NR2B and aggregation of CaMKII holoenzymes. However, only tatCN21 but not KN93 blocked the Ca(2+)-independent "autonomous" activity generated by Thr-286 autophosphorylation, the hallmark feature of CaMKII regulation. Mutational analysis further validated autonomous CaMKII activity as the drug target crucial for post-insult neuroprotection. Overexpression of CaMKII wild type but not the autonomy-deficient T286A mutant significantly increased glutamate-induced neuronal death. Maybe most importantly, tatCN21 also significantly reduced infarct size in a mouse stroke model (middle cerebral arterial occlusion) when injected (1 mg/kg intravenously) 1 h after onset of arterial occlusion. Together, these data demonstrate that inhibition of autonomous CaMKII activity provides a promising therapeutic avenue for post-insult neuro-protection after stroke.
Collapse
Affiliation(s)
- Rebekah S. Vest
- From the Department of Pharmacology, University of Colorado Denver School of Medicine, Aurora, Colorado 80045 and
| | - Heather O'Leary
- From the Department of Pharmacology, University of Colorado Denver School of Medicine, Aurora, Colorado 80045 and
| | - Steven J. Coultrap
- From the Department of Pharmacology, University of Colorado Denver School of Medicine, Aurora, Colorado 80045 and
| | - Mark S. Kindy
- the Department of Neurosciences, Medical University of South Carolina and the Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina 29425
| | - K. Ulrich Bayer
- From the Department of Pharmacology, University of Colorado Denver School of Medicine, Aurora, Colorado 80045 and
| |
Collapse
|
21
|
Vest RS, O'Leary H, Bayer KU. Differential regulation by ATP versus ADP further links CaMKII aggregation to ischemic conditions. FEBS Lett 2009; 583:3577-81. [PMID: 19840793 DOI: 10.1016/j.febslet.2009.10.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 09/24/2009] [Accepted: 10/13/2009] [Indexed: 10/20/2022]
Abstract
CaMKII, a major mediator of synaptic plasticity, forms extra-synaptic clusters under ischemic conditions. This study further supports self-aggregation of CaMKII holoenzymes as the underlying mechanism. Aggregation in vitro was promoted by mimicking ischemic conditions: low pH (6.8 or less), Ca(2+) (and calmodulin), and low ATP and/or high ADP concentration. Mutational analysis showed that high ATP prevented aggregation by a mechanism involving T286 auto-phosphorylation, and indicated requirement for nucleotide binding but not auto-phosphorylation also for extra-synaptic clustering within neurons. These results clarify a previously apparent paradox in the nucleotide and phosphorylation requirement of aggregation, and support a mechanism that involves inter-holoenzyme T286-region/T-site interaction.
Collapse
Affiliation(s)
- Rebekah S Vest
- Department of Pharmacology, University of Colorado Denver, Aurora, CO 80045, United States
| | | | | |
Collapse
|
22
|
Abstract
PURPOSE To determine the distribution and glutamate-mediated activation of nuclear factor (NF) kappaB members in the retina and pan-purified retinal ganglion cells (RGCs) and to characterize steps in the signal transduction events that lead to NFkappaB activation. METHODS Retinal expression patterns and RGCs were evaluated for five NFkappaB proteins with the aid of immunohistochemistry. Retinal explants or RGCs were treated with glutamate with or without the presence of the NDMA receptor antagonist memantine, the calcium chelator EGTA, or a specific inhibitor for calcium/calmodulin-dependent protein kinase-II (CaMKII). Characterizations of NFkappaB activation were performed with the aid of electrophoretic mobility shift assays and supershift assays. RESULTS All five NFkappaB proteins were present in the retina and in the pan-purified RGCs. In response to a glutamate stimulus, all NFkappaB proteins except c-Rel were activated. P65 was unique in that it was not constitutively active but showed a glutamate-inducible activation in the retina and in the cultured RGCs. Memantine, EGTA, or autocamtide-2-related inhibitory peptide (AIP) inhibited NFkappaB activation in the retina. Furthermore, AIP significantly reduced the level of glutamate-induced degradation of IkappaBs. CONCLUSIONS These data indicate that glutamate activates distinct NFkappaB proteins in the retina. P65 activation may be especially important with regard to RGC responses to glutamate given that its activity is induced by conditions known to lead to the death of these cells. The NMDA receptor-Ca(2+)-CaMKII signaling pathway is involved in glutamate-induced NFkappaB activation. Because AIP blocks the degradation of IkappaB, its regulation is clearly downstream of CaMKII.
Collapse
Affiliation(s)
- Wei Fan
- Anatomical Sciences and Neurobiology, University of Louisville School of Medicine. 500 S. Preston St., Louisville, Kentucky
| | - Nigel G. F. Cooper
- Anatomical Sciences and Neurobiology, University of Louisville School of Medicine. 500 S. Preston St., Louisville, Kentucky
- Ophthalmology and Visual Sciences, University of Louisville School of Medicine. 500 S. Preston St., Louisville, Kentucky
| |
Collapse
|
23
|
Goebel DJ. Selective blockade of CaMKII-α inhibits NMDA-induced caspase-3-dependent cell death but does not arrest PARP-1 activation or loss of plasma membrane selectivity in rat retinal neurons. Brain Res 2009; 1256:190-204. [DOI: 10.1016/j.brainres.2008.12.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2008] [Revised: 12/12/2008] [Accepted: 12/12/2008] [Indexed: 01/08/2023]
|
24
|
Easley CA, Brown CM, Horwitz AF, Tombes RM. CaMK-II promotes focal adhesion turnover and cell motility by inducing tyrosine dephosphorylation of FAK and paxillin. CELL MOTILITY AND THE CYTOSKELETON 2008; 65:662-74. [PMID: 18613116 PMCID: PMC2830206 DOI: 10.1002/cm.20294] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Transient elevations in Ca2+ have previously been shown to promote focal adhesion disassembly and cell motility through an unknown mechanism. In this study, evidence is provided to show that CaMK-II, a Ca2+/calmodulin dependent protein kinase, influences fibroblast adhesion and motility. TIRF microscopy reveals a dynamic population of CaMK-II at the cell surface in migrating cells. Inhibition of CaMK-II with two mechanistically distinct, membrane permeant inhibitors (KN-93 and myr-AIP) freezes lamellipodial dynamics, accelerates spreading on fibronectin, enlarges paxillin-containing focal adhesions and blocks cell motility. In contrast, constitutively active CaMK-II is not found at the cell surface, reduces cell attachment, eliminates paxillin from focal adhesions and decreases the phospho-tyrosine levels of both FAK and paxillin; all of these events can be reversed with myr-AIP. Thus, both CaMK-II inhibition and constitutive activation block cell motility through over-stabilization or destabilization of focal adhesions, respectively. Coupled with the existence of transient Ca2+ elevations and a dynamic CaMK-II population, these findings provide the first direct evidence that CaMK-II enables cell motility by transiently and locally stimulating tyrosine dephosphorylation of focal adhesion proteins to promote focal adhesion turnover.
Collapse
Affiliation(s)
- Charles A. Easley
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Claire M. Brown
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
- Department of Biochemistry, Life Sciences Complex Imaging Facility, McGill University, Montreal, Quebec, Canada
| | - Alan F. Horwitz
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Robert M. Tombes
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, Virginia
- Department of Biology, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
25
|
Ishida A, Sueyoshi N, Shigeri Y, Kameshita I. Negative regulation of multifunctional Ca2+/calmodulin-dependent protein kinases: physiological and pharmacological significance of protein phosphatases. Br J Pharmacol 2008; 154:729-40. [PMID: 18454172 DOI: 10.1038/bjp.2008.127] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Multifunctional Ca2+/calmodulin-dependent protein kinases (CaMKs) play pivotal roles in intracellular Ca2+ signaling pathways. There is growing evidence that CaMKs are involved in the pathogenic mechanisms underlying various human diseases. In this review, we begin by briefly summarizing our knowledge of the involvement of CaMKs in the pathogenesis of various diseases suggested to be caused by the dysfunction/dysregulation or aberrant expression of CaMKs. It is widely known that the activities of CaMKs are strictly regulated by protein phosphorylation/dephosphorylation of specific phosphorylation sites. Since phosphorylation status is balanced by protein kinases and protein phosphatases, the mechanism of dephosphorylation/deactivation of CaMKs, corresponding to their 'switching off', is extremely important, as is the mechanism of phosphorylation/activation corresponding to their 'switching on'. Therefore, we focus on the regulation of multifunctional CaMKs by protein phosphatases. We summarize the current understanding of negative regulation of CaMKs by protein phosphatases. We also discuss the biochemical properties and physiological significance of a protein phosphatase that we designated as Ca2+/calmodulin-dependent protein kinase phosphatase (CaMKP), and those of its homologue CaMKP-N. Pharmacological applications of CaMKP inhibitors are also discussed. These compounds may be useful not only for exploring the physiological functions of CaMKP/CaMKP-N, but also as novel chemotherapies for various diseases.
Collapse
Affiliation(s)
- A Ishida
- Laboratory of Molecular Brain Science, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan.
| | | | | | | |
Collapse
|
26
|
Cooper NGF, Laabich A, Fan W, Wang X. The relationship between neurotrophic factors and CaMKII in the death and survival of retinal ganglion cells. PROGRESS IN BRAIN RESEARCH 2008; 173:521-40. [PMID: 18929132 DOI: 10.1016/s0079-6123(08)01136-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
The scientific discourse relating to the causes and treatments for glaucoma are becoming reflective of the need to protect and preserve retinal neurons from degenerative changes, which result from the injurious environment associated with this disease. Knowledge, in particular, of the signal transduction pathways which affect death and survival of the retinal ganglion cells is critical to this discourse and to the development of a suitable neurotherapeutic strategy for this disease. The goal of this chapter is to review what is known of the chief suspects involved in initiating the cell death/survival pathways in these cells, and what still remains to be uncovered. The least controversial aspect of the subject relates to the potential role of neurotrophic factors in the protection of the retinal ganglion cells. On the other hand, the postulated triggers for signaling cell death in glaucoma remain controversial. Certainly, the restricted flow of neurotrophic factors has been cited as one possible trigger. However, the connections between glaucoma and other factors present in the retina, such as glutamate, long held to be a prospective culprit in retinal ganglion cell death are still being questioned. Whatever the outcome of this particular debate, it is clear that the downstream intersections between the cell death and survival pathways should provide important foci for future studies whose goal is to protect retinal neurons, situated as they are, in the stressful environment of a cell destroying disease. The evidence for CaMKII being one of these intersecting points is discussed.
Collapse
Affiliation(s)
- N G F Cooper
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40292, USA.
| | | | | | | |
Collapse
|
27
|
Vest RS, Davies KD, O'Leary H, Port JD, Bayer KU. Dual mechanism of a natural CaMKII inhibitor. Mol Biol Cell 2007; 18:5024-33. [PMID: 17942605 PMCID: PMC2096578 DOI: 10.1091/mbc.e07-02-0185] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Revised: 09/19/2007] [Accepted: 10/05/2007] [Indexed: 11/11/2022] Open
Abstract
Ca(2+)/calmodulin (CaM)-dependent protein kinase II (CaMKII) is a major mediator of cellular Ca(2+) signaling. Several inhibitors are commonly used to study CaMKII function, but these inhibitors all lack specificity. CaM-KIIN is a natural, specific CaMKII inhibitor protein. CN21 (derived from CaM-KIIN amino acids 43-63) showed full specificity and potency of CaMKII inhibition. CNs completely blocked Ca(2+)-stimulated and autonomous substrate phosphorylation by CaMKII and autophosphorylation at T305. However, T286 autophosphorylation (the autophosphorylation generating autonomous activity) was only mildly affected. Two mechanisms can explain this unusual differential inhibitor effect. First, CNs inhibited activity by interacting with the CaMKII T-site (and thereby also interfered with NMDA-type glutamate receptor binding to the T-site). Because of this, the CaMKII region surrounding T286 competed with CNs for T-site interaction, whereas other substrates did not. Second, the intersubunit T286 autophosphorylation requires CaM binding both to the "kinase" and the "substrate" subunit. CNs dramatically decreased CaM dissociation, thus facilitating the ability of CaM to make T286 accessible for phosphorylation. Tat-fusion made CN21 cell penetrating, as demonstrated by a strong inhibition of filopodia motility in neurons and insulin secrection from isolated Langerhans' islets. These results reveal the inhibitory mechanism of CaM-KIIN and establish a powerful new tool for dissecting CaMKII function.
Collapse
Affiliation(s)
- Rebekah S. Vest
- Department of Pharmacology, University of Colorado Denver-School of Medicine, Aurora, CO 80045
| | - Kurtis D. Davies
- Department of Pharmacology, University of Colorado Denver-School of Medicine, Aurora, CO 80045
| | - Heather O'Leary
- Department of Pharmacology, University of Colorado Denver-School of Medicine, Aurora, CO 80045
| | - J. David Port
- Department of Pharmacology, University of Colorado Denver-School of Medicine, Aurora, CO 80045
| | - K. Ulrich Bayer
- Department of Pharmacology, University of Colorado Denver-School of Medicine, Aurora, CO 80045
| |
Collapse
|
28
|
Takeda H, Kitaoka Y, Hayashi Y, Kumai T, Munemasa Y, Fujino H, Kobayashi S, Ueno S. Calcium/calmodulin-dependent protein kinase II regulates the phosphorylation of CREB in NMDA-induced retinal neurotoxicity. Brain Res 2007; 1184:306-15. [PMID: 17961520 DOI: 10.1016/j.brainres.2007.09.055] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2007] [Revised: 09/17/2007] [Accepted: 09/17/2007] [Indexed: 01/15/2023]
Abstract
We examined the role of the phosphorylation of calcium/calmodulin-dependent protein kinase II (CaMKII) and cyclic AMP-response element binding protein (CREB) in N-methyl-d-aspartate (NMDA)-induced neurotoxicity in the rat retina. Western blot analysis showed early elevation of phosphorylated CaMKII (p-CaMKII) protein levels and subsequential elevation of phosphorylated CREB (p-CREB) protein after NMDA injection. Immunohistochemistry showed that p-CaMKII was colocalized with Thy-1-positive retinal ganglion cells (RGCs) after NMDA injection. The increase in the p-CaMKII protein level was significantly inhibited by the preinjection of CaMKII small interfering RNA (siRNA), whereas negative control siRNA did not affect. Moreover, the increase in the p-CREB protein level after NMDA injection was also prevented by preinjection of CaMKII siRNA. In addition, our morphometric study of neurotracer retrograde labeling and Thy-1-positive cells showed that CaMKII siRNA significantly accelerated NMDA-induced RGC loss. Furthermore, the prevention of CREB binding by CRE decoy oligonucleotide also exacerbated RGC loss. These results suggest that the activation of CaMKII may regulate CREB phosphorylation and that the transient phosphorylation of CaMKII and CREB may be a neuroprotective response against NMDA-induced neurotoxicity.
Collapse
Affiliation(s)
- Hiroyuki Takeda
- Department of Ophthalmology, St Marianna University School of Medicine, Kawasaki-Shi, Kanagawa, Japan
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Fan W, Agarwal N, Cooper NGF. The role of CaMKII in BDNF-mediated neuroprotection of retinal ganglion cells (RGC-5). Brain Res 2005; 1067:48-57. [PMID: 16337157 DOI: 10.1016/j.brainres.2005.10.030] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2005] [Revised: 09/05/2005] [Accepted: 10/09/2005] [Indexed: 12/01/2022]
Abstract
The purpose of the study is to determine if expression or secretion of brain-derived neurotrophic factor (BDNF) in retinal ganglion cells (RGC-5) is mediated by NFkappaB or Ca2+/calmodulin-dependent protein kinase II (CaMKII). RGC-5 cells were exposed to 1 mM glutamate for various periods of time, in the presence or absence of prospective regulatory molecules. BDNF mRNA and protein expression were assessed with the aid of real-time PCR and immunoblots, respectively, and BDNF secretion was determined by ELISA. The NFkappaB inhibitor (TLCK and PTD-p65), or a specific CaMKII inhibitor (m-AIP), was used to study association of NFkappaB or CaMKII with BDNF expression/secretion in RGC-5 cells. Glutamate stimulated a transient increase in BDNF mRNA and protein in RGC-5 cells, and also stimulated an early release of BDNF into the culture media. Neutralizing the BDNF or blocking the TrkB receptor enhanced the glutamate-induced cytotoxicity. NFkappaB nuclear translocation was revealed in response to glutamate treatment. Application of TLCK or PTD-p65 inhibited the glutamate-induced BDNF expression and secretion. Inhibition of CaMKII by m-AIP did not affect expression but significantly enhanced the release of BDNF from glutamate challenged cells. Our data suggest that glutamate treatment may stimulate expression of BDNF in RGC-5 cells through NFkappaB activation. A novel mechanism for neuroprotection is proposed for the CaMKII inhibitor, AIP, which appears to protect RGC-5 cells from cytotoxicity by enhancing the release of BDNF from glutamate challenged cells.
Collapse
Affiliation(s)
- Wei Fan
- Department of Anatomical Sciences and Neurobiology, 500 S. Preston St., Louisville, KY 40292, USA
| | | | | |
Collapse
|
30
|
Fan W, Agarwal N, Kumar MD, Cooper NGF. Retinal ganglion cell death and neuroprotection: Involvement of the CaMKIIα gene. ACTA ACUST UNITED AC 2005; 139:306-16. [PMID: 16023257 DOI: 10.1016/j.molbrainres.2005.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2004] [Revised: 05/26/2005] [Accepted: 06/08/2005] [Indexed: 10/25/2022]
Abstract
The purpose of this study is to determine if calcium/calmodulin-dependent protein kinase-II (CaMKII) plays a role in neuronal cell death and if inhibition of this kinase affords some neuroprotection in the RGC-5 retinal ganglion cell line. The RGC-5 cells were treated with glutamate at various concentrations for increasing increments of time. Cytotoxicity was assayed by measuring the lactate dehydrogenase (LDH) leakage from non-viable cells and TUNEL assays. The involvement of caspase-3, Bcl-2 and caspase-8 in glutamate-induced cytotoxicity was determined by immunoblots and/or real time RT-PCR. In addition, the autocamtide-2-related inhibitory peptide (AIP), a specific inhibitor of CaMKII, was used to determine the involvement of CaMKII in glutamate-induced RGC-5 cell death. Application of increasing concentrations of glutamate to RGC-5 cells caused a dose-dependent increase in the level of cell death after 24 h. There was a glutamate-stimulated increase in the expression of caspase-8 and caspase-3 and a corresponding decrease in Bcl-2. The active fragment of caspase-3 increased in glutamate-treated cells. An early transient increase in the expression of CaMKIIalpha(B) gene and a corresponding CaMKIIalpha nuclear translocation was found in glutamate-treated cells. Treatment with AIP blocked the activation of caspase-3 and protected RGC from glutamate-mediated cell death but did not alter the glutamate-enhanced expression levels of caspase-8 or caspase-3. This report shows the likely involvement of a transcript of the CaMKIIalpha gene in the cytotoxicity response of RGC-5 cells similar to previous reports in the neural retina. AIP is shown to be a neuroprotectant for RGC-5 cells as was reported for the neural retina.
Collapse
Affiliation(s)
- Wei Fan
- Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, 500 S. Preston Street, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
31
|
Barnett NL, Grozdanic SD. Glutamate transporter localization does not correspond to the temporary functional recovery and late degeneration after acute ocular ischemia in rats. Exp Eye Res 2004; 79:513-24. [PMID: 15381035 DOI: 10.1016/j.exer.2004.06.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2004] [Accepted: 06/22/2004] [Indexed: 01/26/2023]
Abstract
PURPOSE To determine whether the localization of retinal glutamate transporters is affected by retinal ischaemia and whether their ability to transport glutamate decreases with the progression of ischemic retinal and optic nerve degeneration. METHODS Retinal ischemia was induced in rats by acutely increasing the intraocular pressure (IOP, 110 mmHg/60 min). Reperfusion was permitted for periods up to 60 days post-ischemia. Functional evaluation was performed by monitoring the pupil light reflexes (PLRs) and electroretinograms (flash, flicker ERG and oscillatory potentials). Glutamate transporter localization and D-aspartate (glutamate analogue) uptake were assessed by immunohistochemistry. RESULTS Intense immunoreactivity for the retinal glutamate transporters (GLAST, GLT1, EAAC1 and EAAT5) was observed at all time points after the insult, despite severe retinal degeneration. D-aspartate was also normally accumulated in the ischemic retinas. Ten days post-operatively the PLR ratio (ratio=indirect/direct PLR=34+/-7.5%) was significantly less than the pre-operative value (pre-op=76.7+/-2.6%, p<0.05). However, 25 and 35 days post-operatively PLR ratios did not differ significantly from pre-operative values (44.4+/-6.9 and 53.8+/-9.6%, p>0.05). Forty-five and 60 days post-operatively the PLR ratio declined again and was significantly lower than the pre-operative value (33.8+8.7 and 26.2+8.9%, p<0.05). Statistical analysis revealed that all tested ERG components had significantly higher values at 32, but not at 42 and 58 days post-operatively when compared to the first time point recorded post-operatively (10 days). CONCLUSIONS While retinal glutamate transport is compromised during an acute ischemic insult, consequent retinal recovery and degeneration are not due to a change in the excitatory amino acid transporter localization or D-aspartate (glutamate analogue) uptake. Rat retina and optic nerve are capable of spontaneous, but temporary, functional recovery after an acute ischemic insult.
Collapse
Affiliation(s)
- Nigel L Barnett
- Vision, Touch and Hearing Research Centre, School of Biomedical Sciences, The University of Queensland, Brisbane, Qld 4072, Australia.
| | | |
Collapse
|
32
|
Ishida A, Shigeri Y, Taniguchi T, Kameshita I. Protein phosphatases that regulate multifunctional Ca2+/calmodulin-dependent protein kinases: from biochemistry to pharmacology. Pharmacol Ther 2004; 100:291-305. [PMID: 14652114 DOI: 10.1016/j.pharmthera.2003.09.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Multifunctional Ca(2+)/calmodulin-dependent protein kinases (CaMKs) play pivotal roles in Ca(2+) signaling pathways, such as the regulation of the neuronal functions of learning, memory, and neuronal cell death. The activities of the kinases are strictly regulated by protein phosphorylation/dephosphorylation. Although the activation mechanisms for multifunctional CaMKs through phosphorylation, which correspond to "switch on," have been extensively studied, the negative regulatory mechanisms through dephosphorylation, which correspond to "switch off," have not. In this review, we focused on the regulation of multifunctional CaMKs by the protein phosphatases responsible. We first summarized the current understanding of negative regulation of CaMKs by known protein phosphatases and their physiological significance. We then discussed newly developed methods for detection of protein phosphatases involved in the regulation of CaMKs. We also summarized the biochemical properties of a novel protein phosphatase, which we isolated with the new methods and designated as CaMK phosphatase (CaMKP), and its homologue. Pharmacological implications for neuronal functions including memory and neuronal cell death are discussed from the viewpoint that regulation of protein kinase activity can be elucidated by focusing on protein phosphatases involved in its "switch off" mechanism.
Collapse
Affiliation(s)
- Atsuhiko Ishida
- Department of Biochemistry, Asahikawa Medical College, Asahikawa, 078-8510, Japan.
| | | | | | | |
Collapse
|
33
|
Inomata Y, Hirata A, Koga T, Kimura A, Singh DP, Shinohara T, Tanihara H. Lens epithelium-derived growth factor: neuroprotection on rat retinal damage induced by N-methyl-D-aspartate. Brain Res 2004; 991:163-70. [PMID: 14575888 DOI: 10.1016/j.brainres.2003.08.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The purpose of this study is to investigate possible neuroprotective effects of lens epithelium-derived growth factor (LEDGF) against cell death induced by N-methyl-D-aspartate (NMDA) in the rat retina. LEDGF and/or NMDA were intravitreally injected into rat eyes. NMDA-induced retinal death and protective effects of LEDGF were evaluated by morphometric analysis, cell numbers in the ganglion cell layer (GCL) and the thickness of the inner plexiform layer (IPL). Retrograde labeling with a fluorescent tracer (Fluoro-Gold) was applied for counting retinal ganglion cells (RGCs) that survived after NMDA injection. Terminal deoxyribonucleotidyl transferase (TdT)-mediated fluroscein-16-dUTP nick end-labeling (TUNEL) staining was used to evaluate of retinal cell death. Morphometric analysis and retrograde labeling analysis showed that retinal damage induced by NMDA was protected significantly by LEDGF. TUNEL assay revealed that pretreatment with LEDGF prevents NMDA-induced apoptosis. Retinal damage (ganglion and amacrine cells) induced by NMDA was protected by an intravitreal injection of LEDGF.
Collapse
Affiliation(s)
- Yasuya Inomata
- Department of Ophthalmology and Visual Science, Kumamoto University Graduate School of Medical Sciences, 1-1-1, Honjo, Kumamoto 860-0811, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Moncaster JA, Walsh DT, Gentleman SM, Jen LS, Aruoma OI. Ergothioneine treatment protects neurons against N-methyl-D-aspartate excitotoxicity in an in vivo rat retinal model. Neurosci Lett 2002; 328:55-9. [PMID: 12123858 DOI: 10.1016/s0304-3940(02)00427-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Injection of the glutamate agonist N-methyl-D-aspartate into the vitreous body of the rat eye resulted in a number of morphological changes in the retina. Most apparent was a dramatic reduction in the density and sizes of neurons accompanied by a decrease in amyloid precursor protein and glial fibrillary acidic protein immunoreactivity. Cell counts revealed that 81% of ganglion cells and 43% of non-ganglion cells were lost as a result of the treatment. However, in animals treated with the antioxidant ergothioneine, these figures dropped to 44 and 31%, respectively. Thus, ergothioneine appears to be neuroprotective in this system and the data suggest that antioxidants may provide a useful means of modulating glutamate-based toxicity.
Collapse
Affiliation(s)
- Juliet A Moncaster
- Department of Neuroinflammation, Division of Neuroscience and Psychological Medicine, Faculty of Medicine, Imperial College of Science, Technology and Medicine, Charing Cross Hospital, Fulham Palace Road, London W6 8RF, UK
| | | | | | | | | |
Collapse
|
35
|
Laabich A, Li G, Cooper NG. Characterization of apoptosis-genes associated with NMDA mediated cell death in the adult rat retina. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2001; 91:34-42. [PMID: 11457490 DOI: 10.1016/s0169-328x(01)00116-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Calcium/calmodulin-dependent protein kinase II containing a nuclear localizing signal (CaMKII-alphaB) is altered in retinal neurons exposed to N-methyl-D-aspartate (NMDA). AIP (myristoylated autocamtide-2-related inhibitory peptide), a specific inhibitor of CaMKII provides neuroprotection against NMDA-mediated neurotoxicity. In this study, gene-arrays were used to investigate which apoptosis-associated genes are altered after exposure to NMDA. The data indicate an increased expression (2-7-fold) of five such genes encoding proteins that could be involved in NMDA induced cell death. The up-regulated genes are: FasL; GADD45; GADD153; Nur77 and TNF-R1. Treatment with AIP blocked their altered expression. The results suggest that multiples genes are involved in NMDA-induced excitotoxicity and that AIP, a specific inhibitor for CaMKII, regulates the expression of these apoptosis-associated genes in the retina.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Apoptosis/drug effects
- Apoptosis/genetics
- CCAAT-Enhancer-Binding Proteins/genetics
- DNA-Binding Proteins/genetics
- Excitatory Amino Acid Agonists/toxicity
- Fas Ligand Protein
- Female
- Gene Expression/drug effects
- Gene Expression/physiology
- Intracellular Signaling Peptides and Proteins
- Membrane Glycoproteins/genetics
- N-Methylaspartate/toxicity
- Neurons
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Oligonucleotide Array Sequence Analysis
- Peptides/pharmacology
- Proteins/genetics
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear
- Receptors, Steroid
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type I
- Retina/pathology
- Retina/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription Factor CHOP
- Transcription Factors/genetics
- Transcription, Genetic/drug effects
- Transcription, Genetic/physiology
- GADD45 Proteins
Collapse
Affiliation(s)
- A Laabich
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, 500 South Preston St., Louisville, KY 40202, USA
| | | | | |
Collapse
|