1
|
Carrascosa AJ, Navarrete F, Saldaña R, García-Gutiérrez MS, Montalbán B, Navarro D, Gómez-Guijarro FM, Gasparyan A, Murcia-Sánchez E, Torregrosa AB, Pérez-Doblado P, Gutiérrez L, Manzanares J. Cannabinoid Analgesia in Postoperative Pain Management: From Molecular Mechanisms to Clinical Reality. Int J Mol Sci 2024; 25:6268. [PMID: 38892456 PMCID: PMC11172912 DOI: 10.3390/ijms25116268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/26/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
Postoperative pain (POP) is a challenging clinical phenomenon that affects the majority of surgical patients and demands effective management to mitigate adverse outcomes such as persistent pain. The primary goal of POP management is to alleviate suffering and facilitate a seamless return to normal function for the patient. Despite compelling evidence of its drawbacks, opioid analgesia remains the basis of POP treatment. Novel therapeutic approaches rely on multimodal analgesia, integrating different pharmacological strategies to optimize efficacy while minimizing adverse effects. The recognition of the imperative role of the endocannabinoid system in pain regulation has prompted the investigation of cannabinoid compounds as a new therapeutic avenue. Cannabinoids may serve as adjuvants, enhancing the analgesic effects of other drugs and potentially replacing or at least reducing the dependence on other long-term analgesics in pain management. This narrative review succinctly summarizes pertinent information on the molecular mechanisms, clinical therapeutic benefits, and considerations associated with the plausible use of various cannabinoid compounds in treating POP. According to the available evidence, cannabinoid compounds modulate specific molecular mechanisms intimately involved in POP. However, only two of the eleven clinical trials that evaluated the efficacy of different cannabinoid interventions showed positive results.
Collapse
Affiliation(s)
- Antonio J. Carrascosa
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Raquel Saldaña
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - María S. García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Belinda Montalbán
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Daniela Navarro
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Fernando M. Gómez-Guijarro
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Elena Murcia-Sánchez
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Abraham B. Torregrosa
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Paloma Pérez-Doblado
- Servicio de Anestesiologia y Reanimación, Hospital Universitario 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain; (A.J.C.); (R.S.); (B.M.); (F.M.G.-G.); (E.M.-S.); (P.P.-D.)
| | - Luisa Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, Avda de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain; (F.N.); (M.S.G.-G.); (D.N.); (A.G.); (A.B.T.); (L.G.)
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| |
Collapse
|
2
|
Zeng F, Wade A, Harbert K, Patel S, Holley JS, Dehghanpuor CK, Hopwood T, Marino S, Sophocleous A, Idris AI. Classical cannabinoid receptors as target in cancer-induced bone pain: a systematic review, meta-analysis and bioinformatics validation. Sci Rep 2024; 14:5782. [PMID: 38461339 PMCID: PMC10924854 DOI: 10.1038/s41598-024-56220-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 03/04/2024] [Indexed: 03/11/2024] Open
Abstract
To test the hypothesis that genetic and pharmacological modulation of the classical cannabinoid type 1 (CB1) and 2 (CB2) receptors attenuate cancer-induced bone pain, we searched Medline, Web of Science and Scopus for relevant skeletal and non-skeletal cancer studies from inception to July 28, 2022. We identified 29 animal and 35 human studies. In mice, a meta-analysis of pooled studies showed that treatment of osteolysis-bearing males with the endocannabinoids AEA and 2-AG (mean difference [MD] - 24.83, 95% confidence interval [95%CI] - 34.89, - 14.76, p < 0.00001) or the synthetic cannabinoid (CB) agonists ACPA, WIN55,212-2, CP55,940 (CB1/2-non-selective) and AM1241 (CB2-selective) (MD - 28.73, 95%CI - 45.43, - 12.02, p = 0.0008) are associated with significant reduction in paw withdrawal frequency. Consistently, the synthetic agonists AM1241 and JWH015 (CB2-selective) increased paw withdrawal threshold (MD 0.89, 95%CI 0.79, 0.99, p < 0.00001), and ACEA (CB1-selective), AM1241 and JWH015 (CB2-selective) reduced spontaneous flinches (MD - 4.85, 95%CI - 6.74, - 2.96, p < 0. 00001) in osteolysis-bearing male mice. In rats, significant increase in paw withdrawal threshold is associated with the administration of ACEA and WIN55,212-2 (CB1/2-non-selective), JWH015 and AM1241 (CB2-selective) in osteolysis-bearing females (MD 8.18, 95%CI 6.14, 10.21, p < 0.00001), and treatment with AM1241 (CB2-selective) increased paw withdrawal thermal latency in males (mean difference [MD]: 3.94, 95%CI 2.13, 5.75, p < 0.0001), confirming the analgesic capabilities of CB1/2 ligands in rodents. In human, treatment of cancer patients with medical cannabis (standardized MD - 0.19, 95%CI - 0.35, - 0.02, p = 0.03) and the plant-derived delta-9-THC (20 mg) (MD 3.29, CI 2.24, 4.33, p < 0.00001) or its synthetic derivative NIB (4 mg) (MD 2.55, 95%CI 1.58, 3.51, p < 0.00001) are associated with reduction in pain intensity. Bioinformatics validation of KEGG, GO and MPO pathway, function and process enrichment analysis of mouse, rat and human data revealed that CB1 and CB2 receptors are enriched in a cocktail of nociceptive and sensory perception, inflammatory, immune-modulatory, and cancer pathways. Thus, we cautiously conclude that pharmacological modulators of CB1/2 receptors show promise in the treatment of cancer-induced bone pain, however further assessment of their effects on bone pain in genetically engineered animal models and cancer patients is warranted.
Collapse
Affiliation(s)
- Feier Zeng
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Abbie Wade
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Kade Harbert
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Shrina Patel
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Joshua S Holley
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Cornelia K Dehghanpuor
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Thomas Hopwood
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | - Silvia Marino
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences (UAMS), BioMed II, 238-2, Little Rock, AR, USA
| | - Antonia Sophocleous
- Department of Life Sciences, School of Sciences, European University Cyprus, 6 Diogenes Street, 1516, Nicosia, Cyprus.
| | - Aymen I Idris
- Department of Oncology and Metabolism, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|
3
|
Bajaj S, Jain S, Vyas P, Bawa S, Vohora D. The role of endocannabinoid pathway in the neuropathology of Alzheimer's disease: Can the inhibitors of MAGL and FAAH prove to be potential therapeutic targets against the cognitive impairment associated with Alzheimer's disease? Brain Res Bull 2021; 174:305-322. [PMID: 34217798 DOI: 10.1016/j.brainresbull.2021.06.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/25/2021] [Accepted: 06/29/2021] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease is a neurodegenerative disease characterized by progressive decline of cognitive function in combination with neuronal death. Current approved treatment target single dysregulated pathway instead of multiple mechanism, resulting in lack of efficacy in slowing down disease progression. The proclivity of endocannabinoid system to exert neuroprotective action and mitigate symptoms of neurodegeneration condition has received substantial interest. Growing evidence suggest the endocannabinoids (eCB) system, viz. anadamide (AEA) and arachidonoyl glycerol (2-AG), as potential therapeutic targets with the ability to modify Alzheimer's pathology by targeting the inflammatory, neurodegenerative and cognitive aspects of the disease. In order to modulate endocannabinoid system, number of agents have been reported amongst which are inhibitors of the monoacylglycerol (MAGL) and fatty acid amide hydrolase (FAAH), the enzymes that hydrolyses 2-AG and AEA respectively. However, little is known regarding the exact mechanistic signalling and their effects on pathophysiology and cognitive decline associated with Alzheimer's disease. Both MAGL and FAAH inhibitors possess fascinating properties that may offer a multi-faceted approach for the treatment of Alzheimer's disease such as potential to protect neurons from deleterious effect of amyloid-β, reducing phosphorylation of tau, reducing amyloid-β induced oxidative stress, stimulating neurotrophin to support brain intrinsic repair mechanism etc. Based on empirical evidence, MAGL and FAAH inhibitors might have potential for therapeutic efficacy against cognitive impairment associated with Alzheimer's disease. The aim of this review is to summarize the experimental studies demonstrating the polyvalent properties of MAGL or FAAH inhibitor compounds for the treatment of Alzheimer's disease, and also effect of these on learning and types of memories, which together encourage to study these compounds over other therapeutics targets. Further research in this direction would enhance the molecular mechanisms and development of applicable interventions for the treatment of Alzheimer's disease, which nevertheless stay as the primary unmet need.
Collapse
Affiliation(s)
- Shivanshu Bajaj
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Shreshta Jain
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Preeti Vyas
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sandhya Bawa
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Divya Vohora
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| |
Collapse
|
4
|
Dalton GD, Carney ST, Marshburn JD, Norford DC, Howlett AC. CB 1 Cannabinoid Receptors Stimulate Gβγ-GRK2-Mediated FAK Phosphorylation at Tyrosine 925 to Regulate ERK Activation Involving Neuronal Focal Adhesions. Front Cell Neurosci 2020; 14:176. [PMID: 32655375 PMCID: PMC7324865 DOI: 10.3389/fncel.2020.00176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
CB1 cannabinoid receptors (CB1) are abundantly expressed in the nervous system where they regulate focal adhesion kinase (FAK) and the mitogen-activated protein kinases (MAPK) extracellular signal-regulated kinase 1 and 2 (ERK1/2). However, the role of CB1-stimulated FAK 925 tyrosine phosphorylation (Tyr-P) in regulating ERK1/2 activation remains undefined. Here, immunoblotting analyses using antibodies against FAK phospho-Tyr 925 and ERK2 phospho-Tyr 204 demonstrated CB1-stimulated FAK 925 Tyr-P and ERK2 204 Tyr-P (0–5 min) which was followed by a decline in Tyr-P (5–20 min). CB1 stimulated FAK-Grb2 association and Ras-mediated ERK2 activation. The FAK inhibitors Y11 and PF 573228 abolished FAK 925 Tyr-P and partially inhibited ERK2 204 Tyr-P. FAK 925 Tyr-P and ERK2 204 Tyr-P were adhesion-dependent, required an intact actin cytoskeleton, and were mediated by integrins, Flk-1 vascular endothelial growth factor receptors, and epidermal growth factor receptors. FAK 925 Tyr-P and ERK2 204 Tyr-P were blocked by the Gβγ inhibitor gallein, a GRK2 inhibitor, and GRK2 siRNA silencing, suggesting Gβγ and GRK2 participate in FAK-mediated ERK2 activation. Together, these studies indicate FAK 925 Tyr-P occurs concurrently with CB1-stimulated ERK2 activation and requires the actin cytoskeleton and Gi/oβγ-GRK2-mediated cross-talk between CB1, integrins, and receptor tyrosine kinases (RTKs).
Collapse
Affiliation(s)
- George D Dalton
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Skyla T Carney
- Department of Biological and Biomedical Sciences, Julius L. Chambers Biomedical and Biotechnology Research Institute, North Carolina Central University, Durham, NC, United States
| | - Jamie D Marshburn
- Department of Biological and Biomedical Sciences, Julius L. Chambers Biomedical and Biotechnology Research Institute, North Carolina Central University, Durham, NC, United States
| | - Derek C Norford
- Department of Biological and Biomedical Sciences, Julius L. Chambers Biomedical and Biotechnology Research Institute, North Carolina Central University, Durham, NC, United States
| | - Allyn C Howlett
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
5
|
Lamani M, Malamas MS, Farah SI, Shukla VG, Almeida MF, Weerts CM, Anderson J, Wood JT, Farizatto KLG, Bahr BA, Makriyannis A. Piperidine and piperazine inhibitors of fatty acid amide hydrolase targeting excitotoxic pathology. Bioorg Med Chem 2019; 27:115096. [PMID: 31629610 DOI: 10.1016/j.bmc.2019.115096] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/07/2019] [Accepted: 09/08/2019] [Indexed: 11/30/2022]
Abstract
FAAH inhibitors offer safety advantages by augmenting the anandamide levels "on demand" to promote neuroprotective mechanisms without the adverse psychotropic effects usually seen with direct and chronic activation of the CB1 receptor. FAAH is an enzyme implicated in the hydrolysis of the endocannabinoid N-arachidonoylethanolamine (AEA), which is a partial agonist of the CB1 receptor. Herein, we report the discovery of a new series of highly potent and selective carbamate FAAH inhibitors and their evaluation for neuroprotection. The new inhibitors showed potent nanomolar inhibitory activity against human recombinant and purified rat FAAH, were selective (>1000-fold) against serine hydrolases MGL and ABHD6 and lacked any affinity for the cannabinoid receptors CB1 and CB2. Evaluation of FAAH inhibitors 9 and 31 using the in vitro competitive activity-based protein profiling (ABPP) assay confirmed that both inhibitors were highly selective for FAAH in the brain, since none of the other FP-reactive serine hydrolases in this tissue were inhibited by these agents. Our design strategy followed a traditional SAR approach and was supported by molecular modeling studies based on known FAAH cocrystal structures. To rationally design new molecules that are irreversibly bound to FAAH, we have constructed "precovalent" FAAH-ligand complexes to identify good binding geometries of the ligands within the binding pocket of FAAH and then calculated covalent docking poses to select compounds for synthesis. FAAH inhibitors 9 and 31 were evaluated for neuroprotection in rat hippocampal slice cultures. In the brain tissue, both inhibitors displayed protection against synaptic deterioration produced by kainic acid-induced excitotoxicity. Thus, the resultant compounds produced through rational design are providing early leads for developing therapeutics against seizure-related damage associated with a variety of disorders.
Collapse
Affiliation(s)
- Manjunath Lamani
- Center for Drug Discovery and Department of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA 02155, USA
| | - Michael S Malamas
- Center for Drug Discovery and Department of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA 02155, USA.
| | - Shrouq I Farah
- Center for Drug Discovery and Department of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA 02155, USA
| | - Vidyanand G Shukla
- Center for Drug Discovery and Department of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA 02155, USA
| | - Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Catherine M Weerts
- Center for Drug Discovery and Department of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA 02155, USA
| | - Joseph Anderson
- Center for Drug Discovery and Department of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA 02155, USA
| | - JodiAnne T Wood
- Center for Drug Discovery and Department of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA 02155, USA
| | - Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC 28372, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery and Department of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA 02155, USA
| |
Collapse
|
6
|
Farizatto KLG, Almeida MF, Long RT, Bahr BA. Early Synaptic Alterations and Selective Adhesion Signaling in Hippocampal Dendritic Zones Following Organophosphate Exposure. Sci Rep 2019; 9:6532. [PMID: 31024077 PMCID: PMC6484076 DOI: 10.1038/s41598-019-42934-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/11/2019] [Indexed: 11/09/2022] Open
Abstract
Organophosphates account for many of the world's deadliest poisons. They inhibit acetylcholinesterase causing cholinergic crises that lead to seizures and death, while survivors commonly experience long-term neurological problems. Here, we treated brain explants with the organophosphate compound paraoxon and uncovered a unique mechanism of neurotoxicity. Paraoxon-exposed hippocampal slice cultures exhibited progressive declines in synaptophysin, synapsin II, and PSD-95, whereas reduction in GluR1 was slower and NeuN and Nissl staining showed no indications of neuronal damage. The distinctive synaptotoxicity was observed in dendritic zones of CA1 and dentate gyrus. Interestingly, declines in synapsin II dendritic labeling correlated with increased staining for β1 integrin, a component of adhesion receptors that regulate synapse maintenance and plasticity. The paraoxon-induced β1 integrin response was targeted to synapses, and the two-fold increase in β1 integrin was selective as other synaptic adhesion molecules were unchanged. Additionally, β1 integrin-cofilin signaling was triggered by the exposure and correlations were found between the extent of synaptic decline and the level of β1 integrin responses. These findings identified organophosphate-mediated early and lasting synaptotoxicity which can explain delayed neurological dysfunction later in life. They also suggest that the interplay between synaptotoxic events and compensatory adhesion responses influences neuronal fate in exposed individuals.
Collapse
Affiliation(s)
- Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Ronald T Long
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA. .,Department of Biology, University of North Carolina-Pembroke, Pembroke, North Carolina, USA. .,Department of Chemistry and Physics, University of North Carolina-Pembroke, Pembroke, North Carolina, USA.
| |
Collapse
|
7
|
Hill JD, Zuluaga-Ramirez V, Gajghate S, Winfield M, Persidsky Y. Activation of GPR55 increases neural stem cell proliferation and promotes early adult hippocampal neurogenesis. Br J Pharmacol 2018; 175:3407-3421. [PMID: 29888782 DOI: 10.1111/bph.14387] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 04/30/2018] [Accepted: 05/28/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND AND PURPOSE The cannabinoid system exerts functional regulation of neural stem cell (NSC) proliferation and adult neurogenesis, yet not all effects of cannabinoid-like compounds seen can be attributed to the cannabinoid 1 (CB1 ) or CB2 receptor. The recently de-orphaned GPR55 has been shown to be activated by numerous cannabinoid ligands suggesting that GPR55 is a third cannabinoid receptor. Here, we examined the role of GPR55 activation in NSC proliferation and early adult neurogenesis. EXPERIMENTAL APPROACH The effects of GPR55 agonists (LPI, O-1602, ML184) on human (h) NSC proliferation in vitro were assessed by flow cytometry. Human NSC differentiation was determined by flow cytometry, qPCR and immunohistochemistry. Immature neuron formation in the hippocampus of C57BL/6 and GPR55-/- mice was evaluated by immunohistochemistry. KEY RESULTS Activation of GPR55 significantly increased proliferation rates of hNSCs in vitro. These effects were attenuated by ML193, a selective GPR55 antagonist. ML184 significantly promoted neuronal differentiation in vitro while ML193 reduced differentiation rates as compared to vehicle treatment. Continuous administration of O-1602 into the hippocampus via a cannula connected to an osmotic pump resulted in increased Ki67+ cells within the dentate gyrus. O-1602 increased immature neuron generation, as assessed by DCX+ and BrdU+ cells, as compared to vehicle-treated animals. GPR55-/- animals displayed reduced rates of proliferation and neurogenesis within the hippocampus while O-1602 had no effect as compared to vehicle controls. CONCLUSIONS AND IMPLICATIONS Together, these findings suggest GPR55 activation as a novel target and strategy to regulate NSC proliferation and adult neurogenesis.
Collapse
Affiliation(s)
- Jeremy D Hill
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Viviana Zuluaga-Ramirez
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Sachin Gajghate
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Malika Winfield
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Yuri Persidsky
- Department of Pathology and Laboratory Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.,Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
8
|
Farizatto KLG, Bahr BA. Paraoxon: An Anticholinesterase That Triggers an Excitotoxic Cascade of Oxidative Stress, Adhesion Responses, and Synaptic Compromise. ACTA ACUST UNITED AC 2017; 13:29-37. [PMID: 29805717 DOI: 10.19044/esj.2017.c1p4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The anticholinesterase paraoxon (Pxn) is an organophosphate (OP) and the active metabolite of the insecticide parathion. It potently inhibits the enzyme acetylcholinesterase and leads to enhanced glutamate release, diminished GABA uptake, oxidative damage, and neurodegeneration. The resulting increased levels of acetylcholine can trigger seizures and cause neuronal and excitotoxic damage in the brain. The brain susceptibility related to anticholinesterase toxins extends beyond potential brain damage and death from toxic levels of the agent. Asymptomatic low-level exposure to such toxins can also leave the brain vulnerable or even cause it to exhibit neurological problems later in life. The actions of Pxn and similar neurotoxins have been studied in order to examine the events associated with anticholinesterase toxicity in the brain. A recent study demonstrated that Pxn exposure initiates a pathogenic cascade involving seizure events and subsequent signs of damage including unique presynaptic vulnerability and associated behavioral deficits. In addition, Pxn-mediated synaptotoxicity is also associated with enhanced production of oxidative stress as well as integrin adhesion responses. These findings provide a better understanding of the molecular events involved in Pxn toxicity.
Collapse
Affiliation(s)
- Karen L G Farizatto
- Biotechnology Research and Training Center, William C. Friday Laboratory, University of North Carolina - Pembroke, Pembroke, North Carolina, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, William C. Friday Laboratory, University of North Carolina - Pembroke, Pembroke, North Carolina, USA
| |
Collapse
|
9
|
Farizatto KLG, McEwan SA, Naidoo V, Nikas SP, Shukla VG, Almeida MF, Byrd A, Romine H, Karanian DA, Makriyannis A, Bahr BA. Inhibitor of Endocannabinoid Deactivation Protects Against In Vitro and In Vivo Neurotoxic Effects of Paraoxon. J Mol Neurosci 2017; 63:115-122. [PMID: 28803438 DOI: 10.1007/s12031-017-0963-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 08/02/2017] [Indexed: 12/16/2022]
Abstract
The anticholinesterase paraoxon (Pxn) is related to military nerve agents that increase acetylcholine levels, trigger seizures, and cause excitotoxic damage in the brain. In rat hippocampal slice cultures, high-dose Pxn was applied resulting in a presynaptic vulnerability evidenced by a 64% reduction in synapsin IIb (syn IIb) levels, whereas the postsynaptic protein GluR1 was unchanged. Other signs of Pxn-induced cytotoxicity include the oxidative stress-related production of stable 4-hydroxynonenal (4-HNE)-protein adducts. Next, the Pxn toxicity was tested for protective effects by the fatty acid amide hydrolase (FAAH) inhibitor AM5206, a compound linked to enhanced repair signaling through the endocannabinoid pathway. The Pxn-mediated declines in syn IIb and synaptophysin were prevented by AM5206 in the slice cultures. To test if the protective results in the slice model translate to an in vivo model, AM5206 was injected i.p. into rats, followed immediately by subcutaneous Pxn administration. The toxin caused a pathogenic cascade initiated by seizure events, leading to presynaptic marker decline and oxidative changes in the hippocampus and frontal cortex. AM5206 exhibited protective effects including the reduction of seizure severity by 86%, and improving balance and coordination measured 24 h post-insult. As observed in hippocampal slices, the FAAH inhibitor also prevented the Pxn-induced loss of syn IIb in vivo. In addition, the AM5206 compound reduced the 4-HNE modifications of proteins and the β1 integrin activation events both in vitro and in vivo. These results indicate that Pxn exposure produces oxidative and synaptic toxicity that leads to the behavioral deficits manifested by the neurotoxin. In contrast, the presence of FAAH inhibitor AM5206 offsets the pathogenic cascade elicited by the Pxn anticholinesterase.
Collapse
Affiliation(s)
- Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, 28372-1510, USA
| | - Sara A McEwan
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, 28372-1510, USA.,Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
| | - Vinogran Naidoo
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, 28372-1510, USA.,Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Spyros P Nikas
- Center for Drug Discovery, Northeastern University, Boston, MA, USA
| | | | - Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, 28372-1510, USA
| | - Aaron Byrd
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, 28372-1510, USA
| | - Heather Romine
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, 28372-1510, USA
| | - David A Karanian
- Neurosciences Program, University of Connecticut, Storrs, CT, USA
| | | | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, 28372-1510, USA. .,Neurosciences Program, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
10
|
Abstract
The CB1 and CB2 cannabinoid receptors (CB1R, CB2R) are members of the G protein-coupled receptor (GPCR) family that were identified over 20 years ago. CB1Rs and CB2Rs mediate the effects of Δ9-tetrahydrocannabinol (Δ9-THC), the principal psychoactive constituent of marijuana, and subsequently identified endogenous cannabinoids (endocannabinoids) anandamide and 2-arachidonoyl glycerol. CB1Rs and CB2Rs have both similarities and differences in their pharmacology. Both receptors recognize multiple classes of agonist and antagonist compounds and produce an array of distinct downstream effects. Natural polymorphisms and alternative splice variants may also contribute to their pharmacological diversity. As our knowledge of the distinct differences grows, we may be able to target select receptor conformations and their corresponding pharmacological responses. This chapter will discuss their pharmacological characterization, distribution, phylogeny, and signaling pathways. In addition, the effects of extended agonist exposure and how that affects signaling and expression patterns of the receptors are considered.
Collapse
MESH Headings
- Alternative Splicing/genetics
- Animals
- Humans
- Phylogeny
- Polymorphism, Genetic
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/agonists
- Receptor, Cannabinoid, CB2/genetics
- Receptor, Cannabinoid, CB2/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Allyn C Howlett
- Center for Research on Substance Use and Addiction, Wake Forest University Health Sciences, Winston-Salem, NC, United States
| | - Mary E Abood
- Center for Substance Abuse Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.
| |
Collapse
|
11
|
Bennett MR, Arnold J, Hatton SN, Lagopoulos J. Regulation of fear extinction by long-term depression: The roles of endocannabinoids and brain derived neurotrophic factor. Behav Brain Res 2016; 319:148-164. [PMID: 27867101 DOI: 10.1016/j.bbr.2016.11.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 11/19/2022]
Abstract
The extinction of a conditioned fear response is of great interest in the search for a means of ameliorating adverse neurobiological changes resulting from stress. The discovery that endocannibinoid (EC) levels are inversely related to the extent of such stress, and that the amygdala is a primary site mediating stress, suggests that ECs in this brain region might play a major role in extinction. Supporting this are the observations that the basolateral complex of the amygdala shows an increase in ECs only during extinction and that early clinical trials indicate that cannabinoid-like agents, when taken orally by patients suffering from post traumatic stress disorder (PTSD), reduce insomnia and nightmares. In order to optimize the potential of these agents to ameliorate symptoms of PTSD four important questions need to be answered: first, what is the identity of the cells that release ECs in the amygdala during extinction; second, what are their sites of action; third, what roles do the ECs play in the alleviation of long- depression (LTD), a process central to extinction; and finally, to what extent does brain derived neurotrophic factor (BDNF) facilitate the release of ECs? A review of the relevant literature is presented in an attempt to answer these questions. It is suggested that the principal cell involved in EC synthesis and release during extinction is the so-called excitatory extinction neuron in the basal nucleus of the amygdala. Furthermore that the main site of action of the ECs is the adjacent calcitonin gene-related peptide inhibitory interneurons, whose normal role of blocking the excitatory neurons is greatly diminished. The molecular pathways leading (during extinction trials) to the synthesis and release of ECs from synaptic spines of extinction neurons, that is potentiated by BDNF, are also delineated in this review. Finally, consideration is given to how the autocrine action of BDNF, linked to the release of ECs, can lead to the sustained release of these, so maintaining extinction over long times.
Collapse
Affiliation(s)
- Maxwell R Bennett
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.
| | - Jonathon Arnold
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Sean N Hatton
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Jim Lagopoulos
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia; The Sunshine Coast Mind and Neuroscience, Thompson Institute, The University of the Sunshine Coast, QLD, Australia
| |
Collapse
|
12
|
He X, Zhu Y, Wang M, Jing G, Zhu R, Wang S. Antidepressant effects of curcumin and HU-211 coencapsulated solid lipid nanoparticles against corticosterone-induced cellular and animal models of major depression. Int J Nanomedicine 2016; 11:4975-4990. [PMID: 27757031 PMCID: PMC5055126 DOI: 10.2147/ijn.s109088] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Major depression is a complex neuropsychiatric disorder with few treatment approaches. The use of nontargeted antidepressants induced many side effects with their low efficacy. A more precise targeting strategy is to develop nanotechnology-based drug delivery systems; hence, we employed solid lipid nanoparticles (SLNs) to encapsulate HU-211 and curcumin (Cur). The antidepressant effects of the dual-drug nanoparticles (Cur/SLNs-HU-211) for major depression treatment were investigated in corticosterone-induced cellular and animal models of major depression. Cur/SLNs-HU-211 can effectively protect PC12 cells from corticosterone-induced apoptosis and can release more dopamine, which may be associated with the higher uptake of Cur/SLNs-HU-211 shown by cellular uptake behavior analysis. Additionally, Cur/SLNs-HU-211 significantly reduced the immobility time in forced swim test, enhanced fall latency in rotarod test, and improved the level of dopamine in mice blood. Cur/SLNs-HU-211 can deliver more Cur to the brain and thus produce a significant increase in neurotransmitters level in brain tissue, especially in the hippocampus and striatum. The results of Western blot and immunofluorescence revealed that Cur/SLNs-HU-211 can significantly enhance the expression of CB1, p-MEK1, and p-ERK1/2. Our study suggests that Cur/SLNs-HU-211 may have great potential for major depression treatment.
Collapse
Affiliation(s)
- Xiaolie He
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Yanjing Zhu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Mei Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Guoxin Jing
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Rongrong Zhu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Shilong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
13
|
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
|
14
|
Ernst J, Grabiec U, Greither T, Fischer B, Dehghani F. The endocannabinoid system in the human granulosa cell line KGN. Mol Cell Endocrinol 2016; 423:67-76. [PMID: 26773729 DOI: 10.1016/j.mce.2016.01.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 12/01/2015] [Accepted: 01/06/2016] [Indexed: 10/25/2022]
Abstract
Ovarian steroidogenesis is embedded in a sensitive network of regulatory mechanisms crucial for human fertility. The endocannabinoid system (ECS) represents an intrinsic modulating system involved in the regulation of endocrine functions. In the present study we characterized the ECS in the human granulosa cell line KGN and its impact on gonadotropin sensitivity and steroid hormone synthesis under basal and FSH-stimulated conditions. Expression studies were performed and estradiol was measured. CB1, CB2, DAGL, FAAH, GPR55, MAGL, NAPE-PLD and TRPV1 were expressed without FSH-dependent effects. Treatment with selective cannabinoid receptor agonists reduced basal but not FSH-stimulated estradiol and CYP19. Progesterone was not altered by ECS manipulation. CB1 agonist changed the expression of miRNAs associated with granulosa cell function, e.g. miR-23a, miR-24, miR-181a and miR-320a. Present data indicate a modulating role of the intrinsic ovarian ECS in the regulation of estradiol synthesis.
Collapse
Affiliation(s)
- Jana Ernst
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, D-06097 Halle (Saale), Germany.
| | - Urszula Grabiec
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, D-06097 Halle (Saale), Germany
| | - Thomas Greither
- Center for Reproductive Medicine and Andrology, University Hospital Halle (Saale), Martin Luther University Halle-Wittenberg, Ernst-Grube-Strasse 40, D-06120 Halle (Saale), Germany
| | - Bernd Fischer
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, D-06097 Halle (Saale), Germany
| | - Faramarz Dehghani
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Grosse Steinstrasse 52, D-06097 Halle (Saale), Germany
| |
Collapse
|
15
|
Endogenous and Synthetic Cannabinoids as Therapeutics in Retinal Disease. Neural Plast 2016; 2016:8373020. [PMID: 26881135 PMCID: PMC4736800 DOI: 10.1155/2016/8373020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 11/16/2015] [Accepted: 12/01/2015] [Indexed: 11/25/2022] Open
Abstract
The functional significance of cannabinoids in ocular physiology and disease has been reported some decades ago. In the early 1970s, subjects who smoked Cannabis sativa developed lower intraocular pressure (IOP). This led to the isolation of phytocannabinoids from this plant and the study of their therapeutic effects in glaucoma. The main treatment of this disease to date involves the administration of drugs mediating either the decrease of aqueous humour synthesis or the increase of its outflow and thus reduces IOP. However, the reduction of IOP is not sufficient to prevent visual field loss. Retinal diseases, such as glaucoma and diabetic retinopathy, have been defined as neurodegenerative diseases and characterized by ischemia-induced excitotoxicity and loss of retinal neurons. Therefore, new therapeutic strategies must be applied in order to target retinal cell death, reduction of visual acuity, and blindness. The aim of the present review is to address the neuroprotective and therapeutic potential of cannabinoids in retinal disease.
Collapse
|
16
|
Keegan BMT, Beveridge TJR, Pezor JJ, Xiao R, Sexton T, Childers SR, Howlett AC. Chronic baclofen desensitizes GABA(B)-mediated G-protein activation and stimulates phosphorylation of kinases in mesocorticolimbic rat brain. Neuropharmacology 2015; 95:492-502. [PMID: 25724082 PMCID: PMC4537290 DOI: 10.1016/j.neuropharm.2015.02.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 12/30/2014] [Accepted: 02/11/2015] [Indexed: 01/19/2023]
Abstract
The GABAB receptor is a therapeutic target for CNS and neuropathic disorders; however, few preclinical studies have explored effects of chronic stimulation. This study evaluated acute and chronic baclofen treatments on GABAB-activated G-proteins and signaling protein phosphorylation as indicators of GABAB signaling capacity. Brain sections from rats acutely administered baclofen (5 mg/kg, i.p.) showed no significant differences from controls in GABAB-stimulated GTPγS binding in any brain region, but displayed significantly greater phosphorylation/activation of focal adhesion kinase (pFAK(Tyr397)) in mesocorticolimbic regions (caudate putamen, cortex, hippocampus, thalamus) and elevated phosphorylated/activated glycogen synthase kinase 3-β (pGSK3β(Tyr216)) in the prefrontal cortex, cerebral cortex, caudate putamen, nucleus accumbens, thalamus, septum, and globus pallidus. In rats administered chronic baclofen (5 mg/kg, t.i.d. for five days), GABAB-stimulated GTPγS binding was significantly diminished in the prefrontal cortex, septum, amygdala, and parabrachial nucleus compared to controls. This effect was specific to GABAB receptors: there was no effect of chronic baclofen treatment on adenosine A1-stimulated GTPγS binding in any region. Chronically-treated rats also exhibited increases in pFAK(Tyr397) and pGSK3β(Tyr216) compared to controls, and displayed wide-spread elevations in phosphorylated dopamine- and cAMP-regulated phosphoprotein-32 (pDARPP-32(Thr34)) compared to acutely-treated or control rats. We postulate that those neuroadaptive effects of GABAB stimulation mediated by G-proteins and their sequelae correlate with tolerance to several of baclofen's effects, whereas sustained signaling via kinase cascades points to cross-talk between GABAB receptors and alternative mechanisms that are resistant to desensitization. Both desensitized and sustained signaling pathways should be considered in the development of pharmacotherapies targeting the GABA system.
Collapse
Affiliation(s)
- Bradley M T Keegan
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Thomas J R Beveridge
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Jeffrey J Pezor
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Chemistry, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Ruoyu Xiao
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Tammy Sexton
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Steven R Childers
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Allyn C Howlett
- Center for the Neurobiology of Addiction Treatment, Winston-Salem, NC 27157, USA; Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
17
|
Kokona D, Thermos K. Synthetic and endogenous cannabinoids protect retinal neurons from AMPA excitotoxicity in vivo, via activation of CB1 receptors: Involvement of PI3K/Akt and MEK/ERK signaling pathways. Exp Eye Res 2015; 136:45-58. [DOI: 10.1016/j.exer.2015.05.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/14/2015] [Accepted: 05/15/2015] [Indexed: 11/29/2022]
|
18
|
Varney S, Polston KF, Jessen T, Carneiro AMD. Mice lacking integrin β3 expression exhibit altered response to chronic stress. Neurobiol Stress 2015; 2:51-58. [PMID: 26634222 PMCID: PMC4664197 DOI: 10.1016/j.ynstr.2015.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Recent studies indicate multiple roles for integrin αvβ3 in adult neurons, including response to pharmacological agents such as cocaine and selective serotonin reuptake inhibitors. In this study, we examined the role of the integrin β3 gene (Itgb3) in the response to environmental stimuli by subjecting Itgb3+/+ and Itgb3-/- mice to unpredictable chronic mild stressors. We found that genetic abrogation of integrin β3 expression elicits an exaggerated vulnerability to chronic unpredictable stress in the open field test. In this test, chronic stress elicited significant decreases in stereotypic behavior and horizontal locomotor activity, including increases in anxiety behaviors. Mild chronic stress led to reductions in dopamine turnover in midbrains of Itgb3+/+, but not Itgb3-/- mice, suggesting a disruption of stress-dependent regulation of DA homeostasis. Chronic stress elicited altered synaptic expression of syntaxin and synaptophysin in midbrains of Itgb3-/- mice, when compared to Itgb3+/+. Semi-quantitative Western blot studies revealed that the synaptic expression, but not total tissue expression, of multiple signaling proteins is correlated with integrin αv levels in the midbrain. Moreover, loss of integrin β3 expression modifies this correlation network. Together, these findings demonstrate that Itgb3-/- mice display a pattern of changes indicating disrupted regulation of midbrain synaptic systems involved in conferring resilience to mild stressors.
Collapse
Affiliation(s)
- Seth Varney
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| | - Keith F Polston
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| | - Tammy Jessen
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| | - Ana M D Carneiro
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| |
Collapse
|
19
|
Makriyannis A. 2012 Division of medicinal chemistry award address. Trekking the cannabinoid road: a personal perspective. J Med Chem 2014; 57:3891-911. [PMID: 24707904 DOI: 10.1021/jm500220s] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
My involvement with the field of cannabinoids spans close to 3 decades and covers a major part of my scientific career. It also reflects the robust progress in this initially largely unexplored area of biology. During this period of time, I have witnessed the growth of modern cannabinoid biology, starting from the discovery of its two receptors and followed by the characterization of its endogenous ligands and the identification of the enzyme systems involved in their biosynthesis and biotransformation. I was fortunate enough to start at the beginning of this new era and participate in a number of the new discoveries. It has been a very exciting journey. With coverage of some key aspects of my work during this period of "modern cannabinoid research," this Award Address, in part historical, intends to give an account of how the field grew, the key discoveries, and the most promising directions for the future.
Collapse
Affiliation(s)
- Alexandros Makriyannis
- Center for Drug Discovery and Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University , 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
20
|
Dalton GD, Peterson LJ, Howlett AC. CB₁ cannabinoid receptors promote maximal FAK catalytic activity by stimulating cooperative signaling between receptor tyrosine kinases and integrins in neuronal cells. Cell Signal 2013; 25:1665-77. [PMID: 23571270 PMCID: PMC4165595 DOI: 10.1016/j.cellsig.2013.03.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 03/16/2013] [Accepted: 03/26/2013] [Indexed: 01/28/2023]
Abstract
Tyrosine phosphorylation (Tyr-P) of focal adhesion kinase (FAK) regulates FAK activation. Phosphorylated FAK Tyr 397 binds Src family kinases (Src), which in turn directly phosphorylate FAK Tyr 576/577 to produce maximal FAK enzymatic activity. CB₁ cannabinoid receptors (CB₁) are abundantly expressed in the nervous system and influence FAK activation by presently unknown mechanisms. The current investigation determined that CB₁-stimulated maximal FAK catalytic activity is mediated by Gi/o proteins in N18TG2 neuronal cells, and that G12/13 regulation of Rac1 and RhoA occurs concomitantly. Immunoblotting analyses using antibodies against FAK phospho-Tyr 397 and phospho-Tyr 576/577 demonstrated that the time-course of CB₁-stimulated FAK 576/577 Tyr-P occurred in three phases: Phase I (0-2 min) maximal Tyr-P, Phase II (5-20 min) rapid decline in Tyr-P, and Phase III (>20 min) plateau in Tyr-P at submaximal levels. In contrast, FAK 397 Tyr-P was monophasic and significantly lower in magnitude. FAK 397 Tyr-P and Phase I FAK 576/577 Tyr-P involved protein tyrosine phosphatase (PTP1B and Shp1/Shp2)-mediated Src activation, Protein Kinase A (PKA) inhibition, and integrin activation. Phase I maximal FAK 576/577 Tyr-P also required cooperative signaling between receptor tyrosine kinases (RTKs) and integrins. The integrin antagonist RGDS peptide, Flk-1 vascular endothelial growth factor receptor (VEGFR) antagonist SU5416, and epidermal growth factor receptor (EGFR) antagonist AG 1478 blocked Phase I FAK 576/577 Tyr-P. CB₁ agonists failed to stimulate FAK Tyr-P in the absence of integrin activation upon suspension in serum-free culture media. In contrast, cells grown on the integrin ligands fibronectin and laminin displayed increased FAK 576/577 Tyr-P that was augmented by CB₁ agonists and blocked by the Src inhibitor PP2 and Flk-1 VEGFR antagonist SU5416. Taken together, these studies have identified a complex integrative pathway utilized by CB₁ to stimulate maximal FAK 576/577 Tyr-P in neuronal cells.
Collapse
MESH Headings
- Animals
- Benzoxazines/pharmacology
- Cell Line, Tumor
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/metabolism
- Fibronectins/pharmacology
- Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors
- Focal Adhesion Protein-Tyrosine Kinases/metabolism
- Integrins/antagonists & inhibitors
- Integrins/genetics
- Integrins/metabolism
- Kinetics
- Laminin/pharmacology
- Mice
- Morpholines/pharmacology
- Naphthalenes/pharmacology
- Neurons/cytology
- Neurons/metabolism
- Oligopeptides/pharmacology
- Pertussis Toxin/pharmacology
- Phosphorylation/drug effects
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Receptor, Cannabinoid, CB1/agonists
- Receptor, Cannabinoid, CB1/metabolism
- Signal Transduction/drug effects
- Time Factors
- Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
- Vascular Endothelial Growth Factor Receptor-2/metabolism
- src-Family Kinases/antagonists & inhibitors
- src-Family Kinases/metabolism
Collapse
Affiliation(s)
- George D Dalton
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.
| | | | | |
Collapse
|
21
|
Endogenous cannabinoids revisited: A biochemistry perspective. Prostaglandins Other Lipid Mediat 2013; 102-103:13-30. [DOI: 10.1016/j.prostaglandins.2013.02.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 02/20/2013] [Accepted: 02/21/2013] [Indexed: 12/13/2022]
|
22
|
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
|
23
|
Idris AI, Ralston SH. Role of cannabinoids in the regulation of bone remodeling. Front Endocrinol (Lausanne) 2012; 3:136. [PMID: 23181053 PMCID: PMC3499879 DOI: 10.3389/fendo.2012.00136] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/25/2012] [Indexed: 12/22/2022] Open
Abstract
The endocannabinoid system plays a key role in regulating a variety of physiological processes such as appetite control and energy balance, pain perception, and immune responses. Recent studies have implicated the endocannabinoid system in the regulation of bone cell activity and bone remodeling. These studies showed that endogenous cannabinoid ligands, cannabinoid receptors, and the enzymes responsible for ligand synthesis and breakdown all play important roles in bone mass and in the regulation of bone disease. These findings suggest that the endocannabinoid pathway could be of value as a therapeutic target for the prevention and treatment of bone diseases. Here, we review the role of the skeletal endocannabinoid system in the regulation of bone remodeling in health and disease.
Collapse
Affiliation(s)
- Aymen I. Idris
- Bone and Cancer Group, Edinburgh Cancer Research Centre, The University of EdinburghEdinburgh, UK
| | - Stuart H. Ralston
- Rheumatic Disease Unit, The Centre for Molecular Medicine, The University of EdinburghEdinburgh, UK
| |
Collapse
|
24
|
Nicoll G, Davidson S, Shanley L, Hing B, Lear M, McGuffin P, Ross R, MacKenzie A. Allele-specific differences in activity of a novel cannabinoid receptor 1 (CNR1) gene intronic enhancer in hypothalamus, dorsal root ganglia, and hippocampus. J Biol Chem 2012; 287:12828-34. [PMID: 22362764 PMCID: PMC3339935 DOI: 10.1074/jbc.m111.336750] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/09/2012] [Indexed: 12/30/2022] Open
Abstract
Polymorphisms within intron 2 of the CNR1 gene, which encodes cannabinoid receptor 1 (CB(1)), have been associated with addiction, obesity, and brain volume deficits. We used comparative genomics to identify a polymorphic (rs9444584-C/T) sequence (ECR1) in intron 2 of the CNR1 gene that had been conserved for 310 million years. The C-allele of ECR1 (ECR1(C)) acted as an enhancer in hypothalamic and dorsal root ganglia cells and responded to MAPK activation through the MEKK pathway but not in hippocampal cells. However, ECR1(T) was significantly more active in hypothalamic and dorsal root ganglia cells but, significantly, and in contrast to ECR1(C), was highly active in hippocampal cells where it also responded strongly to activation of MAPK. Intriguingly, rs9444584 is in strong linkage disequilibrium with two other SNPs (rs9450898 (r(2) = 0.841) and rs2023239 (r(2) = 0.920)) that have been associated with addiction, obesity (rs2023239), and reduced fronto-temporal white matter volumes in schizophrenia patients as a result of cannabis misuse (rs9450898). Considering their high linkage disequilibrium and the increased response of ECR1(T) to MAPK signaling when compared with ECR1(C), it is possible that the functional effects of the different alleles of rs9444584 may play a role in the conditions associated with rs9450898 and rs2023239. Further analysis of the different alleles of ECR1 may lead to a greater understanding of the role of CNR1 gene misregulation in these conditions as well as chronic inflammatory pain.
Collapse
Affiliation(s)
- Gemma Nicoll
- From the School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland and
| | - Scott Davidson
- From the School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland and
| | - Lynne Shanley
- From the School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland and
| | - Ben Hing
- From the School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland and
| | - Marissa Lear
- From the School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland and
| | - Peter McGuffin
- the Medical Research Council Social, Genetic and Developmental Psychiatry Centre Centre, Institute of Psychiatry, King's College London, DeCrespigny Park, London SE5 8AF3, United Kingdom
| | - Ruth Ross
- From the School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland and
| | - Alasdair MacKenzie
- From the School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, Scotland and
| |
Collapse
|
25
|
Wisniewski ML, Hwang J, Bahr BA. Submicromolar Aβ42 reduces hippocampal glutamate receptors and presynaptic markers in an aggregation-dependent manner. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1664-74. [PMID: 21978994 DOI: 10.1016/j.bbadis.2011.09.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 08/31/2011] [Accepted: 09/19/2011] [Indexed: 01/01/2023]
Abstract
Synaptic pathology in Alzheimer's disease brains is thought to involve soluble Aβ42 peptide. Here, sterile incubation in PBS caused small Aβ42 oligomer formation as well as heterogeneous, 6E10-immunopositive aggregates of 80-100kDa. The high molecular weight aggregates (H-agg) formed in a time-dependent manner over an extended 30-day period. Interestingly, an inverse relationship between dimeric and H-agg formation was more evident when incubations were performed at 37°C as compared to 23°C, thus providing an experimental strategy with which to address synaptic compromise produced by the different Aβ aggregates. H-agg species formed faster and to higher levels at 37°C compared to 23°C, and the two aggregate preparations were evaluated in hippocampal slice cultures, a sensitive system for monitoring synaptic integrity. Applied daily at 80-600nM for 7days, the Aβ42 preparations caused dose-dependent and aggregation-dependent declines in α-amino-3-hydroxy-5-methyl-4-isoxazolepropionate (AMPA) and N-methyl-d-aspartate (NMDA) receptor subunits as well as in presynaptic components. Unlike the synaptic effects, Aβ42 induced only trace cellular degeneration that was CA1 specific. The 37°C preparation was less effective at decreasing synaptic markers, corresponding with its reduced levels of Aβ42 monomers and dimers. Aβ42 dimers decayed significantly faster at 37°C than 23°C, and more rapidly than monomers at either temperature. These findings indicate that Aβ42 can self-aggregate into potent synaptotoxic oligomers as well as into larger aggregates that may serve to neutralize the toxic formations. These results will add to the growing debate concerning whether high molecular weight Aβ complexes that form amyloid plaques are protective through the sequestration of oligomeric species.
Collapse
|
26
|
Du J, Wang Q, Hu B, Peng Z, Zhao Y, Ma L, Xiong L, Lu Y, Zhu X, Chen S. Involvement of ERK 1/2 activation in electroacupuncture pretreatment via cannabinoid CB1 receptor in rats. Brain Res 2010; 1360:1-7. [PMID: 20654595 DOI: 10.1016/j.brainres.2010.07.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 06/25/2010] [Accepted: 07/13/2010] [Indexed: 11/30/2022]
Abstract
Our previous study demonstrated that pretreatment with electroacupuncture (EA) elicited protective effects against transient cerebral ischemia through cannabinoid receptor type 1 receptor (CB1R). In the present study, we investigated whether or not the extracellular signal regulated-kinase 1/2 (ERK1/2) pathway was involved in the ischemic tolerance induced by EA pretreatment through CB1R. At 24h after the end of the last EA pretreatment, focal cerebral ischemia was induced by middle cerebral artery occlusion for 120min in rats. The neurological scores and infarct volumes were evaluated at 24h after reperfusion. The expression of p-ERK1/2 in the brains was also investigated in the presence or absence of CB1R antagonist AM251. EA pretreatment reduced infarct volumes and improved neurological outcome at 24h after reperfusion, and the beneficial effects were abolished by U0126. The blockade of CB1R by AM251 reversed the up-regulation of p-ERK1/2 expression induced by EA pretreatment. Our findings suggest that the ERK1/2 pathway might be involved in EA pretreatment-induced cerebral ischemic tolerance via cannabinoid CB1 receptor in rats.
Collapse
Affiliation(s)
- Juan Du
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Naidoo V, Nikas SP, Karanian DA, Hwang J, Zhao J, Wood JT, Alapafuja SO, Vadivel SK, Butler D, Makriyannis A, Bahr BA. A new generation fatty acid amide hydrolase inhibitor protects against kainate-induced excitotoxicity. J Mol Neurosci 2010; 43:493-502. [PMID: 21069475 DOI: 10.1007/s12031-010-9472-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 10/28/2010] [Indexed: 02/02/2023]
Abstract
Endocannabinoids, including anandamide (AEA), have been implicated in neuroprotective on-demand responses. Related to such a response to injury, an excitotoxic kainic acid (KA) injection (i.p.) was found to increase AEA levels in the brain. To modulate the endocannabinoid response during events of excitotoxicity in vitro and in vivo, we utilized a new generation compound (AM5206) that selectively inhibits the AEA deactivating enzyme fatty acid amide hydrolase (FAAH). KA caused calpain-mediated spectrin breakdown, declines in synaptic markers, and disruption of neuronal integrity in cultured hippocampal slices. FAAH inhibition with AM5206 protected against the neurodegenerative cascade assessed in the slice model 24 h postinsult. In vivo, KA administration induced seizures and the same neurodegenerative events exhibited in vitro. When AM5206 was injected immediately after KA in rats, the seizure scores were markedly reduced as were levels of cytoskeletal damage and synaptic protein decline. The pre- and postsynaptic proteins were protected by the FAAH inhibitor to levels comparable to those found in healthy control brains. These data support the idea that endocannabinoids are released and converge on pro-survival pathways that prevent excitotoxic progression.
Collapse
Affiliation(s)
- Vinogran Naidoo
- Biotechnology Research and Training Center, University of North Carolina Pembroke, 115 Livermore Drive, Pembroke, NC 28372-1510, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Noonan J, Tanveer R, Klompas A, Gowran A, McKiernan J, Campbell VA. Endocannabinoids prevent β-amyloid-mediated lysosomal destabilization in cultured neurons. J Biol Chem 2010; 285:38543-54. [PMID: 20923768 DOI: 10.1074/jbc.m110.162040] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuronal cell loss underlies the pathological decline in cognition and memory associated with Alzheimer disease (AD). Recently, targeting the endocannabinoid system in AD has emerged as a promising new approach to treatment. Studies have identified neuroprotective roles for endocannabinoids against key pathological events in the AD brain, including cell death by apoptosis. Elucidation of the apoptotic pathway evoked by β-amyloid (Aβ) is thus important for the development of therapeutic strategies that can thwart Aβ toxicity and preserve cell viability. We have previously reported that lysosomal membrane permeabilization plays a distinct role in the apoptotic pathway initiated by Aβ. In the present study, we provide evidence that the endocannabinoid system can stabilize lysosomes against Aβ-induced permeabilization and in turn sustain cell survival. We report that endocannabinoids stabilize lysosomes by preventing the Aβ-induced up-regulation of the tumor suppressor protein, p53, and its interaction with the lysosomal membrane. We also provide evidence that intracellular cannabinoid type 1 receptors play a role in stabilizing lysosomes against Aβ toxicity and thus highlight the functionality of these receptors. Given the deleterious effect of lysosomal membrane permeabilization on cell viability, stabilization of lysosomes with endocannabinoids may represent a novel mechanism by which these lipid modulators confer neuroprotection.
Collapse
Affiliation(s)
- Janis Noonan
- Department of Physiology, School of Medicine and Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
The endocannabinoid system is recognized to play an important role in regulating a variety of physiological processes, including appetite control and energy balance, pain perception, and immune responses. The endocannabinoid system has also recently been implicated in the regulation of bone metabolism. Endogenously produced cannabinoids are hydrophobic molecules derived from hydrolysis of membrane phospholipids. These substances, along with plant-derived and synthetic cannabinoids, interact with the type 1 (CB(1)) and 2 (CB(2)) cannabinoid receptors and the GPR55 receptor to regulate cellular function through a variety of signaling pathways. Endocannabinoids are produced in bone, but the mechanisms that regulate their production are unclear. Skeletal phenotyping of mice with targeted inactivation of cannabinoid receptors and pharmacological studies have shown that cannabinoids play a key role in the regulation of bone metabolism. Mice with CB(1) deficiency have high peak bone mass as a result of an osteoclast defect but develop age-related osteoporosis as a result of impaired bone formation and accumulation of bone marrow fat. Mice with CB(2) deficiency have relatively normal peak bone mass but develop age-related osteoporosis as a result of increased bone turnover with uncoupling of bone resorption from bone formation. Mice with GPR55 deficiency have increased bone mass as a result of a defect in the resorptive activity of osteoclasts, but bone formation is unaffected. Cannabinoids are also produced within synovial tissues, and preclinical studies have shown that cannabinoid receptor ligands are effective in the treatment of inflammatory arthritis. These data indicate that cannabinoid receptors and the enzymes responsible for ligand synthesis and breakdown play important roles in bone remodeling and in the pathogenesis of joint disease.
Collapse
Affiliation(s)
- Aymen I Idris
- Molecular Medicine Centre, Western General Hospital, University of Edinburgh, Scotland, UK
| | | |
Collapse
|
30
|
Hwang J, Adamson C, Butler D, Janero DR, Makriyannis A, Bahr BA. Enhancement of endocannabinoid signaling by fatty acid amide hydrolase inhibition: a neuroprotective therapeutic modality. Life Sci 2009; 86:615-23. [PMID: 19527737 DOI: 10.1016/j.lfs.2009.06.003] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 05/28/2009] [Accepted: 06/03/2009] [Indexed: 12/31/2022]
Abstract
AIMS This review posits that fatty acid amide hydrolase (FAAH) inhibition has therapeutic potential against neuropathological states including traumatic brain injury; Alzheimer's, Huntington's, and Parkinson's diseases; and stroke. MAIN METHODS This proposition is supported by data from numerous in vitro and in vivo experiments establishing metabolic and pharmacological contexts for the neuroprotective role of the endogenous cannabinoid ("endocannabinoid") system and selective FAAH inhibitors. KEY FINDINGS The systems biology of endocannabinoid signaling involves two main cannabinoid receptors, the principal endocannabinoid lipid mediators N-arachidonoylethanolamine ("anandamide") (AEA) and 2-arachidonoyl glycerol (2-AG), related metabolites, and the proteins involved in endocannabinoid biosynthesis, biotransformation, and transit. The endocannabinoid system is capable of activating distinct signaling pathways on-demand in response to pathogenic events or stimuli, thereby enhancing cell survival and promoting tissue repair. Accumulating data suggest that endocannabinoid system modulation at discrete targets is a promising pharmacotherapeutic strategy for treating various medical conditions. In particular, neuronal injury activates cannabinoid signaling in the central nervous system as an intrinsic neuroprotective response. Indirect potentiation of this salutary response through pharmacological inhibition of FAAH, an endocannabinoid-deactivating enzyme, and consequent activation of signaling pathways downstream from cannabinoid receptors have been shown to promote neuronal maintenance and function. SIGNIFICANCE This therapeutic modality has the potential to offer site- and event-specific neuroprotection under conditions where endocannabinoids are being produced as part of a physiological protective mechanism. In contrast, direct application of cannabinoid receptor agonists to the central nervous system may activate CB receptors indiscriminately and invite unwanted psychotrophic effects.
Collapse
Affiliation(s)
- Jeannie Hwang
- Department of Pharmaceutical Sciences and the Neurosciences Program, University of Connecticut, Storrs, Connecticut, USA
| | | | | | | | | | | |
Collapse
|
31
|
Kellogg R, Mackie K, Straiker A. Cannabinoid CB1 receptor-dependent long-term depression in autaptic excitatory neurons. J Neurophysiol 2009; 102:1160-71. [PMID: 19494194 DOI: 10.1152/jn.00266.2009] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Long-term depression (LTD) of synaptic signaling-lasting from tens of minutes to hours or longer-is a widespread form of synaptic plasticity in the brain. Neurons express diverse forms of LTD, including autaptic LTD (autLTD) observed in cultured hippocampal neurons, the mechanism of which remains unknown. We have recently reported that autaptic neurons express both endocannabinoid-mediated depolarization-induced suppression of excitation (DSE) and metabotropic suppression of excitation (MSE). We now report that activating cannabinoid CB(1) receptors is necessary for the induction of autLTD. Most surprisingly, CB(1) does not induce autLTD via the G(i/o) proteins typically activated by this receptor nor with G(s). Rather, the requirements of presynaptic phospholipase C and filled calcium stores suggest G(q). In autLTD, a 3- to 4-min activation of the receptor by the endocannabinoid 2-arachidonoyl glycerol leads to prolonged inhibition while leaving short-term inhibition (e.g., DSE) intact. autLTD requires activation of both metabo- and ionotropic glutamate receptors. autLTD also requires MEK/ERK activation. Under certain conditions, one or more DSE stimuli will elicit autLTD. It is becoming evident that cannabinoids mediate multiple forms of plasticity at a single synapse, stretching temporally from tens of seconds (DSE/MSE) to tens of minutes (autLTD) to hours (CB(1) desensitization). Our findings imply a remarkable flexibility for the cannabinoid signaling system whereby discrete mechanisms of CB(1) activation within a single neuron yield temporally and mechanistically distinct forms of plasticity.
Collapse
Affiliation(s)
- Ryan Kellogg
- Department of Anesthesiology, University of Washington, Seattle, Washington, USA
| | | | | |
Collapse
|
32
|
Chaves GP, Nogueira TCA, Britto LRG, Bordin S, Torrão AS. Retinal removal up-regulates cannabinoid CB(1) receptors in the chick optic tectum. J Neurosci Res 2008; 86:1626-34. [PMID: 18189324 DOI: 10.1002/jnr.21613] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The endocannabinoid system has been implicated in several neurobiological processes, including neurodegeneration and neuroprotection. The aim of this study was to evaluate the effects of unilateral retinal ablation on the expression of the cannabinoid receptor subtype 1 (CB(1)) at both protein and mRNA levels in the optic tectum of the adult chick brain. After different survival times postlesion (2-30 days), the chick brains were subjected to immunohistochemical, immunoblotting, and real-time PCR procedures to evaluate CB(1) expression. TUNEL and Fluoro-Jade B were used to verify the possible occurrence of cell death, and immunostaining for the microtubule-associated protein MAP-2 was performed to verify possible dendritic remodeling after lesions. No cell death could be observed in the deafferented tectum, at least up to 30 days postlesion, although Fluoro-Jade B could reveal degenerating axons and terminals. Retinal ablation seems to generate an increase of CB(1) protein in the optic tectum and other retinorecipient visual areas, which paralleled an increase in MAP-2 staining. On the other hand, CB(1) mRNA levels were not changed after retinal ablation. Our results reveal that CB(1) expression in visual structures of the adult chick brain may be negatively regulated by the retinal innervation. The increase of CB(1) receptor expression observed after retinal removal indicates that these receptors are not presynaptic in retinal axons projecting to the tectum and suggests a role of the cannabinoid system in plasticity processes ensuing after lesions.
Collapse
Affiliation(s)
- Gabriela P Chaves
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
33
|
McLaughlin RJ, Hill MN, Morrish AC, Gorzalka BB. Local enhancement of cannabinoid CB1 receptor signalling in the dorsal hippocampus elicits an antidepressant-like effect. Behav Pharmacol 2007; 18:431-8. [PMID: 17762511 DOI: 10.1097/fbp.0b013e3282ee7b44] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Systemic administration of direct cannabinoid CB1 receptor agonists and inhibitors of the hydrolytic enzyme fatty acid amide hydrolase have been shown to elicit antidepressant effects. Moreover, the endocannabinoid system in the hippocampus is sensitive to both chronic stress and antidepressant administration, suggesting a potential role of this system in emotional changes associated with these regimens. The aim of this study was to determine if cannabinoid CB1 receptors in the hippocampus modulate emotionality in rats as assessed via the forced swim test. Male Sprague-Dawley rats were bilaterally implanted with cannulae directed at the dentate gyrus of the dorsal hippocampus and subsequently received three infusions of either the cannabinoid CB1 receptor agonist HU-210 (1 and 2.5 microg), the fatty acid amide hydrolase inhibitor URB597 (0.5 and 1 microg), the cannabinoid CB1 receptor antagonist AM251 (1 and 2.5 microg), or vehicle (dimethyl sulfoxide) and were assessed in the forced swim test. Infusion of both doses of HU-210 resulted in a dramatic reduction in immobility and increase in swimming behaviour, indicative of an antidepressant response, which was partially reversed by coadministration of AM251. No effect of URB597 administration or any effect following the administration of AM251 alone was, however, observed. These data indicate that activation of CB1 receptors in the dentate gyrus of the hippocampus results in an antidepressant-like response. Collectively, these data highlight the potential importance of changes in the hippocampal endocannabinoid system following stress or antidepressant treatment with respect to the manifestation and/or treatment of depression.
Collapse
Affiliation(s)
- Ryan J McLaughlin
- Department of Psychology, University of British Columbia, Vancouver, Canada
| | | | | | | |
Collapse
|
34
|
Ryzhikov S, Bahr BA. Gephyrin alterations due to protein accumulation stress are reduced by the lysosomal modulator Z-Phe-Ala-diazomethylketone. J Mol Neurosci 2007; 34:131-9. [PMID: 18204977 DOI: 10.1007/s12031-007-9009-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2007] [Accepted: 08/22/2007] [Indexed: 11/25/2022]
Abstract
Inhibitory neurotransmission is important for brain function and requires specific transmitter receptors that are organized in synaptic domains. Gephyrin is a cytoskeletal organization protein that binds tubulin and plays an important role in clustering and organizing select inhibitory neurotransmitter receptors. Here, we tested if gephyrin is altered by protein accumulation stress that is common in age-related neurodegenerative disorders. For this, we used the hippocampal slice model that has been shown to exhibit chloroquine (CQN)-induced protein accumulation, microtubule destabilization, transport failure, and declines in excitatory neurotransmitter receptors and their responses. In addition to the decreases in excitatory receptor subunits and other glutamatergic markers, we found that gephyrin isoforms were reduced across the CQN treatment period. Associated with this decline in gephyrin levels was the production of three gephyrin breakdown products (GBDPs) of 30, 38, and 48 kDa. The induced effects on gephyrin were tested for evidence of recovery through enhancement of lysosomal function that is known to promote protein clearance and microtubule integrity. Using the lysosomal modulator Z-Phe-Ala-diazomethylketone (PADK), gephyrin levels were completely restored in correspondence with the recovery of excitatory glutamatergic components. In addition, GBDPs were significantly reduced after the 2-day PADK treatment, to levels that were at or below those measured in control cultures. These findings suggest that receptor-clustering mechanisms for inhibitory synapses are compromised during protein accumulation events. They also indicate that a lysosomal enhancement strategy can protect gephyrin integrity, which may be vital for the balance between inhibitory and excitatory signaling during age-related diseases.
Collapse
Affiliation(s)
- Sophia Ryzhikov
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA
| | | |
Collapse
|
35
|
Karanian DA, Karim SL, Wood JT, Williams JS, Lin S, Makriyannis A, Bahr BA. Endocannabinoid enhancement protects against kainic acid-induced seizures and associated brain damage. J Pharmacol Exp Ther 2007; 322:1059-66. [PMID: 17545313 DOI: 10.1124/jpet.107.120147] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Endocannabinoids are released in response to pathogenic insults, and inhibitors of endocannabinoid inactivation enhance such on-demand responses that promote cellular protection. Here, AM374 (palmitylsulfonyl fluoride), an irreversible inhibitor of fatty acid amide hydrolase (FAAH), was injected i.p. into rats to test for endocannabinoid enhancement. AM374 caused a prolonged elevation of anandamide levels in several brain regions, including the hippocampus, and resulted in rapid activation of the extracellular signal regulated-kinase/mitogen-activated protein kinase pathway that has been linked to survival. To evaluate the neuroprotective nature of the FAAH inhibitor, we tested AM374 in a seizure model involving rats insulted with kainic acid (KA). AM374 was injected immediately after KA administration, and seizure scores were significantly reduced throughout a 4-h observation period. The KA-induced seizures were associated with calpain-mediated cytoskeletal breakdown, reductions in synaptic markers, and loss of CA1 hippocampal neurons. FAAH inhibition protected against the excitotoxic damage and neuronal loss assessed 48 h postinsult. AM374 also preserved pre- and postsynaptic markers to levels comparable with those found in noninsulted animals, and the synaptic marker preservation strongly correlated with reduced seizure scores. With regard to behavioral deficits in the excitotoxic rats, AM374 produced nearly complete functional protection, significantly improving balance and coordination across different behavioral paradigms. These data indicate that AM374 crosses the blood-brain barrier, enhances endocannabinoid responses in key neuronal circuitries, and protects the brain against excitotoxic damage.
Collapse
Affiliation(s)
- David A Karanian
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA.
| | | | | | | | | | | | | |
Collapse
|
36
|
Chicoine LM, Bahr BA. Excitotoxic protection by polyanionic polysaccharide: evidence of a cell survival pathway involving AMPA receptor-MAPK Interactions. J Neurosci Res 2007; 85:294-302. [PMID: 17131415 DOI: 10.1002/jnr.21117] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Growing numbers of studies indicate that polysaccharides influence signaling events important for brain function. It has been speculated that such polysaccharide modulation of neuronal signals can promote synaptogenesis and cell maintenance. Here, we tested whether dextran sulfate, a polyanion that mimics natural mucopolysaccharides, protects hippocampal neurons against excitotoxic insults. An excitotoxin was applied to primary hippocampal cultures in the absence or presence of a large 500-kDa dextran sulfate (DS-L), a smaller 5-8-kDa species (DS-S), or sulfate-free dextran of 500 kDa. Only DS-L prevented neuronal damage as determined by a membrane permeability assay and phase contrast morphology. The sulfate and size dependence is also characteristic of DS-L's modulatory action on the channel activity of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA)-type glutamate receptors. The extent of neuroprotection correlates with the level of modulation of AMPA responses, and DS-L exhibits comparable EC(50) values for the two effects (3-7 nM). DS-L also modulates the link between AMPA receptors and mitogen-activated protein kinase (MAPK) involving extracellular signal-regulated protein kinase (ERK), well known for its involvement in cell survival and repair. Correspondingly, protection against N-methyl-D-aspartate (NMDA) excitotoxicity was evident in hippocampal slice cultures when DS-L was applied 30 min postinsult. These findings suggest that polysaccharides elicit neuroprotection in the brain, including enhanced repair responses through the AMPA receptor-MAPK axis.
Collapse
|
37
|
Watson PMD, Humphries MJ, Relton J, Rothwell NJ, Verkhratsky A, Gibson RM. Integrin-binding RGD peptides induce rapid intracellular calcium increases and MAPK signaling in cortical neurons. Mol Cell Neurosci 2007; 34:147-54. [PMID: 17150373 DOI: 10.1016/j.mcn.2006.10.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 10/12/2006] [Accepted: 10/17/2006] [Indexed: 10/23/2022] Open
Abstract
Integrins mediate cell adhesion to the extracellular matrix and initiate intracellular signaling. They play key roles in the central nervous system (CNS), participating in synaptogenesis, synaptic transmission and memory formation, but their precise mechanism of action remains unknown. Here we show that the integrin ligand-mimetic peptide GRGDSP induced NMDA receptor-dependent increases in intracellular calcium levels within seconds of presentation to primary cortical neurons. These were followed by transient activation and nuclear translocation of the ERK1/2 mitogen-activated protein kinase. RGD-induced effects were reduced by the NMDA receptor antagonist MK801, and ERK1/2 signaling was specifically inhibited by ifenprodil and PP2, indicating a functional connection between integrins, Src and NR2B-containing NMDA receptors. GRGDSP peptides were not significantly neuroprotective against excitotoxic insults. These results demonstrate a previously undescribed, extremely rapid effect of RGD peptide binding to integrins on cortical neurons that implies a close, functionally relevant connection between adhesion receptors and synaptic transmission.
Collapse
Affiliation(s)
- P Marc D Watson
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Manchester, M13 9PT, UK
| | | | | | | | | | | |
Collapse
|
38
|
Graham ES, Ball N, Scotter EL, Narayan P, Dragunow M, Glass M. Induction of Krox-24 by endogenous cannabinoid type 1 receptors in Neuro2A cells is mediated by the MEK-ERK MAPK pathway and is suppressed by the phosphatidylinositol 3-kinase pathway. J Biol Chem 2006; 281:29085-95. [PMID: 16864584 DOI: 10.1074/jbc.m602516200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Neuro2a cells endogenously express cannabinoid type 1 (CB1) receptors. CB1 stimulation with HU210 activated ERK and induced the transcription factor Krox-24. A functional MEK-ERK pathway is an important requirement for CB1-mediated Krox-24 induction as blockade of MEK signaling by UO126 reduces both basal and CB1-mediated activation of Krox-24. CB1 receptor stimulation did not activate either JNK or p38 MAPK pathways or the pro-proliferation phosphatidylinositol 3-kinase (PI3K)-Akt pathway. However, serum removal or blockade of PI3K signaling by LY294002 transiently stimulated basal Krox-24 expression and increased CB1-mediated induction of Krox-24. This was consistent with a transient increase in pMEK, pERK, and pCREB levels following PI3K blockade. These data demonstrate that CB1-mediated activation of the Krox-24 transcription factor is negatively regulated through the PI3K-Akt pathway and reveals several points of signaling cross-talk between these two important kinase pathways.
Collapse
Affiliation(s)
- E Scott Graham
- Department of Pharmacology, University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | | | | | |
Collapse
|
39
|
Bahr BA, Karanian DA, Makanji SS, Makriyannis A. Targeting the endocannabinoid system in treating brain disorders. Expert Opin Investig Drugs 2006; 15:351-65. [PMID: 16548785 DOI: 10.1517/13543784.15.4.351] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Recent cannabinoid research has a primary focus on developing therapeutics against human diseases. Many studies on cannabinoids indicate important progress for protection against several neurodegenerative disorders. Agonists of cannabinoid receptors activate signalling pathways in the brain that are linked to neuronal repair and cell maintenance, and endogenous ligands can also activate neuroprotective responses. These endocannabinoids are bioactive fatty acid amides and esters that are synthesised in the brain and include arachidonoyl ethanolamide (anandamide) and 2-arachidonoyl glycerol. Endocannabinoids are released in response to pathogenic events, thus representing a potential compensatory repair mechanism. Enhancing this on-demand action of endocannabinoids is a strategy with which to promote endogenous repair signalling. For such enhancement, considerable work has gone into modulating the availability of endocannabinoids by blocking the processes of their deactivation. The targets include the anandamide-hydrolysing enzyme fatty acid amide hydrolase, the carrier-mediated anandamide transport system and 2-arachidonoyl glycerol-deactivating enzyme monoacylglycerol lipase. The activity of endocannabinoids is terminated through transport and degradation and, accordingly, selective inhibitors of these processes effectively exploit the protective nature of cannabinergic responses. This review highlights recent studies implicating the endocannabinoid system in neuroprotection against different disorders of the CNS.
Collapse
Affiliation(s)
- Ben A Bahr
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269, USA.
| | | | | | | |
Collapse
|
40
|
Karanian DA, Brown QB, Makriyannis A, Kosten TA, Bahr BA. Dual modulation of endocannabinoid transport and fatty acid amide hydrolase protects against excitotoxicity. J Neurosci 2006; 25:7813-20. [PMID: 16120783 PMCID: PMC6725251 DOI: 10.1523/jneurosci.2347-05.2005] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The endocannabinoid system has been suggested to elicit signals that defend against several disease states including excitotoxic brain damage. Besides direct activation with CB1 receptor agonists, cannabinergic signaling can be modulated through inhibition of endocannabinoid transport and fatty acid amide hydrolase (FAAH), two mechanisms of endocannabinoid inactivation. To test whether the transporter and FAAH can be targeted pharmacologically to modulate survival/repair responses, the transport inhibitor N-(4-hydroxyphenyl)-arachidonamide (AM404) and the FAAH inhibitor palmitylsulfonyl fluoride (AM374) were assessed for protection against excitotoxicity in vitro and in vivo. AM374 and AM404 both enhanced mitogen-activated protein kinase (MAPK) activation in cultured hippocampal slices. Interestingly, combining the distinct inhibitors produced additive effects on CB1 signaling and associated neuroprotection. After an excitotoxic insult in the slices, infusing the AM374/AM404 combination protected against cytoskeletal damage and synaptic decline, and the protection was similar to that produced by the stable CB1 agonist AM356 (R-methanandamide). AM374/AM404 and the agonist also elicited cytoskeletal and synaptic protection in vivo when coinjected with excitotoxin into the dorsal hippocampus. Correspondingly, potentiating endocannabinoid responses with the AM374/AM404 combination prevented behavioral alterations and memory impairment that are characteristic of excitotoxic damage. The protective effects mediated by AM374/AM404 were (1) evident 7 d after insult, (2) correlated with the preservation of CB1-linked MAPK signaling, and (3) were blocked by a selective CB1 antagonist. These results indicate that dual modulation of the endocannabinoid system with AM374/AM404 elicits neuroprotection through the CB1 receptor. The transporter and FAAH are modulatory sites that may be exploited to enhance cannabinergic signaling for therapeutic purposes.
Collapse
Affiliation(s)
- David A Karanian
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, Connecticut 06269, USA
| | | | | | | | | |
Collapse
|