1
|
Schettler F, Gattor AO, Koch P, Keller M. Characterization of [ 3H]Propionylated Human Peptide YY-A New Probe for Neuropeptide Y Y 2 Receptor Binding Studies. ACS Pharmacol Transl Sci 2025; 8:785-799. [PMID: 40109743 PMCID: PMC11915035 DOI: 10.1021/acsptsci.4c00666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/07/2025] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
The neuropeptide Y (NPY) Y2 receptor (Y2R) is a G-protein-coupled receptor that is involved in the regulation of various physiological processes such as neurotransmitter release, bone metabolism, and memory. Consequently, the Y2R represents a potential drug target, e.g., for the treatment of epilepsy and mood disorders. Until now, the determination of the Y2R binding affinities of Y2R ligands has primarily been performed using 125I-labeled derivatives of the endogenous Y2R agonists NPY and peptide YY (PYY). A tritium-labeled NPY derivative has also been used; however, its suitability for binding assays in sodium-containing buffer is doubtful. We synthesized a tritium-labeled PYY derivative by [3H]propionylation at Lys4 ([3H]2). The radioligand was characterized by saturation binding, association, and dissociation kinetics and was applied in competition binding assays. Specific binding of [3H]2 at intact Chinese hamster ovary cells expressing the hY2R was saturable in both sodium-free buffer (apparent K d = 0.016-0.067 nM) and sodium-containing buffer (175 mM Na+, apparent K d = 0.16-0.18 nM). Competition binding experiments with Y2R reference ligands yielded K i values, which are in good agreement with the reported Y2R binding affinities, showing that [3H]2 represents a useful tritiated tool compound for the determination of Y2R binding affinities also in buffers containing sodium at physiological concentrations.
Collapse
Affiliation(s)
- Franziska Schettler
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93040, Germany
| | - Albert O Gattor
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93040, Germany
| | - Pierre Koch
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93040, Germany
| | - Max Keller
- Institute of Pharmacy, Faculty of Chemistry and Pharmacy, University of Regensburg, Universitätsstraße 31, Regensburg D-93040, Germany
| |
Collapse
|
2
|
Liu Q, Yan R, Wang L, Li R, Zhang D, Liao C, Mao S. Alpha-asarone alleviates cutaneous hyperalgesia by inhibiting hyperexcitability and neurogenic inflammation via TLR4/NF-κB/NLRP3 signaling pathway in a female chronic migraine rat model. Neuropharmacology 2024; 261:110158. [PMID: 39276863 DOI: 10.1016/j.neuropharm.2024.110158] [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: 05/15/2024] [Revised: 06/28/2024] [Accepted: 09/10/2024] [Indexed: 09/17/2024]
Abstract
Migraine is a highly prevalent neurological disorder. Alpha-asarone (ASA), a major active component found in Acorus tatarinowii, plays a crucial role in analgesia and anti-inflammation for neuropathic pain. This study aimed to assess the efficacy of ASA against migraine and elucidate its potential mechanisms using a well-established inflammatory soup (IS) migraine female rat model. Mechanical pain thresholds were assessed daily before IS infusion, followed by post-infusion administration of ASA. Subsequently, spontaneous locomotor activities, exploratory behavior, short-term spatial memory, and photophobia were blindly evaluated after the final drug administration. The rats were then sacrificed for investigation into the underlying mechanisms of action. Network pharmacology was also employed to predict potential targets and pathways of ASA against migraine. The anti-inflammatory activity of ASA and pathway-related proteins were examined in BV2 cells stimulated with lipopolysaccharides (LPS). The results demonstrated that ASA ameliorated cutaneous hyperalgesia and photophobia while improving spatial memory and increasing exploratory behavior in IS rats. ASA attenuated central sensitization-related indicators and excessive glutamate levels while enhancing GABA synthesis. ASA rescued neuronal loss in the cortex and hippocampus of IS rats. Notably, the ability of ASA to improve spatial memory performance in the Y maze test was not observed with sumatriptan, a first-line treatment drug, suggesting the potential involvement of the TLR4 pathway. Moreover, ASA suppressed microglial activation, reduced pro-inflammatory factors, and downregulated TLR4, MyD88, p-NF-κB/NF-κB, NLRP3, caspase-1, IL-1β, and IL-18. Overall, ASA demonstrated its potential to alleviate hyperalgesia and improve behavioral performance in migraine rats by inhibiting hyperexcitability and microglia-related inflammation.
Collapse
Affiliation(s)
- Qi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Ruijie Yan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Ling Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Rui Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Di Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Can Liao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Shengjun Mao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
3
|
Reich N, Hölscher C. Cholecystokinin (CCK): a neuromodulator with therapeutic potential in Alzheimer's and Parkinson's disease. Front Neuroendocrinol 2024; 73:101122. [PMID: 38346453 DOI: 10.1016/j.yfrne.2024.101122] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/04/2024] [Accepted: 01/25/2024] [Indexed: 02/16/2024]
Abstract
Cholecystokinin (CCK) is a neuropeptide modulating digestion, glucose levels, neurotransmitters and memory. Recent studies suggest that CCK exhibits neuroprotective effects in Alzheimer's disease (AD) and Parkinson's disease (PD). Thus, we review the physiological function and therapeutic potential of CCK. The neuropeptide facilitates hippocampal glutamate release and gates GABAergic basket cell activity, which improves declarative memory acquisition, but inhibits consolidation. Cortical CCK alters recognition memory and enhances audio-visual processing. By stimulating CCK-1 receptors (CCK-1Rs), sulphated CCK-8 elicits dopamine release in the substantia nigra and striatum. In the mesolimbic pathway, CCK release is triggered by dopamine and terminates reward responses via CCK-2Rs. Importantly, activation of hippocampal and nigral CCK-2Rs is neuroprotective by evoking AMPK activation, expression of mitochondrial fusion modulators and autophagy. Other benefits include vagus nerve/CCK-1R-mediated expression of brain-derived neurotrophic factor, intestinal protection and suppression of inflammation. We also discuss caveats and the therapeutic combination of CCK with other peptide hormones.
Collapse
Affiliation(s)
- Niklas Reich
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK; Faculty of Health and Medicine, Biomedical & Life Sciences Division, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Christian Hölscher
- Second associated Hospital, Neurology Department, Shanxi Medical University, Taiyuan, Shanxi, China; Henan Academy of Innovations in Medical Science, Neurodegeneration research group, Xinzhen, Henan province, China
| |
Collapse
|
4
|
Liu M, Liu H, Feng F, Krook-Magnuson E, Dudley SC. TRPM7 kinase mediates hypomagnesemia-induced seizure-related death. Sci Rep 2023; 13:7855. [PMID: 37188671 DOI: 10.1038/s41598-023-34789-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 05/08/2023] [Indexed: 05/17/2023] Open
Abstract
Hypomagnesemia (HypoMg) can cause seizures and death, but the mechanism is unknown. Transient receptor potential cation channel subfamily M 7 (TRPM7) is a Mg transporter with both channel and kinase function. In this study, we focused on the kinase role of TRPM7 in HypoMg-induced seizures and death. Wild type C57BL/6J mice and transgenic mice with a global homozygous mutation in the TRPM7 kinase domain (TRPM7K1646R, with no kinase function) were fed with control diet or a HypoMg diet. After 6 weeks of HypoMg diet, mice had significantly decreased serum Mg, elevated brain TRPM7, and a significant rate of death, with females being most susceptible. Deaths were immediately preceded by seizure events. TRPM7K1646R mice showed resistance to seizure-induced death. HypoMg-induced brain inflammation and oxidative stress were suppressed by TRPM7K1646R. Compared to their male counterparts, HypoMg female mice had higher levels of inflammation and oxidative stress in the hippocampus. We concluded that TRPM7 kinase function contributes seizure-induced deaths in HypoMg mice and that inhibiting the kinase reduced inflammation and oxidative stress.
Collapse
Affiliation(s)
- Man Liu
- Cardiovascular Division, Department of Medicine, The Lillehei Heart Institute, University of Minnesota at Twin Cities, 2231 6th Street SE, CCRB 4-141, Minneapolis, MN, 55455, USA
| | - Hong Liu
- Cardiovascular Division, Department of Medicine, The Lillehei Heart Institute, University of Minnesota at Twin Cities, 2231 6th Street SE, CCRB 4-141, Minneapolis, MN, 55455, USA
| | - Feng Feng
- Cardiovascular Division, Department of Medicine, The Lillehei Heart Institute, University of Minnesota at Twin Cities, 2231 6th Street SE, CCRB 4-141, Minneapolis, MN, 55455, USA
| | - Esther Krook-Magnuson
- Department of Neuroscience, University of Minnesota at Twin Cities, Minneapolis, MN, USA
| | - Samuel C Dudley
- Cardiovascular Division, Department of Medicine, The Lillehei Heart Institute, University of Minnesota at Twin Cities, 2231 6th Street SE, CCRB 4-141, Minneapolis, MN, 55455, USA.
| |
Collapse
|
5
|
Lee RHC, Wu CYC, Citadin CT, Couto E Silva A, Possoit HE, Clemons GA, Acosta CH, de la Llama VA, Neumann JT, Lin HW. Activation of Neuropeptide Y2 Receptor Can Inhibit Global Cerebral Ischemia-Induced Brain Injury. Neuromolecular Med 2022; 24:97-112. [PMID: 34019239 PMCID: PMC8606017 DOI: 10.1007/s12017-021-08665-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022]
Abstract
Cardiopulmonary arrest (CA) can greatly impact a patient's life, causing long-term disability and death. Although multi-faceted treatment strategies against CA have improved survival rates, the prognosis of CA remains poor. We previously reported asphyxial cardiac arrest (ACA) can cause excessive activation of the sympathetic nervous system (SNS) in the brain, which contributes to cerebral blood flow (CBF) derangements such as hypoperfusion and, consequently, neurological deficits. Here, we report excessive activation of the SNS can cause enhanced neuropeptide Y levels. In fact, mRNA and protein levels of neuropeptide Y (NPY, a 36-amino acid neuropeptide) in the hippocampus were elevated after ACA-induced SNS activation, resulting in a reduced blood supply to the brain. Post-treatment with peptide YY3-36 (PYY3-36), a pre-synaptic NPY2 receptor agonist, after ACA inhibited NPY release and restored brain circulation. Moreover, PYY3-36 decreased neuroinflammatory cytokines, alleviated mitochondrial dysfunction, and improved neuronal survival and neurological outcomes. Overall, NPY is detrimental during/after ACA, but attenuation of NPY release via PYY3-36 affords neuroprotection. The consequences of PYY3-36 inhibit ACA-induced 1) hypoperfusion, 2) neuroinflammation, 3) mitochondrial dysfunction, 4) neuronal cell death, and 5) neurological deficits. The present study provides novel insights to further our understanding of NPY's role in ischemic brain injury.
Collapse
Affiliation(s)
- Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Cristiane T Citadin
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alexandre Couto E Silva
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Harlee E Possoit
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Garrett A Clemons
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Christina H Acosta
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Victoria A de la Llama
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA
| | - Jake T Neumann
- Department of Biomedical Sciences, West Virginia School of Osteopathic Medicine, Lewisburg, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Sciences Center, 1501 Kings Hwy, Shreveport, USA.
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, USA.
| |
Collapse
|
6
|
Cheon M, Park H, Chung C. Protein kinase C mediates neuropeptide Y-induced reduction in inhibitory neurotransmission in the lateral habenula. Neuropharmacology 2020; 180:108295. [PMID: 32882226 DOI: 10.1016/j.neuropharm.2020.108295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/06/2020] [Accepted: 08/29/2020] [Indexed: 11/30/2022]
Abstract
Neuropeptide Y (NPY) is one of peptide neuromodulators, well known for orexigenic, anxiolytic and antidepressant effects. We previously reported that NPY decreases GABAergic transmission in the lateral habenula (LHb). In the current study, we aim to investigate the underlying signaling pathways that mediate inhibitory action of NPY in the LHb by employing whole-cell patch clamp recording with pharmacological interventions. Here, we revealed that Y1 receptors (Y1Rs) but not Y2Rs mediate NPY-induced decrease of GABAergic transmission in the LHb. Surprisingly, NPY-induced decrease of inhibitory transmission in the LHb was not dependent on adenylyl cyclase (AC)/protein kinase A (PKA)-dependent pathway as reported in other brain areas. Instead, pharmacological blockade of phospholipase C (PLC) or protein kinase C (PKC) activity abolished the decrease of GABAergic transmission by NPY in the LHb. Our findings suggest that Y1Rs in the LHb may trigger the activation of PLC/PKC-dependent pathway but not the classical AC/PKA-dependent pathway to decrease inhibitory transmission of the LHb.
Collapse
Affiliation(s)
- Myunghyun Cheon
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea
| | - Hoyong Park
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea
| | - ChiHye Chung
- Department of Biological Sciences, Konkuk University, Seoul, 05029, South Korea.
| |
Collapse
|
7
|
Ventura F, Muga M, Coelho-Santos V, Fontes-Ribeiro CA, Leitão RA, Silva AP. Protective effect of neuropeptide Y2 receptor activation against methamphetamine-induced brain endothelial cell alterations. Toxicol Lett 2020; 334:53-59. [PMID: 32956829 DOI: 10.1016/j.toxlet.2020.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 09/09/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022]
Abstract
Methamphetamine (METH) consumption is a health problem that leads to neurological and psychiatric disturbances. The cellular alterations behind these conditions have been extensively investigated and it is now well-established that METH causes cerebrovascular alterations being a key feature in drug-induced neuropathology. Although promising advances in understanding the blood-brain barrier (BBB) alterations induced by METH, there is still no available approach to counteract or diminish such effects. Interestingly, several studies show that neuropeptide Y (NPY) has an important protective role against METH-induced neuronal and glial toxicity, as well as behavioral deficits. Despite these beneficial effects of the NPY system, nothing is known about its role in brain endothelial cells under conditions of METH exposure. Thus, our aim was to unravel the effect of NPY and its receptors against METH-induced endothelial cell dysfunction. For that, we used a human brain microvascular endothelial cell line (hCMEC/D3) and our results demonstrate that endothelial cells express both NPY Y1 (Y1R) and Y2 (Y2R) receptors, but only Y2R is upregulated after METH exposure. Moreover, this drug of abuse induced endothelial cell death and elicited the production of reactive oxygen species (ROS) by these cells, which were prevented by the activation of Y2R. Additional, cell death and oxidative stress triggered by METH were dependent on the concentration of the drug. In sum, with the present study we identified for the first time the NPY system, and particularly the Y2R subtype, as a promising target to protect against METH-induced neurovascular dysfunction.
Collapse
Affiliation(s)
- Fabiana Ventura
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal; Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Mariana Muga
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Vanessa Coelho-Santos
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Carlos A Fontes-Ribeiro
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ricardo A Leitão
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal
| | - Ana Paula Silva
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Portugal; Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Portugal; Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
8
|
Zeng X, Niu Y, Qin G, Zhang D, Zhou J, Chen L. Deficiency in the function of inhibitory interneurons contributes to glutamate-associated central sensitization through GABABR2-SynCAM1 signaling in chronic migraine rats. FASEB J 2020; 34:14780-14798. [PMID: 32931071 DOI: 10.1096/fj.202001561r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/11/2020] [Accepted: 08/24/2020] [Indexed: 01/03/2023]
Abstract
The occurrence of pain has always been closely related to a break in the balance between excitatory and inhibitory systems, and the internal relationship between these two systems has not been studied in the pathogenesis of chronic migraine (CM). In this study, we explored how inhibitory interneurons specifically modulate the glutamate-induced hyperexcitability in the periaqueductal gray (PAG) of CM rats. The CM model was established by repeated dural infusion of inflammatory soup (IS) in rats. Then, Baclofen, a gamma-aminobutyric acid type B receptor (GABABR) agonist; CGP35348, a GABABR antagonist; H89, a protein kinase A (PKA) inhibitor; and 8-Bromo-cAMP, a PKA agonist, were applied by intraventricular injection to investigate the detailed CM mechanism. Our results showed that GABABR2 mRNA and protein levels were significantly downregulated (P < .01) in the PAG of CM rats. Similarly, gamma-aminobutyric acid (GABA) and its synthetase glutamate decarboxylase 65/67 (GAD65/67) seriously decreased (P < .01), implying a deficit in the function of inhibitory interneurons in the PAG of CM rats. Afterward, the application of Baclofen and H89 alleviated the IS-evoked hyperalgesia and extenuated vesicular glutamate transporter 2 (VGLUT2), glutamate, calcitonin gene-related peptide (CGRP), and c-Fos expression by regulating the GABABR2/PKA/SynCAM1 pathway in the PAG of CM rats, while the application of CGP35348 and 8-Bromo-cAMP exactly exerted the opposite effect. Importantly, CGP35348 induced an elevation of CGRP, and VGLUT2 expression was relieved by H89. These data suggest that the loss in the function of inhibitory interneurons contributes to glutamate-associated central sensitization through the GABABR2/PKA/SynCAM1 pathway in the PAG of CM rats.
Collapse
Affiliation(s)
- Xiaoxu Zeng
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yingying Niu
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guangcheng Qin
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dunke Zhang
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiying Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lixue Chen
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Wang A, Si Z, Li X, Lu L, Pan Y, Liu J. FK506 Attenuated Pilocarpine-Induced Epilepsy by Reducing Inflammation in Rats. Front Neurol 2019; 10:971. [PMID: 31572289 PMCID: PMC6751399 DOI: 10.3389/fneur.2019.00971] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022] Open
Abstract
Background: The status epilepticus (SE) is accompanied by a local inflammatory response and many oxygen free radicals. FK506 is an effective immunosuppressive agent with neuroprotective and neurotrophic effects, however, whether it can inhibit the inflammatory response and attenuate epilepsy remains unclear. Objective: This study aims to clarify the effect of FK506 on inflammatory response in rats with epilepsy. Methods: A total of 180 rats were randomly and equally divided into the control group, epilepsy group, and FK506 group. The rat SE model in the epilepsy group and FK506 group was induced by lithium chloride combined with pilocarpine. In the FK506 group, FK506 was given before the injection of pilocarpine. The control group was given the same volume of saline. Then the effect of FK506 on epilepsy in rats and the changes of inflammatory factors and free radicals in hippocampus were examined using hematoxylin and eosin (HE) staining, immunohistochemistry, quantitative real-time polymerase chain reaction (qRT-PCR), and western blotting. Results: FK506 ameliorated the course of pilocarpine-induced epilepsy and the neuronal loss in the rat hippocampus after SE. FK506 reduced the increased content of nitric oxide (NO), superoxide dismutase (SOD), and malondialdehyde (MDA) in the hippocampus after SE. Besides, FK506 also significantly reduced the levels of factors involved in inflammatory response such as vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), tumor necrosis factor-α (TNF-α), and Protein Kinase C δ (PKCδ) that rise after epilepsy. Conclusion: FK506 ameliorated the course of pilocarpine-induced epilepsy, significantly reduced free radical content, and inhibited the expression of inflammatory factors, which provided a theoretical basis for the application of FK506 in the treatment of epilepsy.
Collapse
Affiliation(s)
- Aihua Wang
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China.,Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Zhihua Si
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China.,Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Xiaolin Li
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China.,Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Lu Lu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China.,Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Yongli Pan
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China.,Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Jinzhi Liu
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated With Shandong First Medical University, Jinan, China.,Department of Neurology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| |
Collapse
|
10
|
Campos EJ, Martins J, Brudzewsky D, Correia S, Santiago AR, Woldbye DP, Ambrósio AF. Impact of type 1 diabetes mellitus and sitagliptin treatment on the neuropeptide Y system of rat retina. Clin Exp Ophthalmol 2018; 46:783-795. [PMID: 29442423 DOI: 10.1111/ceo.13176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 02/06/2018] [Accepted: 02/10/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Neuropeptide Y (NPY) is a neuromodulator that is expressed in the retina. Increasing evidence suggests that NPY has pronounced anti-inflammatory effects, which might depend on the inhibition of dipeptidyl-peptidase-IV (DPP-IV). The aim of this study was to investigate the impact of type 1 diabetes mellitus (DM) and sitagliptin, a DPP-IV inhibitor, on the NPY system in the retina using an animal model. METHODS Type 1 DM was induced in male Wistar rats by an intraperitoneal injection of streptozotocin. Starting 2 weeks after DM onset, animals were treated orally with sitagliptin (5 mg/kg.day) for 2 weeks. The expression of NPY and NPY receptors (Y1 , Y2 and Y5 receptors) was measured by quantitative polymerase chain reaction, Western blot and/or enzyme-linked immunosorbent assay. The immunoreactivity of NPY and NPY receptors was evaluated by immunohistochemistry, and the [35 S]GTPγS binding assay was used to assess the functional binding of NPY receptors. RESULTS DM decreased the mRNA levels of NPY in the retina, as well as the protein levels of NPY and Y5 receptor. No changes were detected in the localization of NPY and NPY receptors in the retina and in the functional binding of NPY to all receptors. Sitagliptin alone reduced retinal NPY mRNA levels. The effects of DM on the NPY system were not affected by sitagliptin. CONCLUSION DM modestly affects the NPY system in the retina and these effects are not prevented by sitagliptin treatment. These observations suggest that DPP-IV enzyme is not underlying the NPY changes detected in the retina induced by type 1 DM.
Collapse
Affiliation(s)
- Elisa J Campos
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - João Martins
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Dan Brudzewsky
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Sandra Correia
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Ana R Santiago
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| | - David Pd Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - António F Ambrósio
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| |
Collapse
|
11
|
Seldeen KL, Halley PG, Volmar CH, Rodríguez MA, Hernandez M, Pang M, Carlsson SK, Suva LJ, Wahlestedt C, Troen BR, Brothers SP. Neuropeptide Y Y2 antagonist treated ovariectomized mice exhibit greater bone mineral density. Neuropeptides 2018; 67:45-55. [PMID: 29129406 PMCID: PMC5805636 DOI: 10.1016/j.npep.2017.11.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 12/15/2022]
Abstract
Osteoporosis, a disease characterized by progressive bone loss and increased risk of fracture, often results from menopausal loss of estrogen in women. Neuropeptide Y has been shown to negatively regulate bone formation, with amygdala specific deletion of the Y2 receptor resulting in increased bone mass in mice. In this study, ovariectomized (OVX) mice were injected once daily with JNJ-31020028, a brain penetrant Y2 receptor small molecule antagonist to determine the effects on bone formation. Antagonist treated mice had reduced weight and showed increased whole-body bone mineral density compared to vehicle-injected mice. Micro computerized tomography (micro-CT) demonstrated increased vertebral trabecular bone volume, connectivity density and trabecular thickness. Femoral micro-CT analysis revealed increased bone volume within trabecular regions and greater trabecular number, without significant difference in other parameters or within cortical regions. A decrease was seen in serum P1NP, a measure used to confirm positive treatment outcomes in bisphosphonate treated patients. C-terminal telopeptide 1 (CTX-1), a blood biomarker of bone resorption, was decreased in treated animals. The higher bone mineral density observed following Y2 antagonist treatment, as determined by whole-body DEXA scanning, is indicative of either enhanced mineralization or reduced bone loss. Additionally, our findings that ex vivo treatment of bone marrow cells with the Y2 antagonist did not affect osteoblast and osteoclast formation suggests the inhibitor is not affecting these cells directly, and suggests a central role for compound action in this system. Our results support the involvement of Y2R signalling in bone metabolism and give credence to the hypothesis that selective pharmacological manipulation of Y2R may provide anabolic benefits for treating osteoporosis.
Collapse
Affiliation(s)
- K L Seldeen
- Division of Geriatrics and Palliative Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA
| | - P G Halley
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - C H Volmar
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - M A Rodríguez
- Bruce W. Carter VA Geriatric Research Education and Clinical Center (GRECC), Miami, FL, USA; University of Miami Miller School of Medicine, Miami, FL, USA
| | - M Hernandez
- Bruce W. Carter VA Geriatric Research Education and Clinical Center (GRECC), Miami, FL, USA; University of Miami Miller School of Medicine, Miami, FL, USA
| | - M Pang
- Division of Geriatrics and Palliative Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA
| | - S K Carlsson
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - L J Suva
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, University of Arkansas Medical School, Little Rock, AR, USA
| | - C Wahlestedt
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - B R Troen
- Division of Geriatrics and Palliative Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Research Service, Veterans Affairs Western New York Healthcare System, Buffalo, NY, USA
| | - S P Brothers
- Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
12
|
Navidhamidi M, Ghasemi M, Mehranfard N. Epilepsy-associated alterations in hippocampal excitability. Rev Neurosci 2018; 28:307-334. [PMID: 28099137 DOI: 10.1515/revneuro-2016-0059] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 11/03/2016] [Indexed: 11/15/2022]
Abstract
The hippocampus exhibits a wide range of epilepsy-related abnormalities and is situated in the mesial temporal lobe, where limbic seizures begin. These abnormalities could affect membrane excitability and lead to overstimulation of neurons. Multiple overlapping processes refer to neural homeostatic responses develop in neurons that work together to restore neuronal firing rates to control levels. Nevertheless, homeostatic mechanisms are unable to restore normal neuronal excitability, and the epileptic hippocampus becomes hyperexcitable or hypoexcitable. Studies show that there is hyperexcitability even before starting recurrent spontaneous seizures, suggesting although hippocampal hyperexcitability may contribute to epileptogenesis, it alone is insufficient to produce epileptic seizures. This supports the concept that the hippocampus is not the only substrate for limbic seizure onset, and a broader hyperexcitable limbic structure may contribute to temporal lobe epilepsy (TLE) seizures. Nevertheless, seizures also occur in conditions where the hippocampus shows a hypoexcitable phenotype. Since TLE seizures most often originate in the hippocampus, it could therefore be assumed that both hippocampal hypoexcitability and hyperexcitability are undesirable states that make the epileptic hippocampal network less stable and may, under certain conditions, trigger seizures.
Collapse
|
13
|
Yang XL, Chen B, Zhang XQ, Chen X, Yang MH, Zhang W, Chen HR, Zang ZL, Li W, Yang H, Liu SY. Upregulations of CRH and CRHR1 in the Epileptogenic Tissues of Patients with Intractable Infantile Spasms. CNS Neurosci Ther 2016; 23:57-68. [PMID: 27534449 DOI: 10.1111/cns.12598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Revised: 07/12/2016] [Accepted: 07/20/2016] [Indexed: 01/28/2023] Open
Abstract
AIM Infantile spasms (IS) are an age-specific epileptic syndrome with specific clinical symptom and electroencephalogram (EEG) features, lacking treatment options, and a poor prognosis. Excessive endogenous corticotropin-releasing hormone (CRH) in infant brain might result in IS. However, the data from human IS are limited. In our study, we investigated the expressions of CRH and its receptor type 1 (CRHR1) in surgical tissues from patients with IS and autopsy controls. METHODS Specimens surgically removed from 17 patients with IS, and six autopsy controls were included in the study. Real-time PCR, Western blotting, and immunostaining were used to detect the expressions of mRNA, protein expression, and distribution. The correlation between variates was analyzed by Spearman rank correlation. RESULTS The expressions of CRH and CRHR1 were significantly upregulated in the epileptogenic tissues of IS patients compared with the control group. CRH was distributed mainly in neurons, while CRHR1 was distributed in neurons, astrocytes, and microglia. The expression levels of CRH and CRHR1 were positively correlated with the frequency of epileptic spasms. Moreover, the expression of protein kinase C (PKC), which was an important downstream factor of CRHR1, was significantly upregulated in the epileptogenic tissues of patients with IS and was positively correlated with the CRHR1 expression levels and the frequency of epileptic spasms. CONCLUSION These results suggest that the CRH signal transduction pathway might participate in the epileptogenesis of IS, supporting the hypothesis that CRH is related to the pathogenesis of IS.
Collapse
Affiliation(s)
- Xiao-Lin Yang
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Bing Chen
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Xiao-Qing Zhang
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Xin Chen
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Mei-Hua Yang
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Wei Zhang
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Huan-Ran Chen
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Zhen-Le Zang
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Wei Li
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Hui Yang
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| | - Shi-Yong Liu
- Department of Neurosurgery, Second affiliated Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
14
|
Qi Y, Fu M, Herzog H. Y2 receptor signalling in NPY neurons controls bone formation and fasting induced feeding but not spontaneous feeding. Neuropeptides 2016; 55:91-7. [PMID: 26444586 DOI: 10.1016/j.npep.2015.09.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/25/2015] [Accepted: 09/25/2015] [Indexed: 12/24/2022]
Abstract
Y2 receptors have been implicated in the development of obesity and are a potential target for obesity treatment due to their known role of inhibiting neuropeptide Y (NPY) induced feeding responses. However, the precise neuronal population on which Y2 receptors act to fulfil this role is less clear. Here we utilise a novel inducible, postnatal onset NPY neurons specific deletion model to investigate the functional consequences of loss of Y2 signalling in this population of neurons on feeding and energy homeostasis regulation. While the consequences of lack of Y2 signalling in NPY neurons are confirmed in terms of the uncoupling of suppression/increasing of NPY and pro-opiomelanocortin (POMC) mRNA expression in the arcuate nuclei (Arc), respectively, this lack of Y2 signalling surprisingly does not have any significant effect on spontaneous food intake. Fasting induced food intake, however, is strongly increased but only in the first 1h after re-feeding. Consequently no significant changes in body weight are being observed although body weight gain is increased in male mice after postnatal onset Y2 deletion. Importantly, another known function of central Y2 receptor signalling, the suppression of bone formation is conserved in this conditional model with whole body bone mineral content being decreased. Taken together this model confirms the critical role of Y2 signalling to control NPY and associated POMC expression in the Arc, but also highlights the possibility that others, non-NPY neuronal Y2 receptors, are also involved in controlling feeding and energy homeostasis regulation.
Collapse
Affiliation(s)
- Yue Qi
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia.
| | - Melissa Fu
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| |
Collapse
|
15
|
Ledri LN, Melin E, Christiansen SH, Gøtzsche CR, Cifra A, Woldbye DPD, Kokaia M. Translational approach for gene therapy in epilepsy: Model system and unilateral overexpression of neuropeptide Y and Y2 receptors. Neurobiol Dis 2015; 86:52-61. [PMID: 26607785 DOI: 10.1016/j.nbd.2015.11.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 11/12/2015] [Accepted: 11/18/2015] [Indexed: 11/27/2022] Open
Abstract
Although novel treatment strategies based on the gene therapy approach for epilepsy has been encouraging, there is still a gap in demonstrating a proof-of-concept in a clinically relevant animal model and study design. In the present study, a conceptually novel framework reflecting a plausible clinical trial for gene therapy of temporal lobe epilepsy was explored: We investigated (i) whether the post intrahippocampal kainate-induced status epilepticus (SE) model of chronic epilepsy in rats could be clinically relevant; and (ii) whether a translationally designed neuropeptide Y (NPY)/Y2 receptor-based gene therapy approach targeting only the seizure-generating focus unilaterally can decrease seizure frequency in this chronic model of epilepsy. Our data suggest that the intrahippocampal kainate model resembles the disease development of human chronic mesial temporal lobe epilepsy (mTLE): (i) spontaneous seizures originate in the sclerotic hippocampus; (ii) only a part of the animals develops chronic epilepsy; (iii) animals show largely variable seizure frequency that (iv) tends to progressively increase over time. Despite significant hippocampal degeneration caused by the kainate injection, the use of MRI allowed targeting the recombinant adeno-associated viral (rAAV) vectors encoding NPY and Y2 receptor genes to the remaining dorsal and ventral hippocampal areas ipsilateral to the kainate injection. Continuous video-EEG monitoring demonstrated not only prevention of the progressive increase in seizure frequency in rAAV-NPY/Y2 treated animals as compared to the controls, but even 45% decrease of seizure frequency in 80% of the epileptic animals. This translationally designed study in a clinically relevant model of epilepsy suggests that simultaneous overexpression of NPY and Y2 receptors unilaterally in the seizure focus is a relevant and promising approach that can be further validated in more extensive preclinical studies to develop a future treatment strategy for severe, often pharmacoresistant focal epilepsy cases that cannot be offered alternative therapeutic options.
Collapse
Affiliation(s)
- Litsa Nikitidou Ledri
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - Esbjörn Melin
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - Søren H Christiansen
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Nørregade 10, 1017 Copenhagen, Denmark
| | - Casper R Gøtzsche
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Nørregade 10, 1017 Copenhagen, Denmark
| | - Alessandra Cifra
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden
| | - David P D Woldbye
- Laboratory of Neural Plasticity, Department of Neuroscience and Pharmacology, University of Copenhagen, Nørregade 10, 1017 Copenhagen, Denmark
| | - Mérab Kokaia
- Experimental Epilepsy Group, Epilepsy Centre, Lund University Hospital, Sölvegatan 17, 221 84 Lund, Sweden.
| |
Collapse
|
16
|
Differential Effect of Neuropeptides on Excitatory Synaptic Transmission in Human Epileptic Hippocampus. J Neurosci 2015; 35:9622-31. [PMID: 26134645 DOI: 10.1523/jneurosci.3973-14.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Development of novel disease-modifying treatment strategies for neurological disorders, which at present have no cure, represents a major challenge for today's neurology. Translation of findings from animal models to humans represents an unresolved gap in most of the preclinical studies. Gene therapy is an evolving innovative approach that may prove useful for clinical applications. In animal models of temporal lobe epilepsy (TLE), gene therapy treatments based on viral vectors encoding NPY or galanin have been shown to effectively suppress seizures. However, how this translates to human TLE remains unknown. A unique possibility to validate these animal studies is provided by a surgical therapeutic approach, whereby resected epileptic tissue from temporal lobes of pharmacoresistant patients are available for neurophysiological studies in vitro. To test whether NPY and galanin have antiepileptic actions in human epileptic tissue as well, we applied these neuropeptides directly to human hippocampal slices in vitro. NPY strongly decreased stimulation-induced EPSPs in dentate gyrus and CA1 (up to 30 and 55%, respectively) via Y2 receptors, while galanin had no significant effect. Receptor autoradiographic binding revealed the presence of both NPY and galanin receptors, while functional receptor binding was only detected for NPY, suggesting that galanin receptor signaling may be impaired. These results underline the importance of validating findings from animal studies in human brain tissue, and advocate for NPY as a more appropriate candidate than galanin for future gene therapy trials in pharmacoresistant TLE patients.
Collapse
|
17
|
Elbrønd-Bek H, Gøtzsche CR, Skinbjerg M, Christensen DZ, Plenge P, Woldbye DPD. Visualization of Functional Neuropeptide Y Receptors in the Mouse Hippocampus and Neocortex Using [35S]GTPγS Binding. Int J Pept Res Ther 2015. [DOI: 10.1007/s10989-015-9455-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
18
|
Shin EJ, Nam Y, Tu THT, Lim YK, Wie MB, Kim DJ, Jeong JH, Kim HC. Protein kinase Cδ mediates trimethyltin-induced neurotoxicity in mice in vivo via inhibition of glutathione defense mechanism. Arch Toxicol 2015; 90:937-53. [PMID: 25895139 DOI: 10.1007/s00204-015-1516-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/31/2015] [Indexed: 12/11/2022]
Abstract
We investigated whether protein kinase C (PKC) is involved in trimethyltin (TMT)-induced neurotoxicity. TMT treatment (2.8 mg/kg, i.p.) significantly increased PKCδ expression out of PKC isozymes (i.e., α, βI, βII, δ, and ς) in the hippocampus of wild-type (WT) mice. Consistently, treatment with TMT resulted in significant increases in cleaved PKCδ expression. Genetic or pharmacological inhibition (PKCδ knockout or rottlerin) was less susceptible to TMT-induced seizures than WT mice. TMT treatment increased glutathione oxidation, lipid peroxidation, protein oxidation, and levels of reactive oxygen species. These effects were more pronounced in the WT mice than in PKCδ knockout mice. In addition, the ability of TMT to induce nuclear translocation of Nrf2, Nrf2 DNA-binding activity, and upregulation of γ-glutamylcysteine ligase was significantly increased in the PKCδ knockout mice and rottlerin (10 or 20 mg/kg, p.o. × 6)-treated WT mice. Furthermore, neuronal degeneration (as shown by nuclear chromatin clumping and TUNEL staining) in WT mice was most pronounced 2 days after TMT. At the same time, TMT-induced inhibition of phosphoinositol 3-kinase (PI3K)/Akt signaling was evident, thereby decreasing phospho-Bad, expression of Bcl-xL and Bcl-2, and the interaction between phospho-Bad and 14-3-3 protein, and increasing Bax expression and caspase-3 cleavage were observed. Rottlerin or PKCδ knockout significantly protected these changes in anti- and pro-apoptotic factors. Importantly, treatment of the PI3K inhibitor LY294002 (0.8 or 1.6 µg, i.c.v.) 4 h before TMT counteracted protective effects (i.e., Nrf-2-dependent glutathione induction and pro-survival phenomenon) of rottlerin. Therefore, our results suggest that down-regulation of PKCδ and up-regulations of Nrf2-dependent glutathione defense mechanism and PI3K/Akt signaling are critical for attenuating TMT neurotoxicity.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 200-701, Republic of Korea
| | - Yunsung Nam
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 200-701, Republic of Korea
| | - Thu-Hien Thi Tu
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 200-701, Republic of Korea
| | - Yong Kwang Lim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 200-701, Republic of Korea
| | - Myung-Bok Wie
- School of Veterinary Medicine, Kangwon National University, Chunchon, 200-701, Republic of Korea
| | - Dae-Joong Kim
- Department of Anatomy and Cell Biology, Medical School, Kangwon National University, Chunchon, 200-701, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul, 156-756, Republic of Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, 200-701, Republic of Korea.
| |
Collapse
|
19
|
Elbrønd-Bek H, Olling JD, Gøtzsche CR, Waterfield A, Wörtwein G, Woldbye DPD. Neuropeptide Y-stimulated [(35) S]GTPγs functional binding is reduced in the hippocampus after kainate-induced seizures in mice. Synapse 2014; 68:427-36. [PMID: 24985894 DOI: 10.1002/syn.21762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 06/16/2014] [Accepted: 06/26/2014] [Indexed: 11/09/2022]
Abstract
Kainate-induced seizures constitute a model of temporal lobe epilepsy where prominent changes are observed in the hippocampal neuropeptide Y (NPY) system. However, little is known about the functional state and signal transduction of the NPY receptor population resulting from kainate exposure. Thus, in this study, we explored functional NPY receptor activity in the mouse hippocampus and neocortex after kainate-induced seizures using NPY-stimulated [(35) S]GTPγS binding. Moreover, we also studied levels of [(125) I]-peptide YY (PYY) binding and NPY, Y1, Y2, and Y5 receptor mRNA in these kainate-treated mice. Functional NPY binding was unchanged up to 12 h post-kainate, but decreased significantly in all hippocampal regions after 24 h and 1 week. Similarly, a decrease in [(125) I]-PYY binding was found in the dentate gyrus (DG) 1 week post-kainate. However, at 2 h, 6 h, and 12 h, [(125) I]-PYY binding was increased in all regions, and in the CA1 also at 24 h post-kainate. NPY mRNA levels were prominently increased in hippocampal regions, reaching maximum at 12 and 24 h. Y1 and Y5 mRNA levels were lowered in the DG at 24 and 2 h, respectively, while Y2 mRNA levels were elevated at 24 h in the DG and CA3. This study confirms rat kainate studies by showing pronounced adaptive changes in the mouse hippocampus both with regard to NPY synthesis and NPY receptor synthesis and binding, which may contribute to regulating neuronal seizure susceptibility after kainate. However, the potential seizure-suppressant effects of increased NPY gene expression at late time points post-kainate could be attenuated by the novel finding of reduced NPY-receptor G-protein activation.
Collapse
Affiliation(s)
- Heidi Elbrønd-Bek
- Department of Neuroscience and Pharmacology, Laboratory of Neural Plasticity, University of Copenhagen, Copenhagen, Denmark; Department of Neuroscience and Pharmacology, Laboratory of Neuropsychiatry, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
20
|
Xu X, Guo F, He Q, Cai X, Min D, Wang Q, Wang S, Tian L, Cai J, Zhao Y. Altered expression of neuropeptide Y, Y1 and Y2 receptors, but not Y5 receptor, within hippocampus and temporal lobe cortex of tremor rats. Neuropeptides 2014; 48:97-105. [PMID: 24444822 DOI: 10.1016/j.npep.2013.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 12/18/2013] [Accepted: 12/22/2013] [Indexed: 01/24/2023]
Abstract
As an endogenous inhibitor of glutamate-mediated synaptic transmission in mammalian central nervous system, neuropeptide Y (NPY) plays a crucial role in regulating homeostasis of neuron excitability. Loss of balance between excitatory and inhibitory neurotransmission is thought to be a chief mechanism of epileptogenesis. The abnormal expression of NPY and its receptors observed following seizures have been demonstrated to be related to the production of epilepsy. The tremor rat (TRM) is a hereditary epileptic animal model. So far, there is no report concerning whether NPY and its receptors may be involved in TRM pathogenesis. In this study, we focused on the expression of NPY and its three receptor subtypes: Y1R, Y2R and Y5R in the TRM brain. We first found the expression of NPY in TRM hippocampus and temporal lobe cortex was increased compared with control (Wistar) rats. The mRNA and protein expression of Y1R was down-regulated in hippocampus but up-regulated in temporal lobe cortex, whereas Y2R expression was significantly increased in both areas. There was no significant change of Y5R expression in either area. The immunohistochemistry data showed that Y1R, Y2R, Y5R were present throughout CA1, CA3, dentate gyrus (DG) and the entorhinal cortex which is included in the temporal lobe cortex of TRM. In conclusion, our results showed the altered expression of NPY, Y1R and Y2R but not Y5R in hippocampus and temporal lobe cortex of TRM brain. This abnormal expression may be associated with the generation of epileptiform activity and provide a candidate target for treatment of genetic epilepsy.
Collapse
Affiliation(s)
- Xiaoxue Xu
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China; Department of Neurology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Qun He
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Xinze Cai
- Central Lab, The First Hospital of China Medical University, Shenyang 110001, China
| | - Dongyu Min
- Experiment Center of Traditional Chinese Medicine, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China
| | - Qianhui Wang
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Shaocheng Wang
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Liu Tian
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Jiqun Cai
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Yujie Zhao
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China.
| |
Collapse
|
21
|
Agasse F, Xapelli S, Coronas V, Christiansen SH, Rosa AI, Sardá-Arroyo L, Santos T, Ferreira R, Schitine C, Harnois T, Bourmeyster N, Bragança J, Bernardino L, Malva JO, Woldbye DPD. Galanin promotes neuronal differentiation in murine subventricular zone cell cultures. Stem Cells Dev 2013; 22:1693-1708. [PMID: 23327619 DOI: 10.1089/scd.2012.0161] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Neural stem cells of the subventricular zone (SVZ) represent a potentially important source of surrogate cells for the treatment of brain damage. Proper use of these cells for neuronal replacement depends on the ability to drive neuronal differentiation. Several neuromodulators stimulate neurogenesis. Here we examined the effects of the neuropeptide galanin, on neuronal differentiation in murine SVZ cultures. SVZ neurospheres obtained from early postnatal mice were treated with 10 nM to 2 μM galanin. Galanin promoted neuronal differentiation, increasing numbers of NeuN-, vesicular GABA transporter- and tyrosine hydroxylase-expressing neurons. In contrast, galanin neither affected cell proliferation assessed by BrdU incorporation nor cell death evaluated by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Neuronal differentiation was further confirmed at the functional level by measuring [Ca(2+)]i variations in single SVZ cells after KCl and histamine stimulations to distinguish neurons from immature cells. Galanin treatment increased the numbers of neuronal-like responding cells compared to immature cells. Using selective agonists (M617, AR-M1896) and antagonists (galantide, M871) for galanin receptors 1 and 2, we showed that both galanin receptors mediated neuronal differentiation. Early proneuronal effects of galanin included positive regulation of the transcription factor neurogenin-1 (Ngn1). In addition, galanin promoted axonogenesis and dendritogenesis, increasing both the length of phosphorylated stress-activated protein kinase- and Tau-positive axons and the numbers of microtubule associated protein-2 (MAP-2)-labelled dendrites. Moreover, galanin inhibited SVZ cell migration in the transwell assay. Our results show a proneurogenic effect of galanin and open new perspectives for future applications in stem cell-based therapies for neuronal replacement.
Collapse
Affiliation(s)
- Fabienne Agasse
- Center for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês de Pombal, Coimbra, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Gonçalves J, Baptista S, Olesen MV, Fontes-Ribeiro C, Malva JO, Woldbye DP, Silva AP. Methamphetamine-induced changes in the mice hippocampal neuropeptide Y system: implications for memory impairment. J Neurochem 2012; 123:1041-53. [PMID: 23061411 DOI: 10.1111/jnc.12052] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 09/20/2012] [Accepted: 10/08/2012] [Indexed: 11/30/2022]
Abstract
Methamphetamine (METH) is a psychostimulant drug that causes irreversible brain damage leading to several neurological and psychiatric abnormalities, including cognitive deficits. Neuropeptide Y (NPY) is abundant in the mammalian central nervous system (CNS) and has several important functions, being involved in learning and memory processing. It has been demonstrated that METH induces significant alteration in mice striatal NPY, Y(1) and Y(2) receptor mRNA levels. However, the impact of this drug on the hippocampal NPY system and its consequences remain unknown. Thus, in this study, we investigated the effect of METH intoxication on mouse hippocampal NPY levels, NPY receptors function, and memory performance. Results show that METH increased NPY, Y(2) and Y(5) receptor mRNA levels, as well as total NPY binding accounted by opposite up- and down-regulation of Y(2) and Y(1) functional binding, respectively. Moreover, METH-induced impairment in memory performance and AKT/mammalian target of rapamycin pathway were both prevented by the Y(2) receptor antagonist, BIIE0246. These findings demonstrate that METH interferes with the hippocampal NPY system, which seems to be associated with memory failure. Overall, we concluded that Y(2) receptors are involved in memory deficits induced by METH intoxication.
Collapse
Affiliation(s)
- Joana Gonçalves
- Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | |
Collapse
|
23
|
Gonçalves J, Ribeiro CF, Malva JO, Silva AP. Protective role of neuropeptide Y Y2receptors in cell death and microglial response following methamphetamine injury. Eur J Neurosci 2012; 36:3173-83. [DOI: 10.1111/j.1460-9568.2012.08232.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
24
|
Baptista S, Bento AR, Gonçalves J, Bernardino L, Summavielle T, Lobo A, Fontes-Ribeiro C, Malva JO, Agasse F, Silva AP. Neuropeptide Y promotes neurogenesis and protection against methamphetamine-induced toxicity in mouse dentate gyrus-derived neurosphere cultures. Neuropharmacology 2012; 62:2413-23. [DOI: 10.1016/j.neuropharm.2012.02.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 02/13/2012] [Accepted: 02/14/2012] [Indexed: 10/28/2022]
|
25
|
Olesen MV, Christiansen SH, Gøtzsche CR, Holst B, Kokaia M, Woldbye DPD. Y5 neuropeptide Y receptor overexpression in mice neither affects anxiety- and depression-like behaviours nor seizures but confers moderate hyperactivity. Neuropeptides 2012; 46:71-9. [PMID: 22342800 DOI: 10.1016/j.npep.2012.01.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Revised: 01/11/2012] [Accepted: 01/27/2012] [Indexed: 02/07/2023]
Abstract
Neuropeptide Y (NPY) has been implicated in anxiolytic- and antidepressant-like behaviour as well as seizure-suppressant effects in rodents. Although these effects appear to be predominantly mediated via other NPY receptors (Y1 and/or Y2), several studies have also indicated a role for Y5 receptors. Gene therapy using recombinant viral vectors to induce overexpression of NPY, Y1 or Y2 receptors in the hippocampus or amygdala has previously been shown to modulate emotional behaviour and seizures in rodents. The present study explored the potential effects of gene therapy with the Y5 receptor, by testing effects of recombinant adeno-associated viral vector (rAAV) encoding Y5 (rAAV-Y5) in anxiety- and depression-like behaviour as well as in kainate-induced seizures in adult mice. The rAAV-Y5 vector injected into the hippocampus and amygdala induced a pronounced and sustained increase in Y5 receptor mRNA expression and functional Y5 receptor binding, but no significant effects were found with regard to anxiety- and depression-like behaviours or seizure susceptibility. Instead, rAAV-mediated Y5 receptor transgene overexpression resulted in moderate hyperactivity in the open field test. These results do not support a potential role for single transgene overexpression of Y5 receptors for modulating anxiety-/depression-like behaviours or seizures in adult mice. Whether the induction of hyperactivity by rAAV-Y5 could be relevant for other conditions remains to be studied.
Collapse
Affiliation(s)
- M V Olesen
- Protein Laboratory & Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
26
|
Gøtzsche CR, Nikitidou L, Sørensen AT, Olesen MV, Sørensen G, Christiansen SH, Ängehagen M, Woldbye DP, Kokaia M. Combined gene overexpression of neuropeptide Y and its receptor Y5 in the hippocampus suppresses seizures. Neurobiol Dis 2012; 45:288-96. [DOI: 10.1016/j.nbd.2011.08.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 07/23/2011] [Accepted: 08/15/2011] [Indexed: 01/13/2023] Open
|
27
|
Ferreira R, Santos T, Cortes L, Cochaud S, Agasse F, Silva AP, Xapelli S, Malva JO. Neuropeptide Y inhibits interleukin-1 beta-induced microglia motility. J Neurochem 2011; 120:93-105. [DOI: 10.1111/j.1471-4159.2011.07541.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
28
|
Olesen MV, Christiansen SH, Gøtzsche CR, Nikitidou L, Kokaia M, Woldbye DPD. Neuropeptide Y Y1 receptor hippocampal overexpression via viral vectors is associated with modest anxiolytic-like and proconvulsant effects in mice. J Neurosci Res 2011; 90:498-507. [PMID: 21971867 DOI: 10.1002/jnr.22770] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 07/15/2011] [Accepted: 07/24/2011] [Indexed: 02/06/2023]
Abstract
Neuropeptide Y (NPY) exerts anxiolytic- and antidepressant-like effects in rodents that appear to be mediated via Y1 receptors. Gene therapy using recombinant viral vectors to induce overexpression of NPY in the hippocampus or amygdala has previously been shown to confer anxiolytic-like effect in rodents. The present study explored an alternative and more specific approach: overexpression of Y1 receptors. Using a recombinant adeno-associated viral vector (rAAV) encoding the Y1 gene (rAAV-Y1), we, for the first time, induced overexpression of functional transgene Y1 receptors in the hippocampus of adult mice and tested the animals in anxiety- and depression-like behavior. Hippocampal Y1 receptors have been suggested to mediate seizure-promoting effect, so the effects of rAAV-induced Y1 receptor overexpression were also tested in kainate-induced seizures. Y1 receptor transgene overexpression was found to be associated with modest anxiolytic-like effect in the open field and elevated plus maze tests, but no effect was seen on depression-like behavior using the tail suspension and forced swim tests. However, the rAAV-Y1 vector modestly aggravated kainate-induced seizures. These data indicate that rAAV-induced overexpression of Y1 receptors in the hippocampus could confer anxiolytic-like effect accompanied by a moderate proconvulsant adverse effect. Further studies are clearly needed to determine whether Y1 gene therapy might have a future role in the treatment of anxiety disorders.
Collapse
Affiliation(s)
- M V Olesen
- Laboratory of Neuropsychiatry, Psychiatric Centre Copenhagen and Protein Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
29
|
Kim YS, Choi MY, Kim YH, Jeon BT, Lee DH, Roh GS, Kang SS, Kim HJ, Cho GJ, Choi WS. Protein kinase Cdelta is associated with 14-3-3 phosphorylation in seizure-induced neuronal death. Epilepsy Res 2010; 92:30-40. [PMID: 20813501 DOI: 10.1016/j.eplepsyres.2010.08.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 07/22/2010] [Accepted: 08/08/2010] [Indexed: 11/29/2022]
Abstract
Prolonged seizures cause significant damage to the brain, and cellular damage due to status epilepticus may be related to the pathogenesis of epilepsy. Protein kinase Cdelta (PKCδ) mediates multiple cell death signalings, and 14-3-3 proteins regulate survival pathways in brain, sequestering certain pro-apoptotic proteins. Presently, we examined the association between PKCδ and 14-3-3 with seizure-induced neuronal death using mouse model. Status epilepticus was induced by systemic kainic acid. Kainate-induced seizures caused an increase in levels of cleaved PKCδ in the hippocampus, along with up-regulation of cleaved caspase-3 and phospho-14-3-3ζ (Ser58), as well as extensive hippocampal cell death as visualized with Fluoro-Jade B and anti-active caspase-3 staining. Furthermore, co-immunoprecipitation or double immunofluorescence analysis revealed that PKCδ interacts with 14-3-3, and interaction between PKCδ and 14-3-3 was significantly enhanced in the hippocampus after seizures, paralleling increased interaction between Bad and Bcl-x(L). Moreover, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL)-positive cells had upregulated phospho-14-3-3ζ (Ser58) in the hippocampus after seizures. These findings suggest that PKCδ and phospho-14-3-3 are associated with apoptotic cell death in the hippocampus after seizures, and targeting PKCδ or phospho-14-3-3 may be potently protective against seizure-induced neuronal injury.
Collapse
Affiliation(s)
- Yoon Sook Kim
- Department of Anatomy and Neurobiology, School of Medicine, Institute of Health Science, Gyeongsang National University, Chilam-dong 92, Jinju, Gyeongnam 660-751, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Woldbye DPD, Angehagen M, Gøtzsche CR, Elbrønd-Bek H, Sørensen AT, Christiansen SH, Olesen MV, Nikitidou L, Hansen TVO, Kanter-Schlifke I, Kokaia M. Adeno-associated viral vector-induced overexpression of neuropeptide Y Y2 receptors in the hippocampus suppresses seizures. ACTA ACUST UNITED AC 2010; 133:2778-88. [PMID: 20688813 DOI: 10.1093/brain/awq219] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Gene therapy using recombinant adeno-associated viral vectors overexpressing neuropeptide Y in the hippocampus exerts seizure-suppressant effects in rodent epilepsy models and is currently considered for clinical application in patients with intractable mesial temporal lobe epilepsy. Seizure suppression by neuropeptide Y in the hippocampus is predominantly mediated by Y2 receptors, which, together with neuropeptide Y, are upregulated after seizures as a compensatory mechanism. To explore whether such upregulation could prevent seizures, we overexpressed Y2 receptors in the hippocampus using recombinant adeno-associated viral vectors. In two temporal lobe epilepsy models, electrical kindling and kainate-induced seizures, vector-based transduction of Y2 receptor complementary DNA in the hippocampus of adult rats exerted seizure-suppressant effects. Simultaneous overexpression of Y2 and neuropeptide Y had a more pronounced seizure-suppressant effect. These results demonstrate that overexpression of Y2 receptors (alone or in combination with neuropeptide Y) could be an alternative strategy for epilepsy treatment.
Collapse
Affiliation(s)
- David P D Woldbye
- Experimental Epilepsy Group, Wallenberg Neuroscience Center, Lund University Hospital, Lund, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kim SH, Kim MK, Yu HS, Kim HS, Park IS, Park HG, Kang UG, Kim YS. Electroconvulsive seizure increases phosphorylation of PKC substrates, including GAP-43, MARCKS, and neurogranin, in rat brain. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34:115-21. [PMID: 19837121 DOI: 10.1016/j.pnpbp.2009.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Revised: 10/08/2009] [Accepted: 10/08/2009] [Indexed: 01/06/2023]
Abstract
Protein kinase C (PKC) has been suggested as a molecular target related to the pathogenetic and therapeutic mechanisms of mood disorders in which electroconvulsive seizure (ECS) is effective. However, the reports concerning the effects of ECS on PKC are anecdotal and need further clarification. In this study, we examined the effects of ECS treatment on the phosphorylation of PKC substrates, including GAP-43, MARCKS, and neurogranin. Immunoblot using anti-p-PKC substrate antibodies revealed that a single ECS treatment induced temporal changes in the phosphorylation level of PKC substrates in rat brain, reflecting the effects on PKC activity. Phosphorylation of GAP-43 and MARCKS, representative PKC substrates related to synaptic remodeling, increased from 5 to 30 min, after a transient decrease at 0 min immediately after ECS, and returned to basal levels at 60 min in rat frontal cortex, hippocampus, and cerebellum. Phosphorylation of neurogranin, another PKC substrate, showed a similar pattern of temporal changes in the frontal cortex and hippocampus. Immunohistochemical analysis revealed that p-GAP-43 and p-MARCKS were densely stained throughout the neuronal cells of the prefrontal cortex and hippocampus, and the Purkinje cells of cerebellum, after ECS treatment. Brief and transient activation of PKC may be translated into long-term biochemical changes, resulting in synaptic plasticity. Taken together, the acute effects of ECS on PKC activity, which could be an underpinning of long-term biochemical changes induced by ECS, may contribute to understand the molecular mechanism of ECS.
Collapse
Affiliation(s)
- Se Hyun Kim
- Department of Psychiatry and Behavioral Science and Institute of Human Behavioral Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Olling JD, Ulrichsen J, Correll M, Woldbye DPD. Gene expression in the neuropeptide Y system during ethanol withdrawal kindling in rats. Alcohol Clin Exp Res 2009; 34:462-70. [PMID: 20028355 DOI: 10.1111/j.1530-0277.2009.01110.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
BACKGROUND Multiple episodes of ethanol intoxication and withdrawal result in progressive, irreversible intensification of the withdrawal reaction, a process termed "ethanol withdrawal kindling." Previous studies show that a single episode of chronic ethanol intoxication and withdrawal causes prominent changes in neuropeptide Y (NPY) and its receptors that have been implicated in regulating withdrawal hyperexcitability. This study for the first time examined the NPY system during ethanol withdrawal kindling. METHODS Ethanol withdrawal kindling was studied in rats receiving 16 episodes of 2 days of chronic ethanol intoxication by intragastric intubations followed by 5 days withdrawal. The study included 6 groups: 4 multiple withdrawal episode (MW) groups [peak withdrawal plus (MW+)/minus (MW-) seizures, 3-day (MW3d), and 1-month (MW1mth) withdrawal], a single withdrawal episode group (SW), and an isocalorically fed control group. Gene expression of NPY and its receptors Y1, Y2, and Y5 was studied in the hippocampal dentate gyrus (DG) and CA3/CA1, as well as piriform cortex (PirCx), and neocortex (NeoCx). RESULTS MW+/- as well as SW groups showed decreased NPY gene expression in all hippocampal areas compared with controls, but, in the DG and CA3, decreases were significantly smaller in the MW- group compared with the SW group. In the MW+/- and SW groups, Y1, Y2, and Y5 mRNA levels were decreased in most brain areas compared with controls; however, decreases in Y1 and Y5 mRNA were augmented in the MW+/- groups compared with the SW group. The MW+ group differed from the MW- group in the PirCx, where Y2 gene expression was significantly higher. CONCLUSION Multiple withdrawal episodes reversibly decreased NPY and NPY receptor mRNA levels at peak withdrawal, with smaller decreases in NPY mRNA levels and augmented decreases in Y1/Y5 mRNA levels compared with a SW episode. Multiple withdrawal-induced seizures increased the Y2 mRNA levels in PirCx. These complex changes in NPY system gene expression could play a role in the ethanol withdrawal kindling process.
Collapse
Affiliation(s)
- Janne D Olling
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen & University Hospital Rigshospitalet, Copenhagen, Denmark
| | | | | | | |
Collapse
|
33
|
Voss LJ, Sleigh JW, Barnard JPM, Kirsch HE. The Howling Cortex: Seizures and General Anesthetic Drugs. Anesth Analg 2008; 107:1689-703. [PMID: 18931234 DOI: 10.1213/ane.0b013e3181852595] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
34
|
Fuortes MG, Faria LC, Merlin LR. Impact of protein kinase C activation on epileptiform activity in the hippocampal slice. Epilepsy Res 2008; 82:38-45. [PMID: 18715754 PMCID: PMC2633120 DOI: 10.1016/j.eplepsyres.2008.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Revised: 06/03/2008] [Accepted: 07/02/2008] [Indexed: 11/24/2022]
Abstract
There is evidence suggesting that protein kinase C (PKC) activation can prevent the enhanced network excitability associated with status epilepticus and group I metabotropic glutamate receptor (mGluR)-induced epileptogenesis. However, we observed no suppression of mGluR-induced burst prolongation in the guinea pig hippocampal slice when applied in the presence of the PKC activator phorbol-12,13-dibutyrate (PDBu). Furthermore, PDBu alone converted picrotoxin-induced interictal bursts into ictal-length discharges ranging from 2 to 6s in length. This effect could not be elicited by the inactive analog 4-alpha-PDBu and was suppressed with the PKC inhibitor chelerythrine, indicating PKC dependence. PKC activation can enhance neurotransmitter release, and both glutamate and acetylcholine are capable of eliciting similar prolonged synchronized discharges. However, neither mGluR1 nor NMDA receptor antagonist suppressed PDBu-driven burst prolongation, suggesting that increased glutamate release alone is unlikely to account for the PKC-induced expression of ictaform discharges. Similarly, atropine, a broad-spectrum muscarinic receptor antagonist, had no effect on PKC-induced burst prolongation. By contrast, AMPA/kainate receptor antagonist abolished PKC-induced burst prolongation, and mGluR5 antagonist significantly blunted the maximum burst length induced by PKC. These data suggest that PKC-induced prolongation of epileptiform bursts is dependent on changes specific to mGluR5 and AMPA/kainate receptors and not mediated simply by a generalized increase in transmitter release.
Collapse
Affiliation(s)
- Michaelangelo G. Fuortes
- Neural and Behavioral Sciences Program, School of Graduate Studies, State University of New York Downstate Medical Center, Brooklyn, NY 11203
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203
| | - Leonardo C. Faria
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY 11203
| | - Lisa R. Merlin
- Neural and Behavioral Sciences Program, School of Graduate Studies, State University of New York Downstate Medical Center, Brooklyn, NY 11203
- The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York Downstate Medical Center, Brooklyn, NY 11203
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY 11203
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, NY 11203
| |
Collapse
|
35
|
Merlin LR. Impact of protein kinase C activation on status epilepticus and epileptogenesis: oh, what a tangled web. Epilepsy Curr 2008; 8:101-3. [PMID: 18596877 DOI: 10.1111/j.1535-7511.2008.00256.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Deficits in Phosphorylation of GABAA Receptors by Intimately Associated Protein Kinase C Activity Underlie Compromised Synaptic Inhibition during Status Epilepticus. Terunuma M, Xu J, Vithlani M, Sieghart W, Kittler J, Pangalos M, Haydon PG, Coulter DA, Moss SJ. J Neurosci 2008;28(2):376–384. Status epilepticus (SE) is a progressive and often lethal human disorder characterized by continuous or rapidly repeating seizures. Of major significance in the pathology of SE are deficits in the functional expression of GABAA receptors (GABAARs), the major sites of fast synaptic inhibition in the brain. We demonstrate that SE selectively decreases the phosphorylation of GABAARs on serine residues 408/9 (S408/9) in the β3 subunit by intimately associated protein kinase C isoforms. Dephosphorylation of S408/9 unmasks a basic patch-binding motif for the clathrin adaptor AP2, enhancing the endocytosis of selected GABAAR subtypes from the plasma membrane during SE. In agreement with this, enhancing S408/9 phosphorylation or selectively blocking the binding of the β3 subunit to AP2 increased GABAAR cell surface expression levels and restored the efficacy of synaptic inhibition in SE. Thus, enhancing phosphorylation of GABAARs or selectively blocking their interaction with AP2 may provide novel therapeutic strategies to ameliorate SE.
Collapse
|
36
|
|
37
|
Abstract
We briefly survey the current knowledge and concepts regarding structure and function of the neuropeptide Y Y2 receptor and its agonists, especially as related to pharmacology of the receptor and its roles in pathological processes. Specific structural features are considered that could be responsible for the known compartmentalization and participation of the receptor in cell and tissue organization. This is further discussed in relation to changes of levels of the Y2 receptor in pathological conditions (especially in epilepsy and drug abuse), to endocytosis and recycling, and to participation in wound healing, retinopathy and angiogenesis. Properties of the receptor and of Y2 agonists are considered and reviewed in connection to the negative regulation of transmitter release, feeding, mood and social behavior. The possible involvement of the Y2 receptor in diabetes, carcinogenesis and bone formation is also reviewed.
Collapse
Affiliation(s)
- S L Parker
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | | |
Collapse
|