1
|
Duncan RS, Riordan SM, Hall CW, Payne AJ, Chapman KD, Koulen P. N-acylethanolamide metabolizing enzymes are upregulated in human neural progenitor-derived neurons exposed to sub-lethal oxidative stress. Front Cell Neurosci 2022; 16:902278. [PMID: 36003139 PMCID: PMC9393304 DOI: 10.3389/fncel.2022.902278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/05/2022] [Indexed: 11/28/2022] Open
Abstract
N-acyl amides (NAAs) are a class of lipids that consist of an acyl group N-linked to an amino acid, neurotransmitter, taurine or ethanolamide group (N-acylethanolamines or NAEs) and include some endocannabinoids (eCB) such as anandamide. These lipids are synthesized in a wide variety of organisms and in multiple cell types, including neurons. NAEs are involved in numerous cellular and physiological processes and their concentrations are elevated in response to ischemia and physical trauma to play a role in neuroprotection. The neuroprotective properties of eCB NAEs make the protein targets of these compounds attractive targets for clinical intervention for a variety of conditions. The most promising of these targets include cannabinoid receptor type 1 (CB1), cannabinoid receptor type 2 (CB2), fatty acid amide hydrolase (FAAH), N-acylethanolamine acid amidase (NAAA), and N-acyl phosphatidylethanolamine phospholipase D (NAPE-PLD). Further characterization of these targets in a more contemporary model system of neurodegeneration and neuroprotection will allow us to fully describe their role and mechanism of action in neuroprotection against oxidative stress leading to better utilization in the clinical setting. Human stem cell-derived or human neural progenitor cell-derived cells, such as ReN cells, have become more utilized for the study of human neuronal development and neurodegenerative diseases. ReN cells can be easily differentiated thereby circumventing the need for using transformed cell lines and primary neurons as cell model systems. In this study, we determined whether ReN cells, a superior cell model system for studying neurodevelopment, differentiation, and neuroprotection, express proteins involved in canonical eCB NAE signaling and whether oxidative stress can induce their expression. We determined that sublethal oxidative stress upregulates the expression of all eCB proteins tested. In addition, we determined that oxidative stress increases the nuclear localization of FAAH, and to a lesser extent, NAAA and NAPE-PLD. This study is a first step toward determining how oxidative stress affects CB1, CB2, FAAH, NAAA, and NAPE-PLD expression and their potential defense against oxidative stress. As such, our data is important for further determining the role of eCB metabolizing proteins and eCB receptors against oxidative stress.
Collapse
Affiliation(s)
- R. Scott Duncan
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri–Kansas City, Kansas City, MO, United States
| | - Sean M. Riordan
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri–Kansas City, Kansas City, MO, United States
| | - Conner W. Hall
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri–Kansas City, Kansas City, MO, United States
| | - Andrew J. Payne
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri–Kansas City, Kansas City, MO, United States
| | - Kent D. Chapman
- Department of Biological Sciences, Center for Plant Lipid Research, University of North Texas, Denton, TX, United States
| | - Peter Koulen
- Department of Ophthalmology, Vision Research Center, School of Medicine, University of Missouri–Kansas City, Kansas City, MO, United States
- Department of Biological Sciences, Center for Plant Lipid Research, University of North Texas, Denton, TX, United States
- Department of Biomedical Sciences, School of Medicine, University of Missouri–Kansas City, Kansas City, MO, United States
- *Correspondence: Peter Koulen,
| |
Collapse
|
2
|
Ferreira C, Almeida C, Tenreiro S, Quintas A. Neuroprotection or Neurotoxicity of Illicit Drugs on Parkinson's Disease. Life (Basel) 2020; 10:life10060086. [PMID: 32545328 PMCID: PMC7344445 DOI: 10.3390/life10060086] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson's Disease (PD) is currently the most rapid growing neurodegenerative disease and over the past generation, its global burden has more than doubled. The onset of PD can arise due to environmental, sporadic or genetic factors. Nevertheless, most PD cases have an unknown etiology. Chemicals, such as the anthropogenic pollutant 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and amphetamine-type stimulants, have been associated with the onset of PD. Conversely, cannabinoids have been associated with the treatment of the symptoms'. PD and medical cannabis is currently under the spotlight, and research to find its benefits on PD is on-going worldwide. However, the described clinical applications and safety of pharmacotherapy with cannabis products are yet to be fully supported by scientific evidence. Furthermore, the novel psychoactive substances are currently a popular alternative to classical drugs of abuse, representing an unknown health hazard for young adults who may develop PD later in their lifetime. This review addresses the neurotoxic and neuroprotective impact of illicit substance consumption in PD, presenting clinical evidence and molecular and cellular mechanisms of this association. This research area is utterly important for contemporary society since illicit drugs' legalization is under discussion which may have consequences both for the onset of PD and for the treatment of its symptoms.
Collapse
Affiliation(s)
- Carla Ferreira
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
- Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário–Quinta da Granja, Monte de Caparica, P-2825-084 Caparica, Portugal
- Faculty of Medicine of Porto University, Al. Prof. Hernâni Monteiro, P-4200–319 Porto, Portugal
| | - Catarina Almeida
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
| | - Sandra Tenreiro
- CEDOC–Chronic Diseases Research Center, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, P-1150-082 Lisboa, Portugal;
| | - Alexandre Quintas
- Molecular Pathology and Forensic Biochemistry Laboratory, Centro de Investigação Interdisciplinar Egas Moniz, P-2825-084 Caparica, Portugal; (C.F.); (C.A.)
- Laboratório de Ciências Forenses e Psicológicas Egas Moniz, Campus Universitário–Quinta da Granja, Monte de Caparica, P-2825-084 Caparica, Portugal
- Correspondence:
| |
Collapse
|
3
|
Ignatowska J, Mironiuk-Puchalska E, Grześkowiak P, Wińska P, Wielechowska M, Bretner M, Karatsai O, Rędowicz MJ, Koszytkowska-Stawińska M. New insight into nucleo α-amino acids - Synthesis and SAR studies on cytotoxic activity of β-pyrimidine alanines. Bioorg Chem 2020; 100:103864. [PMID: 32446118 DOI: 10.1016/j.bioorg.2020.103864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 04/15/2020] [Accepted: 04/17/2020] [Indexed: 12/14/2022]
Abstract
Three series of the β-pyrimidine alanines, including willardiine - a naturally occurring amino acid, were prepared from the l-serine-derived sulfamidates. Compounds 3b, 4a and 4b demonstrated antiproliferative activity toward the studied cancer cell lines, albeit the effect of these compounds on human brain astrocytoma MOG-G-CCM cells was more significant than on human neuroblastoma SK-N-AS cells. The cytosine analog of willardiine, compound 4b, reduced viability of MOG-G-CCM cells with EC50 = 36 ± 2 μM, more effectively than AMPA antagonist GYKI 52466. Willardiine showed possible capability of affecting invasiveness of glioblastoma U251 MG cells with no effect on their viability and morphology. Compound 3d, the ethyl ester of willardiine, featured activity toward binding domain hHS1S2I of the GluR2 receptor. Docking analysis revealed that the location mode of compound 3d at the S1S2 domain of hGluR2 (PDB ID: 3R7X) might differ from that of willardiine.
Collapse
Affiliation(s)
- Jolanta Ignatowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Ewa Mironiuk-Puchalska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Piotr Grześkowiak
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Patrycja Wińska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Monika Wielechowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Maria Bretner
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Olena Karatsai
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Maria Jolanta Rędowicz
- Laboratory of Molecular Basis of Cell Motility, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland
| | | |
Collapse
|
4
|
Ceprian M, Fulton D. Glial Cell AMPA Receptors in Nervous System Health, Injury and Disease. Int J Mol Sci 2019; 20:E2450. [PMID: 31108947 PMCID: PMC6566241 DOI: 10.3390/ijms20102450] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/11/2019] [Accepted: 04/22/2019] [Indexed: 12/16/2022] Open
Abstract
Glia form a central component of the nervous system whose varied activities sustain an environment that is optimised for healthy development and neuronal function. Alpha-amino-3-hydroxy-5-methyl-4-isoxazole (AMPA)-type glutamate receptors (AMPAR) are a central mediator of glutamatergic excitatory synaptic transmission, yet they are also expressed in a wide range of glial cells where they influence a variety of important cellular functions. AMPAR enable glial cells to sense the activity of neighbouring axons and synapses, and as such many aspects of glial cell development and function are influenced by the activity of neural circuits. However, these AMPAR also render glia sensitive to elevations of the extracellular concentration of glutamate, which are associated with a broad range of pathological conditions. Excessive activation of AMPAR under these conditions may induce excitotoxic injury in glial cells, and trigger pathophysiological responses threatening other neural cells and amplifying ongoing disease processes. The aim of this review is to gather information on AMPAR function from across the broad diversity of glial cells, identify their contribution to pathophysiological processes, and highlight new areas of research whose progress may increase our understanding of nervous system dysfunction and disease.
Collapse
Affiliation(s)
- Maria Ceprian
- Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain.
- Departamento de Bioquímica y Biología Molecular, CIBERNED, IRICYS. Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Daniel Fulton
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
5
|
Sarne Y. Beneficial and deleterious effects of cannabinoids in the brain: the case of ultra-low dose THC. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2019; 45:551-562. [PMID: 30864864 DOI: 10.1080/00952990.2019.1578366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This article reviews the neurocognitive advantages and drawbacks of cannabinoid substances, and discusses the possible physiological mechanisms that underlie their dual activity. The article further reviews the neurocognitive effects of ultra-low doses of ∆9-tetrahydrocannabinol (THC; 3-4 orders of magnitude lower than the conventional doses) in mice, and proposes such low doses of THC as a possible remedy for various brain injuries and for the treatment of age-related cognitive decline.
Collapse
Affiliation(s)
- Yosef Sarne
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
6
|
Cannabinoid signalling in the immature brain: Encephalopathies and neurodevelopmental disorders. Biochem Pharmacol 2018; 157:85-96. [DOI: 10.1016/j.bcp.2018.08.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 08/09/2018] [Indexed: 12/19/2022]
|
7
|
Ilyasov AA, Milligan CE, Pharr EP, Howlett AC. The Endocannabinoid System and Oligodendrocytes in Health and Disease. Front Neurosci 2018; 12:733. [PMID: 30416422 PMCID: PMC6214135 DOI: 10.3389/fnins.2018.00733] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cannabinoid-based interventions are being explored for central nervous system (CNS) pathologies such as neurodegeneration, demyelination, epilepsy, stroke, and trauma. As these disease states involve dysregulation of myelin integrity and/or remyelination, it is important to consider effects of the endocannabinoid system on oligodendrocytes and their precursors. In this review, we examine research reports on the effects of the endocannabinoid system (ECS) components on oligodendrocytes and their precursors, with a focus on therapeutic implications. Cannabinoid ligands and modulators of the endocannabinoid system promote cell signaling in oligodendrocyte precursor survival, proliferation, migration and differentiation, and mature oligodendrocyte survival and myelination. Agonist stimulation of oligodendrocyte precursor cells (OPCs) at both CB1 and CB2 receptors counter apoptotic processes via Akt/PI3K, and promote proliferation via Akt/mTOR and ERK pathways. CB1 receptors in radial glia promote proliferation and conversion to progenitors fated to become oligodendroglia, whereas CB2 receptors promote OPC migration in neonatal development. OPCs produce 2-arachidonoylglycerol (2-AG), stimulating cannabinoid receptor-mediated ERK pathways responsible for differentiation to arborized, myelin basic protein (MBP)-producing oligodendrocytes. In cell culture models of excitotoxicity, increased reactive oxygen species, and depolarization-dependent calcium influx, CB1 agonists improved viability of oligodendrocytes. In transient and permanent middle cerebral artery occlusion models of anoxic stroke, WIN55212-2 increased OPC proliferation and maturation to oligodendroglia, thereby reducing cerebral tissue damage. In several models of rodent encephalomyelitis, chronic treatment with cannabinoid agonists ameliorated the damage by promoting OPC survival and oligodendrocyte function. Pharmacotherapeutic strategies based upon ECS and oligodendrocyte production and survival should be considered.
Collapse
Affiliation(s)
- Alexander A Ilyasov
- Graduate Program in Neuroscience, Wake Forest School of Medicine, Winston Salem, NC, United States.,Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Carolanne E Milligan
- Graduate Program in Neuroscience, Wake Forest School of Medicine, Winston Salem, NC, United States.,Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Emily P Pharr
- Graduate Program in Neuroscience, Wake Forest School of Medicine, Winston Salem, NC, United States.,Department of Neurology and Comprehensive Multiple Sclerosis Center, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Allyn C Howlett
- Graduate Program in Neuroscience, Wake Forest School of Medicine, Winston Salem, NC, United States.,Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
8
|
Inhibition of Fatty Acid Amide Hydrolase (FAAH) by Macamides. Mol Neurobiol 2018; 56:1770-1781. [PMID: 29926378 DOI: 10.1007/s12035-018-1115-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/11/2018] [Indexed: 10/28/2022]
Abstract
The pentane extract of the Peruvian plant, Lepidium meyenii (Maca), has been demonstrated to possess neuroprotective activity in previous in vitro and in vivo studies (Pino-Figueroa et al. in Ann N Y Acad Sci 1199:77-85, 2010; Pino-Figueroa et al. in Am J Neuroprot Neuroregener 3:87-92, 2011). This extract contains a number of macamides that may act on the endocannabinoid system (Pino-Figueroa et al. in Ann N Y Acad Sci 1199:77-85, 2010; Pino-Figueroa et al., 2011; Dini et al. in Food Chem 49:347-349, 1994). The aim of this study was to characterize the inhibitory activity of four of these maccamides (N-benzylstearamide, N-benzyloleamide, N-benzyloctadeca-9Z,12Z-dienamide, and N-benzyloctadeca-9Z,12Z,15Z-trienamide) on fatty acid amide hydrolase (FAAH), an enzyme that is responsible for endocannabinoid degradation in the nervous system (Kumar et al. in Anaesthesia 56:1059-1068, 2001). The four compounds were tested at concentrations between 1 and 100 μM, utilizing an FAAH inhibitor screening assay. The results demonstrated concentration-dependent FAAH inhibitory activities for the four macamides tested. N-Benzyloctadeca-9Z,12Z-dienamide demonstrated the highest FAAH inhibitory activity whereas N-benzylstearamide had the lowest inhibitory activity. In addition, N-benzylstearamide, N-benzyloleamide, and N-benzyloctadeca-9Z,12Z-dienamide demonstrated time-dependent inhibition when tested after a pre-incubation period, indicating that the mechanism of inhibition for these compounds most likely is irreversible. Of interest, unsaturation in the fatty acid moiety resulted in greater FAAH inhibitory activity. LC/MS/MS analysis demonstrated that FAAH was able to hydrolyze N-benzyloctadeca-9Z,12Z-dienamide, suggesting that N-benzyloctadeca-9Z,12Z-dienamide is also a slow substrate for FAAH. These results provide useful information about the mechanism of action of Lepidium meyenii and may help with the development of new compounds with FAAH inhibitory or modulatory activity.
Collapse
|
9
|
Zhou J, Burkovskiy I, Yang H, Sardinha J, Lehmann C. CB2 and GPR55 Receptors as Therapeutic Targets for Systemic Immune Dysregulation. Front Pharmacol 2016; 7:264. [PMID: 27597829 PMCID: PMC4992728 DOI: 10.3389/fphar.2016.00264] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/05/2016] [Indexed: 11/25/2022] Open
Abstract
The endocannabinoid system (ECS) is involved in many physiological processes and has been suggested to play a critical role in the immune response and the central nervous system (CNS). Therefore, ECS modulation has potential therapeutic effects on immune dysfunctional disorders, such as sepsis and CNS injury-induced immunodeficiency syndrome (CIDS). In sepsis, excessive release of pro- and anti-inflammatory mediators results in multi-organ dysfunction, failure, and death. In CIDS, an acute CNS injury dysregulates a normally well-balanced interplay between CNS and the immune system, leading to increased patients' susceptibility to infections. In this review, we will discuss potential therapeutic modulation of the immune response in sepsis and CNS injury by manipulation of the ECS representing a novel target for immunotherapy.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Anesthesia, Dalhousie UniversityHalifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie UniversityHalifax, NS, Canada
| | - Ian Burkovskiy
- Department of Anesthesia, Dalhousie UniversityHalifax, NS, Canada
- Department of Pharmacology, Dalhousie UniversityHalifax, NS, Canada
| | - Hyewon Yang
- Department of Anesthesia, Dalhousie UniversityHalifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie UniversityHalifax, NS, Canada
| | - Joel Sardinha
- Department of Anesthesia, Dalhousie UniversityHalifax, NS, Canada
| | - Christian Lehmann
- Department of Anesthesia, Dalhousie UniversityHalifax, NS, Canada
- Department of Microbiology and Immunology, Dalhousie UniversityHalifax, NS, Canada
- Department of Pharmacology, Dalhousie UniversityHalifax, NS, Canada
| |
Collapse
|
10
|
Blanco E, Galeano P, Holubiec MI, Romero JI, Logica T, Rivera P, Pavón FJ, Suarez J, Capani F, Rodríguez de Fonseca F. Perinatal asphyxia results in altered expression of the hippocampal acylethanolamide/endocannabinoid signaling system associated to memory impairments in postweaned rats. Front Neuroanat 2015; 9:141. [PMID: 26578900 PMCID: PMC4630311 DOI: 10.3389/fnana.2015.00141] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 10/19/2015] [Indexed: 01/25/2023] Open
Abstract
Perinatal asphyxia (PA) is an obstetric complication that strongly affects the CNS. The endocannabinoid system (ECS) is a lipid transmitter system involved in several physiological processes including synaptic plasticity, neurogenesis, memory, and mood. Endocannabinoids, and other acylethanolamides (AEs) without endocannabinoid activity, have recently received growing attention due to their potential neuroprotective functions in neurological disorders, including cerebral ischemia. In the present study, we aimed to analyze the changes produced by PA in the major metabolic enzymes and receptors of the ECS/AEs in the hippocampus using a rodent model of PA. To induce PA, we removed uterine horns from ready-to-deliver rats and immersed them into a water bath during 19 min. Animals delivered spontaneously or by cesarean section were employed as controls. At 1 month of age, cognitive functions were assessed and immunohistochemical procedures were carried out to determine the expression of NeuN and glial fibrillary acidic protein, enzymes responsible for synthesis (DAGLα and NAPE-PLD) and degradation (FAAH) of ECS/AEs and their receptors (CB1 and PPARα) in the hippocampus. Postweaned asphyctic rats showed impaired recognition and spatial reference memory that were accompanied by hippocampal astrogliosis and changes in the expression of enzymes and receptors. The most remarkable findings in asphyctic rats were a decrease in the expression of NAPE-PLD and PPARα in both hippocampal areas CA1 and CA3. In addition, postweaned cesarean delivery rats showed an increase in the immunolabeling for FAAH in the hippocampal CA3 area. Since, NAPE-PLD and PPARα are proteins that participate in the biochemical process of AEs, specially the neuroprotective oleoylethanolamide, these results suggest that PA dysregulates this system. These data encourage conducting future studies using AEs as potential neuroprotective compounds in animal models of PA.
Collapse
Affiliation(s)
- Eduardo Blanco
- Unidad de Gestión Clínica de Salud Mental, Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga Málaga, Spain ; Departament de Pedagogia i Psicologia, Facultat d'Educació, Psicologia i Treball Social, Universitat de Lleida Lleida, Spain
| | - Pablo Galeano
- Instituto de Investigaciones Bioquímicas de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas, Fundación Instituto Leloir Buenos Aires, Argentina ; Facultad de Medicina, Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini", Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires Buenos Aires, Argentina
| | - Mariana I Holubiec
- Facultad de Medicina, Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini", Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires Buenos Aires, Argentina
| | - Juan I Romero
- Facultad de Medicina, Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini", Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires Buenos Aires, Argentina
| | - Tamara Logica
- Facultad de Medicina, Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini", Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires Buenos Aires, Argentina
| | - Patricia Rivera
- Unidad de Gestión Clínica de Salud Mental, Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga Málaga, Spain
| | - Francisco J Pavón
- Unidad de Gestión Clínica de Salud Mental, Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga Málaga, Spain
| | - Juan Suarez
- Unidad de Gestión Clínica de Salud Mental, Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga Málaga, Spain
| | - Francisco Capani
- Facultad de Medicina, Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini", Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires Buenos Aires, Argentina
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Laboratorio de Medicina Regenerativa, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, Universidad de Málaga Málaga, Spain
| |
Collapse
|
11
|
Anandamide protects HT22 cells exposed to hydrogen peroxide by inhibiting CB1 receptor-mediated type 2 NADPH oxidase. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2014; 2014:893516. [PMID: 25136404 PMCID: PMC4127243 DOI: 10.1155/2014/893516] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 11/25/2022]
Abstract
Background. Endogenous cannabinoid anandamide (AEA) protects neurons from oxidative injury in rodent models; however the mechanism of AEA-induced neuroprotection remains to be determined. Activation of neuronal NADPH oxidase 2 (Nox2) contributes to oxidative damage of the brain, and inhibition of Nox2 can attenuate cerebral oxidative stress. We aimed to determine whether the neuronal Nox2 was involved in protection mediated by AEA. Methods. The mouse hippocampal neuron cell line HT22 was exposed to hydrogen peroxide (H2O2) to mimic oxidative injury of neurons. The protective effect of AEA was assessed by measuring cell metabolic activity, apoptosis, lactate dehydrogenase (LDH) release, cellular morphology, intracellular reactive oxygen species (ROS), and antioxidant and oxidant levels and Nox2 expression. Results. HT22 cells exposed to H2O2 demonstrated morphological changes, decreased LDH release, reduced metabolic activity, increased levels of intracellular ROS and oxidized glutathione (GSSG), reduced levels of superoxide dismutase (SOD), and reduced glutathione (GSH) and increased expression of Nox2. AEA prevented these effects, a property abolished by simultaneous administration of CB1 antagonist AM251 or CB1-siRNA. Conclusion. Nox2 inhibition is involved in AEA-induced cytoprotection against oxidative stress through CB1 activation in HT22 cells.
Collapse
|
12
|
Fishbein-Kaminietsky M, Gafni M, Sarne Y. Ultralow doses of cannabinoid drugs protect the mouse brain from inflammation-induced cognitive damage. J Neurosci Res 2014; 92:1669-77. [DOI: 10.1002/jnr.23452] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 06/15/2014] [Accepted: 06/16/2014] [Indexed: 01/05/2023]
Affiliation(s)
- Miriam Fishbein-Kaminietsky
- The Adelson Center for the Biology of Addictive Diseases and The Mauerberger Chair in Neuropharmacology; Sackler Faculty of Medicine, Tel-Aviv University; Tel-Aviv Israel
| | - Mikhal Gafni
- The Adelson Center for the Biology of Addictive Diseases and The Mauerberger Chair in Neuropharmacology; Sackler Faculty of Medicine, Tel-Aviv University; Tel-Aviv Israel
| | - Yosef Sarne
- The Adelson Center for the Biology of Addictive Diseases and The Mauerberger Chair in Neuropharmacology; Sackler Faculty of Medicine, Tel-Aviv University; Tel-Aviv Israel
| |
Collapse
|
13
|
Tree KC, Scotto di Perretolo M, Peyronnet J, Cayetanot F. In utero cannabinoid exposure alters breathing and the response to hypoxia in newborn mice. Eur J Neurosci 2014; 40:2196-204. [PMID: 24717006 DOI: 10.1111/ejn.12588] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 01/21/2014] [Accepted: 03/13/2014] [Indexed: 11/26/2022]
Abstract
Cannabis is one of the most commonly used recreational drugs at ages highly correlated with potential pregnancy. Endocannabinoid signalling regulates important stages of neuronal development. When cannabinoid receptors, which are widely distributed through the nervous system, are activated by exogenous cannabinoids, breathing in adult rats is depressed. Here, we show that, in newborn mice, endocannabinoids, through the activation of cannabinoid receptor type 1 (CB1 R), participate in the modulation of respiration and its control. Blocking CB1 Rs at birth suppressed the brake exerted by endocannabinoids on ventilation in basal and in hypoxic conditions. The number of apnoeas and their duration were also minimized by activation of CB1 Rs in normoxic and in hypoxic conditions. However, prenatal cannabis intoxication, caused by a daily injection of WIN55,212-2, in pregnant mice durably modified respiration of the offspring, as shown by hyperventilation in basal conditions, an altered chemoreflex in response to hypoxia, and longer apnoeas. When CB1 Rs were blocked in WIN55,212-2 treated newborns, persistent hyperventilation was still observed, which could partly be explained by a perturbation of the central respiratory network. In fact, in vitro medullary preparations from WIN55,212-2 treated pups, free of peripheral or of supramedullary structures, showed an altered fictive breathing frequency. In conclusion, the endocannabinoid pathway at birth seems to modulate breathing and protect the newborn against apnoeas. However, when exposed prenatally to an excess of cannabinoid, the breathing neuronal network in development seems to be modified, probably rendering the newborn more vulnerable in the face of an unstable environment.
Collapse
Affiliation(s)
- Keda C Tree
- Institut de Neurosciences de la Timone UMR 7289, Aix Marseille Université, CNRS, Marseille, France
| | | | | | | |
Collapse
|
14
|
Yang R, Cui HJ, Wang H, Wang Y, Liu JH, Li Y, Lu Y. N-Stearoyltyrosine Protects Against Glutamate-Induced Oxidative Toxicity by an Apoptosis-Inducing Factor (AIF)-Mediated Caspase-Independent Cell Death Pathway. J Pharmacol Sci 2014; 124:169-79. [DOI: 10.1254/jphs.13184fp] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
15
|
Almukadi H, Wu H, Böhlke M, Kelley CJ, Maher TJ, Pino-Figueroa A. The macamide N-3-methoxybenzyl-linoleamide is a time-dependent fatty acid amide hydrolase (FAAH) inhibitor. Mol Neurobiol 2013; 48:333-9. [PMID: 23853040 DOI: 10.1007/s12035-013-8499-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 06/19/2013] [Indexed: 10/26/2022]
Abstract
The Peruvian plant Lepidium meyenii (Maca) has been shown to possess neuroprotective activity both in vitro and in vivo. Previous studies have also demonstrated the activity of the pentane extract and its macamides, the most representative lipophilic constituents of Maca, in the endocannabinoid system as fatty acid amide hydrolase (FAAH) inhibitors. One of the most active macamides, N-3-methoxybenzyl-linoleamide, was studied to determine its mechanism of interaction with FAAH and whether it has inhibitory activity on mono-acyl glycerol lipase (MAGL), the second enzyme responsible for endocannabinoid degradation. Macamide concentrations from 1 to 100 μM were tested using FAAH and MAGL inhibitor assay methods and showed no effect on MAGL. Tests with other conditions were performed in order to characterize the inhibitory mechanism of FAAH inhibition. N-3-methoxybenzyl-linoleamide displayed significant time-dependent and dose-dependent FAAH inhibitory activity. The mechanism of inhibition was most likely irreversible or slowly reversible. These results suggest the potential application of macamides isolated from Maca as FAAH inhibitors, as they might act on the central nervous system to provide analgesic, anti-inflammatory, or neuroprotective effects, by modulating the release of neurotransmitters.
Collapse
Affiliation(s)
- Haifa Almukadi
- Department of Pharmaceutical Sciences, MCPHS University, 179 Longwood Ave., Boston, MA, 02115, USA
| | | | | | | | | | | |
Collapse
|
16
|
Zogopoulos P, Vasileiou I, Patsouris E, Theocharis S. The neuroprotective role of endocannabinoids against chemical-induced injury and other adverse effects. J Appl Toxicol 2013; 33:246-64. [DOI: 10.1002/jat.2828] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 08/31/2012] [Accepted: 09/01/2012] [Indexed: 12/21/2022]
Affiliation(s)
- Panagiotis Zogopoulos
- 1st Department of Pathology, Medical School; National and Kapodistrian University of Athens; Athens; Greece
| | - Ioanna Vasileiou
- 1st Department of Pathology, Medical School; National and Kapodistrian University of Athens; Athens; Greece
| | - Efstratios Patsouris
- 1st Department of Pathology, Medical School; National and Kapodistrian University of Athens; Athens; Greece
| | - Stamatios Theocharis
- 1st Department of Pathology, Medical School; National and Kapodistrian University of Athens; Athens; Greece
| |
Collapse
|
17
|
Dalous J, Pansiot J, Pham H, Chatel P, Nadaradja C, D'Agostino I, Vottier G, Schwendimann L, Vanneaux V, Charriaut-Marlangue C, Titomanlio L, Gressens P, Larghero J, Baud O. Use of Human Umbilical Cord Blood Mononuclear Cells to Prevent Perinatal Brain Injury: A Preclinical Study. Stem Cells Dev 2013; 22:169-79. [DOI: 10.1089/scd.2012.0183] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jérémie Dalous
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Julien Pansiot
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Hoa Pham
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Paul Chatel
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Céline Nadaradja
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Irene D'Agostino
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Gaëlle Vottier
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Leslie Schwendimann
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Valérie Vanneaux
- Unité de Thérapie Cellulaire et Centre d'Investigation Clinique en Biothérapies CIC-BT501, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Christiane Charriaut-Marlangue
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Luigi Titomanlio
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Pierre Gressens
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
| | - Jérôme Larghero
- Unité de Thérapie Cellulaire et Centre d'Investigation Clinique en Biothérapies CIC-BT501, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Olivier Baud
- INSERM UMR 676, Université Paris-Diderot, PRES Sorbonne Paris-Cité, Hôpital Robert Debré, Assistance Publique-Hôpitaux de Paris, Paris, France
- PremUP Foundation, Paris, France
- NICU, Université Paris-Diderot, Hôpital Robert Debré, APHP, Paris, France
| |
Collapse
|
18
|
Lara-Celador I, Castro-Ortega L, Alvarez A, Goñi-de-Cerio F, Lacalle J, Hilario E. Endocannabinoids reduce cerebral damage after hypoxic-ischemic injury in perinatal rats. Brain Res 2012; 1474:91-9. [PMID: 22841538 DOI: 10.1016/j.brainres.2012.07.045] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Revised: 06/25/2012] [Accepted: 07/23/2012] [Indexed: 12/19/2022]
Abstract
Hypoxic-ischemic (HI) insult during the perinatal period remains as one of the most common causes of brain injury and produces long-term neurological deficits, and there is a growing need for effective therapies. The aim of the present work was to perform a prospective study designed to assess the possible protector effect of two endocannabinoids: 2-arachidonoylglycerol (2AG) and anandamide (AEA) in the brain after HI injury in perinatal rat model. We evaluate their effects on cell death and check several cellular parameters. 7-days-old Wistar rats were assigned to four different experimental groups (n=7-10): Sham, HI, and HI treated with 2AG or AEA. The injury was induced by the left carotid artery ligature and subsequent exposure to 8% O(2) for 120 min. Immediately after the injury, treated groups received a single dose of 2AG (1mg/kg) or AEA (5mg/kg) and then animals were sacrificed 24, 72 h or 7 days after the HI event. Brains fixed by perfusion were stained with Nissl for morphological studies, and non-fixed brains were dissociated and analyzed by flow cytometry to quantify apoptosis, mitochondrial state, intracellular calcium and reactive oxygen species. Our results show that both 2AG and AEA have beneficial effects after HI injury in this rat model, producing a remarkable amelioration of brain injury, reducing apoptotic cell death, contributing to the maintenance of mitochondrial functionality, and improving cellular parameters such as the influx of calcium and ROS production.
Collapse
Affiliation(s)
- Idoia Lara-Celador
- Department of Cell Biology and Histology, School of Medicine and Dentistry, University of the Basque Country, E-48940 Leioa, Vizcaya, Spain.
| | | | | | | | | | | |
Collapse
|
19
|
Long-term behavioral and biochemical effects of an ultra-low dose of Δ9-tetrahydrocannabinol (THC): neuroprotection and ERK signaling. Exp Brain Res 2012; 221:437-48. [DOI: 10.1007/s00221-012-3186-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 07/03/2012] [Indexed: 10/28/2022]
|
20
|
Luchicchi A, Pistis M. Anandamide and 2-arachidonoylglycerol: Pharmacological Properties, Functional Features, and Emerging Specificities of the Two Major Endocannabinoids. Mol Neurobiol 2012; 46:374-92. [DOI: 10.1007/s12035-012-8299-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2012] [Accepted: 07/03/2012] [Indexed: 12/18/2022]
|
21
|
Sarne Y, Asaf F, Fishbein M, Gafni M, Keren O. The dual neuroprotective-neurotoxic profile of cannabinoid drugs. Br J Pharmacol 2012; 163:1391-401. [PMID: 21323910 DOI: 10.1111/j.1476-5381.2011.01280.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Extensive in vitro and in vivo studies have shown that cannabinoid drugs have neuroprotective properties and suggested that the endocannabinoid system may be involved in endogenous neuroprotective mechanisms. On the other hand, neurotoxic effects of cannabinoids in vitro and in vivo were also described. Several possible explanations for these dual, opposite effects of cannabinoids on cellular fate were suggested, and it is conceivable that various factors may determine the final outcome of the cannabinoid effect in vivo. In the current review, we focus on one of the possible reasons for the dual neuroprotective/neurotoxic effects of cannabinoids in vivo, namely, the opposite effects of low versus high doses of cannabinoids. While many studies reported neuroprotective effects of the conventional doses of cannabinoids in various experimental models for acute brain injuries, we have shown that a single administration of an extremely low dose of Δ(9) -tetrahydrocannabinol (THC) (3-4 orders of magnitude lower than the conventional doses) to mice induced long-lasting mild cognitive deficits that affected various aspects of memory and learning. These findings led to the idea that this low dose of THC, which induces minor damage to the brain, may activate preconditioning and/or postconditioning mechanisms and thus will protect the brain from more severe insults. Indeed, our recent findings support this assumption and show that a pre- or a postconditioning treatment with extremely low doses of THC, several days before or after brain injury, provides effective long-term cognitive neuroprotection. The future therapeutical potential of these findings is discussed.
Collapse
Affiliation(s)
- Yosef Sarne
- The Adelson Center for the Biology of Addictive Diseases and The Mauerberger Chair in Neuropharmacology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | | | | | | | | |
Collapse
|
22
|
Marco EM, Laviola G. The endocannabinoid system in the regulation of emotions throughout lifespan: a discussion on therapeutic perspectives. J Psychopharmacol 2012; 26:150-63. [PMID: 21693551 DOI: 10.1177/0269881111408459] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alterations in emotion regulation processes may form the basis of psychopathologies. The endocannabinoid (eCB) system, composed of endogenous ligands, the enzymatic machinery in charge of their metabolism and the specific metabotropic receptors, has emerged as a major neuromodulatory system critically involved in the control of emotional homeostasis and stress responsiveness. Data from animal models indicate that the eCB system plays a key role in brain development, and is probably involved in the control of emotional states from early developmental stages. The present review summarizes the latest information on the role of the eCB system in emotionality and anxiety-related disorders throughout the lifespan. Putative therapeutic strategies based on the pharmacological modulation of this system will be discussed. Given the fact that the pharmacological modulation of the eCB system has recently arisen as a promising strategy in the management of anxiety and mood disorders, the potential efficacy of this pharmacological approach (i.e. blockers of the catabolic pathway) will be discussed, as well as pharmacological alternatives such as modulators of cannabinoid receptors other than the classical CB1 receptor, or administration of other plant-derived compounds (e.g. cannabidiol).
Collapse
Affiliation(s)
- Eva M Marco
- Department of Animal Physiology (Animal Physiology II), Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain.
| | | |
Collapse
|
23
|
Lou ZY, Zhao CB, Xiao BG. Immunoregulation of experimental autoimmune encephalomyelitis by the selective CB1 receptor antagonist. J Neurosci Res 2011; 90:84-95. [DOI: 10.1002/jnr.22721] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 02/23/2011] [Accepted: 05/23/2011] [Indexed: 12/12/2022]
|
24
|
The Cannabinoid WIN 55212-2 Mitigates Apoptosis and Mitochondrial Dysfunction After Hypoxia Ischemia. Neurochem Res 2011; 37:161-70. [DOI: 10.1007/s11064-011-0594-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Revised: 07/29/2011] [Accepted: 09/02/2011] [Indexed: 12/25/2022]
|
25
|
Holopainen IE, Laurén HB. Glutamate signaling in the pathophysiology and therapy of prenatal insults. Pharmacol Biochem Behav 2011; 100:825-34. [PMID: 21443898 DOI: 10.1016/j.pbb.2011.03.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 02/25/2011] [Accepted: 03/16/2011] [Indexed: 01/18/2023]
Abstract
Birth asphyxia and hypoxia-ischemia (HI) are important factors affecting the normal development and maturation of the central nervous system (CNS). Depending on the maturity of the brain, HI-induced damage at different ages is region-selective, the white matter (WM) peripheral to the lateral ventricles being selectively vulnerable to damage in premature infants. As a squeal of primary or secondary HI in the preterm infant, the brain injury comprises periventricular leukomalasia (PVL), accompanied by neuronal and axonal damage, which affects several brain regions. Premature delivery and improved neonatal intensive care have led to a survival rate of about 75% to 90% of infants weighting under 1500g both in Europe and in the United States. However, about 5-10% of these survivors exhibit cerebral palsy (CP), and many have cognitive, behavioral, attentional or socialization deficits. In this review, we first shortly discuss developmental changes in the expression of the excitatory glutamate receptors (GluRs), and then in more detail elucidate the contribution of GluRs to oligodendrocyte (OL) damage both in experimental models and in preterm human infants. Finally, therapeutic interventions targeted at GluRs at the young age are discussed in the light of results obtained from recent experimental HI animal models and from humans.
Collapse
Affiliation(s)
- Irma E Holopainen
- Department of Pharmacology, Drug Development and Therapeutics, and Medicity Research Laboratory, Institute of Biomedicine University of Turku, Tykistökatu 6A, 4th floor, FIN-20014 Turku, Finland.
| | | |
Collapse
|
26
|
Pre- and post-conditioning treatment with an ultra-low dose of Δ9-tetrahydrocannabinol (THC) protects against pentylenetetrazole (PTZ)-induced cognitive damage. Behav Brain Res 2011; 220:194-201. [PMID: 21315768 DOI: 10.1016/j.bbr.2011.02.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/01/2011] [Accepted: 02/02/2011] [Indexed: 12/15/2022]
Abstract
Preconditioning, a phenomenon where a minor noxious stimulus protects from a subsequent more severe insult, and post-conditioning, where the protective intervention is applied following the insult, offer new insight into the neuronal mechanism(s) of neuroprotection and may provide new strategies for the prevention and treatment of brain damage. We have previously reported that a single administration of an extremely low dose of Δ(9)-tetrahydrocannabinol (THC; the psychoactive ingredient of marijuana) to mice induced minor long-lasting cognitive deficits. In the present study we examined the possibility that such a low dose of THC will protect the mice from more severe cognitive deficits induced by the epileptogenic drug pentylenetetrazole (PTZ). THC (0.002 mg/kg, a dose that is 3-4 orders of magnitude lower than the doses that induce the conventional effects of THC) was administered 1-7 days before, or 1-3 days after the injection of PTZ (60 mg/kg). The consequences of this treatment were studied 3-7 weeks later by various behavioral tests that evaluated different aspects of memory and learning. We found that a single administration of THC either before or after PTZ abolished the PTZ-induced long-lasting cognitive deficits. Biochemical studies indicated a concomitant reduction in phosphorylated-ERK (extracellular signal-regulated kinase) in the cerebella of mice 7 weeks following the injection of THC. Our results suggest that a pre- or post-conditioning treatment with extremely low doses of THC, several days before or after brain injury, may provide safe and effective long-term neuroprotection.
Collapse
|
27
|
Griesmaier E, Schlager G, Wegleiter K, Hermann M, Urbanek M, Simbruner G, Keller M. Role of p75NTR in NMDAR-mediated excitotoxic brain injury in neonatal mice. Brain Res 2010; 1355:31-40. [PMID: 20692240 DOI: 10.1016/j.brainres.2010.07.095] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 07/21/2010] [Accepted: 07/27/2010] [Indexed: 11/30/2022]
Abstract
BACKGROUND Perinatal brain injury in preterm infants is a major cause of neurological handicap. The role of the neurotrophin receptor p75 (p75(NTR)) in the pathogenesis and repair of neonatal excitotoxic brain injury is unknown. Depending on a complex interplay of neurotrophin signalling, p75(NTR) can, in addition to its trophic function, also induce apoptosis. HYPOTHESIS We hypothesised that excitotoxicity increases p75(NTR) expression and p75(NTR) knockout (KO) mice have a significantly smaller lesion size upon excitotoxicity as compared to wild-type (WT) mice. METHODS We used an established animal model of neonatal excitotoxic brain injury mimicking several key aspects of human preterm brain damage. We subjected five-day-old WT and KO mice to excitotoxic injury by means of a single intracranial ibotenate injection (N-methyl-D-aspartate receptor agonist, NMDAR) into one brain hemisphere. Lesion size, number of activated caspase-3- and apoptosis-inducing factor (AIF)-positive cells were determined as outcome parameters. Gender analyses were taken into account retrospectively. RESULTS NMDAR-mediated excitotoxicity induced an upregulation of p75(NTR) expression in the peri-lesion area. Lesion size was significantly increased in female KO as compared to male KO animals. Knockout of p75(NTR) reduced the number of activated caspase-3 but not AIF-positive cells after NMDAR-mediated excitotoxic injury independently of gender. CONCLUSION Since NMDAR-mediated excitotoxic brain injury induced p75(NTR) expression and caspase-3-activated apoptosis in p75(NTR) KO animals was decreased, we conclude that activation of p75(NTR) contributes to NMDAR-mediated apoptosis in the neonatal brain. An increase in lesion size in female animals after excitotoxic brain injury suggests that in females p75(NTR) seems to play a dual role.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Apoptosis/drug effects
- Apoptosis/physiology
- Brain Injury, Chronic/chemically induced
- Brain Injury, Chronic/metabolism
- Brain Injury, Chronic/pathology
- Disease Models, Animal
- Female
- Male
- Mice
- Mice, Knockout
- Neurotoxins/toxicity
- Receptors, N-Methyl-D-Aspartate/drug effects
- Receptors, N-Methyl-D-Aspartate/metabolism
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/physiology
Collapse
Affiliation(s)
- Elke Griesmaier
- Department of Paediatrics IV, Neonatology, Neuropaediatrics and Metabolic Diseases, Innsbruck Medical University, Anichstrasse 35, 6020 Innsbruck, Austria.
| | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Half of the most severe cases of cerebral palsy (CP) survive to adulthood, but because this longevity is relatively recent, there is no empirical experience of their life expectancy past middle age. The last 2 decades have seen significant developments in the management of persons with CP, involving specialist services from an increasing number of disciplines that require coordination to maximize their effectiveness. This article provides an overview of CP. The author discusses definitions of CP, its epidemiology, pathologies, and range of possible clinical descriptions, and briefly touches on management and prevention.
Collapse
Affiliation(s)
- Eve Blair
- Division of Population Sciences, Centre for Child Health Research, University of Western Australia at The Telethon Institute for Child Health Research, West Perth, WA 6872, Australia.
| |
Collapse
|
29
|
Tree K, Caravagna C, Hilaire G, Peyronnet J, Cayetanot F. Anandamide centrally depresses the respiratory rhythm generator of neonatal mice. Neuroscience 2010; 170:1098-109. [PMID: 20800658 DOI: 10.1016/j.neuroscience.2010.08.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 08/02/2010] [Accepted: 08/19/2010] [Indexed: 11/26/2022]
Abstract
Endogenous cannabinoid receptors are widely distributed throughout the CNS, including the brainstem, and modulate a variety of functions, including breathing. In adult rats, activation of cannabinoid 1 receptors has been shown to depress breathing. Here in neonatal mice, we used in vitro electrophysiology, pharmacology, and immunohistochemistry to analyse the central effects of the endocannabinoid anandamide (AEA) on the activity of the medullary respiratory rhythm generator (RRG). First of all, in vitro electrophysiology on medullary preparations has revealed that bath application of AEA (30 μM, 15 min) significantly depressed respiratory activity. Secondly, applying pre-treatments with alpha-1 (Prazosin, 5 μM, 10 min) and alpha-2 (Yohimbine, 5 μM, 10 min) adrenoceptor antagonists prior to AEA application abolished the AEA-induced depression of the RRG. Finally, immunostaining revealed a dense network of fibres positive for the cannabinoid 1 receptor in the ventrolateral medulla (VLM), a region known to contain both the RRG and the modulatory A1/C1 catecholaminergic group. Moreover, cannabinoid 1 receptor positive fibres were found in close apposition with A1/C1 catecholaminergic cells, identified by the presence of tyrosine hydroxylase. In regard of our electrophysiological, pharmacological and immunostaining results, we conclude that AEA has a central depressive effect on the neonatal RRG, probably via the medullary A1/C1 catecholaminergic neurons which are already known to modulate the respiratory rhythm generator.
Collapse
Affiliation(s)
- K Tree
- Centre de Recherche en Neurobiologie-Neurophysiologie de Marseille CRN2M, Département de Physiologie Neurovégétative (PNV), Université Paul Cézanne Aix Marseille III, Faculté des Sciences et Techniques St. Jérôme, UMR, 6231 CNRS, Marseille, France
| | | | | | | | | |
Collapse
|
30
|
Melis M, Pistis M. Endocannabinoid signaling in midbrain dopamine neurons: more than physiology? Curr Neuropharmacol 2010; 5:268-77. [PMID: 19305743 PMCID: PMC2644494 DOI: 10.2174/157015907782793612] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 03/30/2007] [Accepted: 04/03/2007] [Indexed: 11/22/2022] Open
Abstract
Different classes of neurons in the CNS utilize endogenous cannabinoids as retrograde messengers to shape afferent activity in a short- and long-lasting fashion. Transient suppression of excitation and inhibition as well as long-term depression or potentiation in many brain regions require endocannabinoids to be released by the postsynaptic neurons and activate presynaptic CB1 receptors. Memory consolidation and/or extinction and habit forming have been suggested as the potential behavioral consequences of endocannabinoid-mediated synaptic modulation. HOWEVER, ENDOCANNABINOIDS HAVE A DUAL ROLE: beyond a physiological modulation of synaptic functions, they have been demonstrated to participate in the mechanisms of neuronal protection under circumstances involving excessive excitatory drive, glutamate excitotoxicity, hypoxia-ischemia, which are key features of several neurodegenerative disorders. In this framework, the recent discovery that the endocannabinoid 2-arachidonoyl-glycerol is released by midbrain dopaminergic neurons, under both physiological synaptic activity to modulate afferent inputs and pathological conditions such as ischemia, is particularly interesting for the possible implication of these molecules in brain functions and dysfunctions. Since dopamine dysfunctions underlie diverse neuropsychiatric disorders including schizophrenia, psychoses, and drug addiction, the importance of better understanding the correlation between an unbalanced endocannabinoid signal and the dopamine system is even greater. Additionally, we will review the evidence of the involvement of the endocannabinoid system in the pathogenesis of Parkinson's disease, where neuroprotective actions of cannabinoid-acting compounds may prove beneficial.The modulation of the endocannabinoid system by pharmacological agents is a valuable target in protection of dopamine neurons against functional abnormalities as well as against their neurodegeneration.
Collapse
Affiliation(s)
- M Melis
- B.B. Brodie Department of Neuroscience and Center of Excellence for the Neurobiology of Addiction, University of Cagliari, Monserrato, 09042, Italy
| | | |
Collapse
|
31
|
Pansiot J, Loron G, Olivier P, Fontaine R, Charriaut-Marlangue C, Mercier JC, Gressens P, Baud O. Neuroprotective effect of inhaled nitric oxide on excitotoxic-induced brain damage in neonatal rat. PLoS One 2010; 5:e10916. [PMID: 20532231 PMCID: PMC2879374 DOI: 10.1371/journal.pone.0010916] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2010] [Accepted: 05/07/2010] [Indexed: 11/18/2022] Open
Abstract
Background Inhaled nitric oxide (iNO) is one of the most promising therapies used in neonates. However, little information is known about its impact on the developing brain submitted to excitotoxic challenge. Methodology/Principal Findings We investigated here the effect of iNO in a neonatal model of excitotoxic brain lesions. Rat pups and their dams were placed in a chamber containing 20 ppm NO during the first week of life. At postnatal day (P)5, rat pups were submitted to intracranial injection of glutamate agonists. At P10, rat pups exposed to iNO exhibited a significant decrease of lesion size in both the white matter and cortical plate compared to controls. Microglia activation and astrogliosis were found significantly decreased in NO-exposed animals. This neuroprotective effect was associated with a significant decrease of several glutamate receptor subunits expression at P5. iNO was associated with an early (P1) downregulation of pCREB/pAkt expression and induced an increase in pAkt protein concentration in response to excitotoxic challenge (P7). Conclusion This study is the first describe and investigate the neuroprotective effect of iNO in neonatal excitotoxic-induced brain damage. This effect may be mediated through CREB pathway and subsequent modulation of glutamate receptor subunits expression.
Collapse
Affiliation(s)
- Julien Pansiot
- INSERM, Hôpital Robert Debré, Paris, France
- INSERM, UMR 676, Hôpital Robert Debré, Paris, France
| | - Gauthier Loron
- INSERM, Hôpital Robert Debré, Paris, France
- INSERM, UMR 676, Hôpital Robert Debré, Paris, France
| | - Paul Olivier
- INSERM, Hôpital Robert Debré, Paris, France
- INSERM, UMR 676, Hôpital Robert Debré, Paris, France
| | - Romain Fontaine
- INSERM UMR 711, Université Pierre et Marie Curie, Faculté de Médecine, Hôpital de la Salpêtrière, Paris, France
| | | | | | - Pierre Gressens
- INSERM, UMR 676, Hôpital Robert Debré, Paris, France
- APHP, Neonatal Intensive Care Unit, Hôpital Robert Debré, Paris, France
| | - Olivier Baud
- INSERM, Hôpital Robert Debré, Paris, France
- INSERM, UMR 676, Hôpital Robert Debré, Paris, France
- APHP, Neonatal Intensive Care Unit, Hôpital Robert Debré, Paris, France
- PremUP Foundation, Paris, France
- * E-mail:
| |
Collapse
|
32
|
Inhibition by anandamide of 6-hydroxydopamine-induced cell death in PC12 cells. Int J Cell Biol 2010; 2010:818497. [PMID: 20182544 PMCID: PMC2825649 DOI: 10.1155/2010/818497] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 10/22/2009] [Accepted: 11/10/2009] [Indexed: 11/17/2022] Open
Abstract
6-hydroxydopamine (6-OHDA) is a selective neurotoxin that is widely used to investigate cell death and protective strategies in models of Parkinson's disease. Here, we investigated the effects of the endogenous cannabinoid, anandamide, on 6-OHDA-induced toxicity in rat adrenal phaeochromocytoma PC12 cells. Morphological analysis and caspase-3 activity assay revealed that anandamide inhibited 6-OHDA-induced apoptosis. The protection was not affected by antagonists of either cannabinoid receptors (CB1 or CB2) or the vanilloid receptor TRPV1. Anandamide-dependent protection was reduced by pretreatment with LY294002 (inhibitor of phosphatidylinositol 3-kinase, PI3K) and unaffected by U0126 (inhibitor of extracellularly-regulated kinase). Interestingly, phosphorylation of c-Jun-NH2-terminal kinase (JNK) in cells exposed to 6-OHDA was strongly reduced by anandamide pre-treatment. Furthermore, 6-OHDA induced c-Jun activation and increased Bim expression, both of which were inhibited by anandamide. Together, these data demonstrate antiapoptotic effects of anandamide and also suggest a role for activation of PI3K and inhibition of JNK signalling in anandamide-mediated protection against 6-OHDA.
Collapse
|
33
|
Duncan RS, Chapman KD, Koulen P. The neuroprotective properties of palmitoylethanolamine against oxidative stress in a neuronal cell line. Mol Neurodegener 2009; 4:50. [PMID: 20003317 PMCID: PMC2799406 DOI: 10.1186/1750-1326-4-50] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 12/10/2009] [Indexed: 01/16/2023] Open
Abstract
Background N-acylethanolamines (NAEs) are lipids upregulated in response to cell and tissue injury and are involved in cytoprotection. Arachidonylethanolamide (AEA) is a well characterized NAE that is an endogenous ligand at cannabinoid and vanilloid receptors, but it exists in small quantities relative to other NAE types. The abundance of other NAE species, such as palmitoylethanolamine (PEA), together with their largely unknown function and receptors, has prompted us to examine the neuroprotective properties and mechanism of action of PEA. We hypothesized that PEA protects HT22 cells from oxidative stress and activates neuroprotective kinase signaling pathways. Results Indeed PEA protected HT22 cells from oxidative stress in part by mediating an increase in phosphorylated Akt (pAkt) and ERK1/2 immunoreactivity as well as pAkt nuclear translocation. These changes take place within a time frame consistent with neuroprotection. Furthermore, we determined that changes in pAkt immunoreactivity elicited by PEA were not mediated by activation of cannabinoid receptor type 2 (CB2), thus indicating a novel mechanism of action. These results establish a role for PEA as a neuroprotectant against oxidative stress, which occurs in a variety of neurodegenerative diseases. Conclusions The results from this study reveal that PEA protects HT22 cells from oxidative stress and alters the localization and expression levels of kinases known to be involved in neuroprotection by a novel mechanism. Overall, these results identify PEA as a neuroprotectant with potential as a possible therapeutic agent in neurodegenerative diseases involving oxidative stress.
Collapse
Affiliation(s)
- R Scott Duncan
- Departments of Basic Medical Science and Ophthalmology, University of Missouri - Kansas City, 2411 Holmes St, Kansas City, MO 64108-2792, USA.
| | | | | |
Collapse
|
34
|
Intracellular mechanisms of N-acylethanolamine-mediated neuroprotection in a rat model of stroke. Neuroscience 2009; 166:252-62. [PMID: 19963043 DOI: 10.1016/j.neuroscience.2009.11.069] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 11/11/2009] [Accepted: 11/29/2009] [Indexed: 11/23/2022]
Abstract
N-acyl ethanolamines (NAEs) are endogenous lipids that are synthesized in response to tissue injury, including ischemia and stroke, suggesting they may exhibit neuroprotective properties. We hypothesized that NAE 16:0 (palmitoylethanolamine) is neuroprotective against ischemia-reperfusion injury in rats, a widely employed model of stroke, and that neuroprotection is mediated through an intracellular mechanism independent of known NAE receptors. Administration of NAE 16:0 from 30 min before to 2 h after stroke significantly reduced cortical and subcortical infarct volume, and correlated with an improvement of the neurological phenotype, as assessed by the neurological deficit score. We here show that NAE 16:0-mediated neuroprotection was independent of cannabinoid (CB1) and vanilloid (VR1) receptor activation, known NAE receptors on the plasma membrane, as determined by inclusion of specific inhibitors. The inclusion of an NAE uptake inhibitor (AM404), however, completely reversed NAE 16:0-mediated neuroprotection, suggesting that NAE 16:0s effects are through an intracellular mechanism. NAE 16:0 produced a significant reduction in the number of cells undergoing apoptosis and reversed ischemia-induced upregulation of several proteins, including inducible nitric oxide synthase and transcription factor NFkappaB. Our findings suggest that NAE 16:0-mediated neuroprotection is due to the reduction of neuronal apoptosis and inflammation in the brain.
Collapse
|
35
|
Anavi-Goffer S, Mulder J. The polarised life of the endocannabinoid system in CNS development. Chembiochem 2009; 10:1591-8. [PMID: 19533710 DOI: 10.1002/cbic.200800827] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The spatiotemporal expression of cannabinoid receptors and endocannabinoid-metabolising enzymes during brain development guides major developmental processes including neurogenesis, cell differentiation, cell migration, neuronal specification and synaptogenesis.Endocannabinoids (eCBs) play an important role in fine-tuning neurotransmission and have recently been shown to play an important role in brain development. The spatiotemporal expression of cannabinoid receptors and endocannabinoid-metabolising enzymes during development guides major developmental processes including neurogenesis, cell differentiation, cell migration, neuronal specification and synaptogenesis. Furthermore, pharmacological experiments and transgenic animal models have shown the impact of disrupted eCB signalling on normal brain development and revealed the danger of both cannabis abuse and exposure to cannabinoid drugs during embryonic development, childhood and adolescence. In this review, we focus on the dynamic expression of eCB components and the physiological role eCBs play during brain development.
Collapse
Affiliation(s)
- Sharon Anavi-Goffer
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | |
Collapse
|
36
|
Griesmaier E, Keller M. Neuroprotective strategies in excitotoxic brain injury: potential applications to the preterm brain. FUTURE NEUROLOGY 2009. [DOI: 10.2217/fnl.09.28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Neuronal and oligodendroglial cell death owing to increased glutamate levels plays an important role in the pathophysiology of hypoxic-, ischemic- and inflammation-mediated brain injury as well as in disorders such as epilepsy, Alzheimer’s, Parkinson’s or Huntington’s disease. In addition, excitotoxic brain injury is known to be a major contributing factor to brain injury in preterm infants. Excitotoxicity is characterized as excessive glutamatergic activation of postsynaptic receptors that consequently leads to cell injury and cell death. The major excitatory amino acid neurotransmitter is glutamate. Glutamate plays a key role in brain development, affecting progenitor cell differentiation, proliferation, migration and survival. In physiological conditions the presence of glutamate in the synapse is regulated by ATP-dependent glutamate transporters in neurons and glial cells, with astrocytes being responsible for a major part of glutamate uptake in the brain. In pathologic circumstances the function of the transporters is impaired, leading to glutamate accumulation in the synaptic cleft and in turn excessive activation of postsynaptic glutamate receptors with subsequent massive Ca2+ influx, activation of neuronal nitric oxide synthase, translocation of proapoptotic genes to the mitochondria, mitochondrial dysfunction, release of cytochrome C into the cytosol, activation of caspases and subsequent cell death. Based on the pathogenic concept of an overactivation of the excitatory pathways, glutamate receptors have been a longstanding therapeutic target for rational drug design. This article reviews the pathophysiology of excitotoxic brain injury in the example of preterm brain injury, as well as current research on therapeutic antiexcitotoxic strategies.
Collapse
Affiliation(s)
- Elke Griesmaier
- Department of Pediatrics IV, Medical University Innsbruck, Austria, Anichstr. 35, 6020 Innsbruck, Austria
| | - Matthias Keller
- Department of Pediatrics I University Hospital Essen, Hufelandstraße 55, 45147 Essen Germany
| |
Collapse
|
37
|
Armstrong C, Morgan RJ, Soltesz I. Pursuing paradoxical proconvulsant prophylaxis for epileptogenesis. Epilepsia 2009; 50:1657-69. [PMID: 19552655 DOI: 10.1111/j.1528-1167.2009.02173.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
There are essentially two potential treatment options for any acquired disorder: symptomatic or prophylactic. For acquired epilepsies that follow a variety of different brain insults, there remains a complete lack of prophylactic treatment options, whereas at the same time these epilepsies are notoriously resistant, once they have emerged, to symptomatic treatments with antiepileptic drugs. The development of prophylactic strategies is logistically challenging, both for basic researchers and clinicians. Nevertheless, cannabinoid-targeting drugs provide a very interesting example of a system within the central nervous system (CNS) that can have very different acute and long-term effects on hyperexcitability and seizures. In this review, we outline research on cannabinoids suggesting that although cannabinoid antagonists are acutely proconvulsant, they may have beneficial effects on long-term hyperexcitability following brain insults of multiple etiologies, making them promising candidates for further investigation as prophylactics against acquired epilepsy. We then discuss some of the implications of this finding on future attempts at prophylactic treatments, specifically, the very short window within which prevention may be possible, the possibility that traditional anticonvulsants may interfere with prophylactic strategies, and the importance of moving beyond anticonvulsants-even to proconvulsants-to find the ideal preventative strategy for acquired epilepsy.
Collapse
Affiliation(s)
- Caren Armstrong
- Department of Anatomy and Neurobiology, University of California, Irvine, California, USA.
| | | | | |
Collapse
|
38
|
Zhang M, Martin BR, Adler MW, Razdan RJ, Kong W, Ganea D, Tuma RF. Modulation of cannabinoid receptor activation as a neuroprotective strategy for EAE and stroke. J Neuroimmune Pharmacol 2009; 4:249-59. [PMID: 19255856 PMCID: PMC2855650 DOI: 10.1007/s11481-009-9148-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 02/04/2009] [Indexed: 12/25/2022]
Abstract
Recognition of the importance of the endocannabinoid system in both homeostasis and pathologic responses raised interest recently in the development of therapeutic agents based on this system. The CB(2) receptor, a component of the endocannabinoid system, has significant influence on immune function and inflammatory responses. Inflammatory responses are major contributors to central nervous system (CNS) injury in a variety of diseases. In this report, we present evidence that activation of CB(2) receptors, by selective CB(2) agonists, reduces inflammatory responses that contribute to CNS injury. The studies demonstrate neuroprotective effects in experimental autoimmune encephalomyelitis, a model of multiple sclerosis, and in a murine model of cerebral ischemia/reperfusion injury. In both cases, CB(2) receptor activation results in reduced white cell rolling and adhesion to cerebral microvessels, a reduction in immune cell invasion, and improved neurologic function after insult. In addition, administration of the CB(1) antagonist SR141716A reduces infarct size following ischemia/reperfusion injury. Administration of both a selective CB(2) agonist and a CB(1) antagonist has the unique property of increasing blood flow to the brain during the occlusion period, suggesting an effect on collateral blood flow. In summary, selective CB(2) receptor agonists and CB(1) receptor antagonists have significant potential for neuroprotection in animal models of two devastating diseases that currently lack effective treatment options.
Collapse
Affiliation(s)
- Ming Zhang
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Billy R. Martin
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Martin W. Adler
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | - Weimin Kong
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Doina Ganea
- Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Ronald F. Tuma
- Center for Substance Abuse Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| |
Collapse
|
39
|
Seierstad M, Breitenbucher JG. Discovery and development of fatty acid amide hydrolase (FAAH) inhibitors. J Med Chem 2009; 51:7327-43. [PMID: 18983142 DOI: 10.1021/jm800311k] [Citation(s) in RCA: 144] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Mark Seierstad
- Johnson & Johnson Pharmaceutical Research and Development, L.L.C., 3210 Merryfield Row, San Diego, California 92121, USA
| | | |
Collapse
|
40
|
The endocannabinoid system is modulated in response to spinal cord injury in rats. Neurobiol Dis 2009; 33:57-71. [DOI: 10.1016/j.nbd.2008.09.015] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 09/03/2008] [Accepted: 09/09/2008] [Indexed: 01/22/2023] Open
|
41
|
Correa FG, Mestre L, Docagne F, Borrell J, Guaza C. The endocannabinoid anandamide from immunomodulation to neuroprotection. Implications for multiple sclerosis. VITAMINS AND HORMONES 2009; 81:207-30. [PMID: 19647114 DOI: 10.1016/s0083-6729(09)81009-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Over the last decade, the endocannabinoid system (ECS) has emerged as a potential target for multiple sclerosis (MS) management. A growing amount of evidence suggests that cannabinoids may be neuroprotective during CNS inflammation. Advances in the understanding of the physiology and pharmacology of the ECS have potentiated the interest of several components of this system as useful biological targets for disease management. Alterations of the ECS have been recently implicated in a number of neuroinflammatory and neurodegenerative conditions, so that the pharmacological modulation of cannabinoid (CB) receptors and/or of the enzymes controlling synthesis, transport, and degradation of these lipid mediators is considered an option to treat several neurological diseases. This chapter focuses on our current understanding of the function of anandamide (AEA), its biological and therapeutic implications, as well as a description of its effects on neuroimmune modulation.
Collapse
Affiliation(s)
- Fernando G Correa
- Functional and Systems Neurobiology Department, Cajal Institute, CSIC, Avda Doctor Arce, Madrid, Spain
| | | | | | | | | |
Collapse
|
42
|
Ferriero DM. Cannabinoids--can what hurts you make you stronger? Commentary on the article by Alvarez et al. on page 653. Pediatr Res 2008; 64:590-1. [PMID: 19034198 DOI: 10.1203/pdr.0b013e3181894a3e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Donna M Ferriero
- Department of Neurology, University of California, San Francisco, CA 94143-0663, USA.
| |
Collapse
|
43
|
Abstract
The human costs of stroke are very large and growing; it is the third largest cause of death in the United States and survivors are often faced with loss of ability to function independently. There is a large need for therapeutic approaches that act to protect neurons from the injury produced by ischemia and reperfusion. The goal of this review is to introduce and discuss the available data that endogenous cannabinoid signaling is altered during ischemia and that it contributes to the consequences of ischemia-induced injury. Overall, the available data suggest that inhibition of CB1 receptor activation together with increased CB2 receptor activation produces beneficial effects.
Collapse
Affiliation(s)
- Cecilia J Hillard
- Department of Pharmacology and Toxicology, Medical College of Wisconsin; Milwaukee, WI 53226, USA.
| |
Collapse
|
44
|
Vulnerability of white matter towards antenatal hypoxia is linked to a species-dependent regulation of glutamate receptor subunits. Proc Natl Acad Sci U S A 2008; 105:16779-84. [PMID: 18922769 DOI: 10.1073/pnas.0803004105] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
White-matter damage is a leading cause of neurological handicap. Although hypoxia-ischemia and excitotoxicity are major pathogenic factors, a role for genetic influences was suggested recently. Thus, protracted gestational hypoxia was associated with white-matter damage (WMD) in rat pups but not in mouse pups. Indeed, microglial activation and vessel-wall density on postnatal days (P)1 and P10 were found increased in both mouse and rat pups, but cell death, astrogliosis, and myelination were only significantly altered in hypoxic rat pups. We investigated whether this species-related difference was ascribable to effects of antenatal hypoxia on the expression of glutamate receptor subunits by using immunocytochemistry, PCR, and excitotoxic double hit insult. Quantitative PCR in hypoxic mouse pups on P1 showed 2- to 4-fold down-regulation of the AMPA-receptor subunits -1, 2, and -4; of the kainate-receptor subunit GluR7; and of the metabotropic receptor subunits mGluR1, -2, -3, -5, and -7. None of the glutamate-receptor subunits was down-regulated in the hypoxic rat pups. NR2B was the only NMDA-receptor subunit that was down-regulated in hypoxic mice but not in hypoxic rat on P1. Ifenprodil administration to induce functional inhibition of NMDA containing NR2B-subunit receptors prevented hypoxia-induced myelination delay in rat pups. Intracerebral injection of a glutamate agonist produced a larger decrease in ibotenate-induced excitotoxic lesions in hypoxic mouse pups than in normoxic mouse pups. Gestational hypoxia may regulate the expression of specific glutamate-receptor subunits in fetal mice but not in fetal rats. Therefore, genetic factors may influence the susceptibility of rodents to WMD.
Collapse
|
45
|
Youssef FF, Ramchandani J, Manswell S, McRae A. Adult-onset calorie restriction attenuates kainic acid excitotoxicity in the rat hippocampal slice. Neurosci Lett 2007; 431:118-22. [PMID: 18179872 DOI: 10.1016/j.neulet.2007.11.064] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Revised: 11/04/2007] [Accepted: 11/11/2007] [Indexed: 10/22/2022]
Abstract
Lifelong calorie restriction is the only known intervention that has been shown to consistently increase life span and reduce the effects of aging on the brain. Given the difficulties of replicating lifelong calorie restriction within human populations, we have sought to assess the effects of short-term adult-onset calorie restriction upon acute excitotoxic insults in the rat hippocampus. Adult animals (approximately 6 months of age) underwent calorie restriction (alternate day feeding) for 7-10 weeks. Utilizing both electrophysiological and immunocytochemical techniques, we report that calorie restriction had no effect upon long-term potentiation (LTP), a measure of neuronal function. In control animals, application of kainic acid (20 microM) resulted in only 35% recovery of CA1 population spikes post-insult. However calorie-restricted animals showed significantly improved recovery after kainic acid treatment (64%). This data was supported by immunocytochemical studies which noted widespread loss of microtubule-associated protein (MAP 2) immunoreactivity in control slices following treatment with kainic acid; however MAP 2 staining was preserved in the CA1 and CA3 regions of calorie-restricted animals. Interestingly there was no significant difference in the recovery of population spikes between groups when slices were treated with N-methyl-d-aspartate (15 microM). We conclude that short-term adult-onset calorie restriction does not alter normal neuronal function and serves to protect the hippocampus from acute kainic acid excitotoxicity.
Collapse
Affiliation(s)
- Farid F Youssef
- Department of Preclinical Sciences, The University of the West Indies, St. Augustine Campus, Trinidad and Tobago.
| | | | | | | |
Collapse
|
46
|
Wolfberg AJ, Dammann O, Gressens P. Anti-inflammatory and immunomodulatory strategies to protect the perinatal brain. Semin Fetal Neonatal Med 2007; 12:296-302. [PMID: 17418653 DOI: 10.1016/j.siny.2007.01.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Infection and inflammation contribute to perinatal brain damage, particularly to the white matter. Although combating perinatal inflammation can be dangerous, because inflammation might have beneficial effects for mother and fetus, it is worthwhile reviewing potential anti-inflammatory neuroprotective compounds, along with their potential adverse effects. Further research on the possible neuroprotective roles of existing medications and substances is necessary.
Collapse
Affiliation(s)
- Adam J Wolfberg
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Tufts New England Medical Center, Boston, MA, USA
| | | | | |
Collapse
|
47
|
Youssef FF, Hormuzdi SG, Irving AJ, Frenguelli BG. Cannabinoid modulation of neuronal function after oxygen/glucose deprivation in area CA1 of the rat hippocampus. Neuropharmacology 2007; 52:1327-35. [PMID: 17382973 DOI: 10.1016/j.neuropharm.2006.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Revised: 12/12/2006] [Accepted: 12/14/2006] [Indexed: 01/17/2023]
Abstract
Endocannabinoids released during cerebral ischemia have been implicated as neuroprotective agents. We assessed the role of cannabinoid receptors in modulating the response of neurons to oxygen/glucose deprivation (OGD), a model for in vitro ischemia, in rat hippocampal slices using extracellular recording techniques. Under control conditions, 15 min OGD resulted in only 50% recovery of CA1 field excitatory postsynaptic potentials (fEPSPs) 60 min post-insult. This post-OGD depression of function was primarily NMDA receptor-dependent as the NMDA receptor antagonist MK-801 (50 microM) promoted recovery of synaptic transmission to 76% of the baseline. Treatment with the CB1 receptor antagonist AM251 (1 microM), which prevented the depression of excitatory synaptic transmission caused by WIN55,212-2 (1 microM), also markedly enhanced recovery of function (71% of control). The enhanced recovery after OGD in the presence of AM251 was independent of both GABA(A) receptors and NMDA receptors since co-application of AM251 with either bicuculline (10 microM) or MK-801 (50 microM) did not alter recovery, or further improved recovery, respectively. These results suggest endocannabinoids released during OGD may modulate synaptic transmission and post-OGD neuronal outcome via activation of an AM251-sensitive cannabinoid receptor.
Collapse
Affiliation(s)
- Farid F Youssef
- Department of Preclinical Sciences, Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago, W.I.
| | | | | | | |
Collapse
|
48
|
Keller M, Yang J, Griesmaier E, Gorna A, Sarkozy G, Urbanek M, Gressens P, Simbruner G. Erythropoietin is neuroprotective against NMDA-receptor-mediated excitotoxic brain injury in newborn mice. Neurobiol Dis 2006; 24:357-66. [PMID: 16959492 DOI: 10.1016/j.nbd.2006.07.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2006] [Revised: 06/23/2006] [Accepted: 07/17/2006] [Indexed: 11/23/2022] Open
Abstract
Using an established mouse model of human periventricular leukomalacia, we investigated whether EPO could reduce excitotoxic damage. When administered 1 h following intracerebral injection of 10 microg ibotenic acid at day 5 of life, both a single injection of EPO (5000 IU/kg bw) and repetitive administrations of EPO reduced white and gray matter lesion size. The therapeutic window for protection was small as the protective effect of EPO was lost when EPO administration was delayed to 4 h post-insult. EPO-mediated upregulation of EPO-R, but not EPO, mRNA was observed within 4 h of the excitotoxic insult. The EPO effect was gender independent. Minor hematopoetic effects were observed following EPO treatment. We conclude that a single dose of EPO is sufficient to reduce excitotoxic brain injury and may therefore possess therapeutic relevance in the clinical setting.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Brain/drug effects
- Brain/metabolism
- Brain/physiopathology
- Cytoprotection/drug effects
- Cytoprotection/physiology
- Disease Models, Animal
- Drug Administration Schedule
- Erythropoietin/pharmacology
- Erythropoietin/therapeutic use
- Female
- Glutamic Acid/metabolism
- Humans
- Ibotenic Acid/antagonists & inhibitors
- Ibotenic Acid/metabolism
- Infant, Newborn
- Injections, Intraventricular
- Leukomalacia, Periventricular/drug therapy
- Leukomalacia, Periventricular/metabolism
- Leukomalacia, Periventricular/physiopathology
- Male
- Mice
- Nerve Degeneration/drug therapy
- Nerve Degeneration/physiopathology
- Nerve Degeneration/prevention & control
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Neurotoxins/antagonists & inhibitors
- Neurotoxins/metabolism
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptors, Erythropoietin/genetics
- Receptors, N-Methyl-D-Aspartate/agonists
- Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
- Time Factors
Collapse
Affiliation(s)
- Matthias Keller
- Department of Pediatrics IV, Neonatology, Neuropediatrics and Metabolic Diseases, Medical University Innsbruck, Austria, and INSERM U 676 and Service de Neurologie Pediatrique, Hopital Robert Debre 48, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Pacher P, Bátkai S, Kunos G. The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol Rev 2006; 58:389-462. [PMID: 16968947 PMCID: PMC2241751 DOI: 10.1124/pr.58.3.2] [Citation(s) in RCA: 1525] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The recent identification of cannabinoid receptors and their endogenous lipid ligands has triggered an exponential growth of studies exploring the endocannabinoid system and its regulatory functions in health and disease. Such studies have been greatly facilitated by the introduction of selective cannabinoid receptor antagonists and inhibitors of endocannabinoid metabolism and transport, as well as mice deficient in cannabinoid receptors or the endocannabinoid-degrading enzyme fatty acid amidohydrolase. In the past decade, the endocannabinoid system has been implicated in a growing number of physiological functions, both in the central and peripheral nervous systems and in peripheral organs. More importantly, modulating the activity of the endocannabinoid system turned out to hold therapeutic promise in a wide range of disparate diseases and pathological conditions, ranging from mood and anxiety disorders, movement disorders such as Parkinson's and Huntington's disease, neuropathic pain, multiple sclerosis and spinal cord injury, to cancer, atherosclerosis, myocardial infarction, stroke, hypertension, glaucoma, obesity/metabolic syndrome, and osteoporosis, to name just a few. An impediment to the development of cannabinoid medications has been the socially unacceptable psychoactive properties of plant-derived or synthetic agonists, mediated by CB(1) receptors. However, this problem does not arise when the therapeutic aim is achieved by treatment with a CB(1) receptor antagonist, such as in obesity, and may also be absent when the action of endocannabinoids is enhanced indirectly through blocking their metabolism or transport. The use of selective CB(2) receptor agonists, which lack psychoactive properties, could represent another promising avenue for certain conditions. The abuse potential of plant-derived cannabinoids may also be limited through the use of preparations with controlled composition and the careful selection of dose and route of administration. The growing number of preclinical studies and clinical trials with compounds that modulate the endocannabinoid system will probably result in novel therapeutic approaches in a number of diseases for which current treatments do not fully address the patients' need. Here, we provide a comprehensive overview on the current state of knowledge of the endocannabinoid system as a target of pharmacotherapy.
Collapse
Affiliation(s)
- Pál Pacher
- Laboratory of Physiological Studies, National Institute of Alcohol Abuse and Alcoholism, National Institutes of Health, 5625 Fishers Lane, Room 2S-24, Bethesda, MD 20892-9413, USA
| | | | | |
Collapse
|