1
|
Wang Q, Guo C, Wang T, Shuai P, Wu W, Huang S, Li Y, Zhao P, Zeng C, Yi L. Drug protection against radiation-induced neurological injury: mechanisms and developments. Arch Toxicol 2024:10.1007/s00204-024-03933-w. [PMID: 39724149 DOI: 10.1007/s00204-024-03933-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
In daily life, individuals are frequently exposed to various forms of radiation, which, when adhering to safety standards, typically result in relatively minor health effects. However, accidental exposure to radiation levels that exceed these safety standards can lead to significant health consequences. This study focuses on the analysis of radiation-induced damage to the nervous system and the mechanisms of pharmacological protection. The findings indicate that radiation can adversely affect neural structures, memory, and neurobehaviour. A range of pharmacological agents, including traditional Chinese medicine, Western medicine, and other therapeutic drugs, can be employed to safeguard the nervous system from radiation damage. The primary protective mechanisms of these agents encompass antioxidant effects, attenuation of apoptosis, and reduction of neurogenesis. A comprehensive review of these topics will offer new insights for the development and investigation of drugs aimed at mitigating radiation-induced damage to the nervous system.
Collapse
Affiliation(s)
- Qingyu Wang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Caimao Guo
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Tiantian Wang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Peimeng Shuai
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Wenyu Wu
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shuqi Huang
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Yuanyuan Li
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pei Zhao
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Chengkai Zeng
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Lan Yi
- Institute of Pharmacy and Pharmacology, Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
2
|
He XF, Yang XF, Li G, Zhao Y, Luo J, Xu JH, Zheng HQ, Zhang LY, Hu XQ. Physical Exercise Improves the Neuronal Function in Ischemic Stroke Via Microglial CB 2R/P2Y12 Signaling. Mol Neurobiol 2024:10.1007/s12035-024-04391-2. [PMID: 39066973 DOI: 10.1007/s12035-024-04391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024]
Abstract
Physical exercise (PE) may be the single most important and accessible lifestyle habit throughout life, it inhibits the neuroinflammatory response and protects the brain against damage. As the innate cells in brain, microglia undergo morphological and functional changes to communicate with neurons protecting the neurons from injury. Herein, aiming at exploring the effects of PE on the communication between microglia-neuron during acute ischemic cerebral infarction, we carried out running wheel training before the conduction of transient middle cerebral artery occlusion (tMCAO) in C57BL/6 J and Cx3cr1-GFP mice. We found that microglial P2Y12 expression in the peri-infarct area was decreased, microglial dynamics and microglia-neuron communications were impaired, using in vivo two-photon imaging. PE up-regulated the microglial P2Y12 expression, increased the microglial dynamics, and promoted the contacts of microglia with neurons. As a result, PE inhibited neuronal Ca2+ overloads and protected against damage of the neuronal mitochondria in acute tMCAO. Mechanistically, PE increased the cannabinoid receptor 2 (CB2R) in microglia, promoted the phosphorylation of Nrf2 (NF-E2-related factor 2) at ser-344, increased the transcription factor level of Mafk, and up-regulated the level of P2Y12, whereby PE increased the levels of CB2R to promote microglia-neuron contacts to monitor and protect neuronal function.
Collapse
Affiliation(s)
- Xiao-Fei He
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Xiao-Feng Yang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, Guangzhou, 510663, Guangdong, China
| | - Yun Zhao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Jing Luo
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Jing-Hui Xu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Hai-Qing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Li-Ying Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| | - Xi-Quan Hu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
3
|
Reid HMO, Trepanier O, Gross A, Poberezhnyk P, Snowden T, Conway K, Breit KR, Rodriguez C, Thomas JD, Christie BR. Prenatal ethanol and cannabis exposure have sex- and region-specific effects on somatostatin and neuropeptide Y interneurons in the rat hippocampus. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1289-1301. [PMID: 38789401 PMCID: PMC11236510 DOI: 10.1111/acer.15350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND Cannabis is increasingly being legalized and socially accepted around the world and is often used with alcohol in social settings. We recently showed that in utero exposure to both substances can alter the density of parvalbumin-expressing interneurons in the hippocampus. Here we investigate the effects of in utero alcohol and cannabis exposure, alone or in combination, on somatostatin- and neuropeptide Y-positive (NPY) interneurons. These are separate classes of interneurons important for network synchrony and inhibition in the hippocampus. METHODS A 2 (Ethanol, Air) × 2 (tetrahydrocannabinol [THC], Vehicle) design was used to expose pregnant Sprague-Dawley rats to either ethanol or air, in addition to either THC or the inhalant vehicle solution, during gestational days 5-20. Immunohistochemistry for somatostatin- and NPY-positive interneurons was performed in 50 μm tissue sections obtained at postnatal day 70. RESULTS Exposure to THC in utero had region-specific and sex-specific effects on the density of somatostatin-positive interneurons in the adult rat hippocampus. A female-specific decrease in NPY interneuron cell density was observed in the CA1 region following THC exposure. Combined exposure to alcohol and THC reduced NPY neurons selectively in the ventral dentate gyrus hippocampal subfield. However, overall, co-exposure to alcohol and cannabis had neither additive nor synergistic effects on interneuron populations in other areas of the hippocampus. CONCLUSIONS These results illustrate how alcohol and cannabis exposure in utero may affect hippocampal function by altering inhibitory processes in a sex-specific manner.
Collapse
Affiliation(s)
- Hannah M O Reid
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
| | - Owen Trepanier
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
| | - Allyson Gross
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
| | - Polina Poberezhnyk
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
| | - Taylor Snowden
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
| | - Kate Conway
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
| | - Kristen R Breit
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
- Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
- Department of Psychology, West Chester University of Pennsylvania, West Chester, Pennsylvania, USA
| | - Cristina Rodriguez
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| | - Jennifer D Thomas
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Canada, Victoria, British Columbia, Canada
- Island Medical Program and Department of Cellular and Physiological Sciences, University of British Columbia, Victoria, British Columbia, Canada
- Center for Behavioral Teratology, San Diego State University, San Diego, California, USA
| |
Collapse
|
4
|
Rodrigues RS, Moreira JB, Mateus JM, Barateiro A, Paulo SL, Vaz SH, Lourenço DM, Ribeiro FF, Soares R, Loureiro-Campos E, Bielefeld P, Sebastião AM, Fernandes A, Pinto L, Fitzsimons CP, Xapelli S. Cannabinoid type 2 receptor inhibition enhances the antidepressant and proneurogenic effects of physical exercise after chronic stress. Transl Psychiatry 2024; 14:170. [PMID: 38555299 PMCID: PMC10981758 DOI: 10.1038/s41398-024-02877-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 04/02/2024] Open
Abstract
Chronic stress is a major risk factor for neuropsychiatric conditions such as depression. Adult hippocampal neurogenesis (AHN) has emerged as a promising target to counteract stress-related disorders given the ability of newborn neurons to facilitate endogenous plasticity. Recent data sheds light on the interaction between cannabinoids and neurotrophic factors underlying the regulation of AHN, with important effects on cognitive plasticity and emotional flexibility. Since physical exercise (PE) is known to enhance neurotrophic factor levels, we hypothesised that PE could engage with cannabinoids to influence AHN and that this would result in beneficial effects under stressful conditions. We therefore investigated the actions of modulating cannabinoid type 2 receptors (CB2R), which are devoid of psychotropic effects, in combination with PE in chronically stressed animals. We found that CB2R inhibition, but not CB2R activation, in combination with PE significantly ameliorated stress-evoked emotional changes and cognitive deficits. Importantly, this combined strategy critically shaped stress-induced changes in AHN dynamics, leading to a significant increase in the rates of cell proliferation and differentiation of newborn neurons, overall reduction in neuroinflammation, and increased hippocampal levels of BDNF. Together, these results show that CB2Rs are crucial regulators of the beneficial effects of PE in countering the effects of chronic stress. Our work emphasises the importance of understanding the mechanisms behind the actions of cannabinoids and PE and provides a framework for future therapeutic strategies to treat stress-related disorders that capitalise on lifestyle interventions complemented with endocannabinoid pharmacomodulation.
Collapse
Affiliation(s)
- R S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Université de Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - J B Moreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - J M Mateus
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - A Barateiro
- Central Nervous System, blood and peripheral inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - S L Paulo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - S H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - D M Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - F F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - R Soares
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - E Loureiro-Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - P Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - A M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - A Fernandes
- Central Nervous System, blood and peripheral inflammation, Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - L Pinto
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - C P Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - S Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
5
|
de Souza PB, de Araujo Borba L, Castro de Jesus L, Valverde AP, Gil-Mohapel J, Rodrigues ALS. Major Depressive Disorder and Gut Microbiota: Role of Physical Exercise. Int J Mol Sci 2023; 24:16870. [PMID: 38069198 PMCID: PMC10706777 DOI: 10.3390/ijms242316870] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/18/2023] Open
Abstract
Major depressive disorder (MDD) has a high prevalence and is a major contributor to the global burden of disease. This psychiatric disorder results from a complex interaction between environmental and genetic factors. In recent years, the role of the gut microbiota in brain health has received particular attention, and compelling evidence has shown that patients suffering from depression have gut dysbiosis. Several studies have reported that gut dysbiosis-induced inflammation may cause and/or contribute to the development of depression through dysregulation of the gut-brain axis. Indeed, as a consequence of gut dysbiosis, neuroinflammatory alterations caused by microglial activation together with impairments in neuroplasticity may contribute to the development of depressive symptoms. The modulation of the gut microbiota has been recognized as a potential therapeutic strategy for the management of MMD. In this regard, physical exercise has been shown to positively change microbiota composition and diversity, and this can underlie, at least in part, its antidepressant effects. Given this, the present review will explore the relationship between physical exercise, gut microbiota and depression, with an emphasis on the potential of physical exercise as a non-invasive strategy for modulating the gut microbiota and, through this, regulating the gut-brain axis and alleviating MDD-related symptoms.
Collapse
Affiliation(s)
- Pedro Borges de Souza
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Laura de Araujo Borba
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Louise Castro de Jesus
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Ana Paula Valverde
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| | - Joana Gil-Mohapel
- Island Medical Program, Faculty of Medicine, University of British Columbia, Victoria, BC V8P 5C2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ana Lúcia S. Rodrigues
- Center of Biological Sciences, Department of Biochemistry, Universidade Federal de Santa Catarina, Florianópolis 88037-000, SC, Brazil; (P.B.d.S.); (L.d.A.B.); (L.C.d.J.); (A.P.V.)
| |
Collapse
|
6
|
Costas‐Insua C, Guzmán M. Endocannabinoid signaling in glioma. Glia 2023; 71:127-138. [PMID: 35322459 PMCID: PMC9790654 DOI: 10.1002/glia.24173] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/16/2022] [Accepted: 03/17/2022] [Indexed: 12/30/2022]
Abstract
High-grade gliomas constitute the most frequent and aggressive form of primary brain cancer in adults. These tumors express cannabinoid CB1 and CB2 receptors, as well as other elements of the endocannabinoid system. Accruing preclinical evidence supports that pharmacological activation of cannabinoid receptors located on glioma cells exerts overt anti-tumoral effects by modulating key intracellular signaling pathways. The mechanism of this cannabinoid receptor-evoked anti-tumoral activity in experimental models of glioma is intricate and may involve an inhibition not only of cancer cell survival/proliferation, but also of invasiveness, angiogenesis, and the stem cell-like properties of cancer cells, thereby affecting the complex tumor microenvironment. However, the precise biological role of the endocannabinoid system in the generation and progression of glioma seems very context-dependent and remains largely unknown. Increasing our basic knowledge on how (endo)cannabinoids act on glioma cells could help to optimize experimental cannabinoid-based anti-tumoral therapies, as well as the preliminary clinical testing that is currently underway.
Collapse
Affiliation(s)
- Carlos Costas‐Insua
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Department of Biochemistry and Molecular BiologyInstituto Universitario de Investigación Neuroquímica (IUIN), Complutense UniversityMadridSpain,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
| | - Manuel Guzmán
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED)MadridSpain,Department of Biochemistry and Molecular BiologyInstituto Universitario de Investigación Neuroquímica (IUIN), Complutense UniversityMadridSpain,Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
| |
Collapse
|
7
|
Watts JJ, Guma E, Chavez S, Tyndale RF, Ross RA, Houle S, Wilson AA, Chakravarty M, Rusjan PM, Mizrahi R. In vivo brain endocannabinoid metabolism is related to hippocampus glutamate and structure - a multimodal imaging study with PET, 1H-MRS, and MRI. Neuropsychopharmacology 2022; 47:1984-1991. [PMID: 35906490 PMCID: PMC9485131 DOI: 10.1038/s41386-022-01384-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 06/17/2022] [Accepted: 07/07/2022] [Indexed: 01/18/2023]
Abstract
Dysregulation of hippocampus glutamatergic neurotransmission and reductions in hippocampal volume have been associated with psychiatric disorders. The endocannabinoid system modulates glutamate neurotransmission and brain development, including hippocampal remodeling. In humans, elevated levels of anandamide and lower activity of its catabolic enzyme fatty acid amide hydrolase (FAAH) are associated with schizophrenia diagnosis and psychotic symptom severity, respectively (Neuropsychopharmacol, 29(11), 2108-2114; Biol. Psychiatry 88 (9), 727-735). Although preclinical studies provide strong evidence linking anandamide and FAAH to hippocampus neurotransmission and structure, these relationships remain poorly understood in humans. We recruited young adults with and without psychotic disorders and measured FAAH activity, hippocampal glutamate and glutamine (Glx), and hippocampal volume using [11C]CURB positron emission tomography (PET), proton magnetic resonance spectroscopy (1H-MRS) and T1-weighted structural MRI, respectively. We hypothesized that higher FAAH activity would be associated with greater hippocampus Glx and lower hippocampus volume, and that these effects would differ in patients with psychotic disorders relative to healthy control participants. After attrition and quality control, a total of 37 participants (62% male) completed [11C]CURB PET and 1H-MRS of the left hippocampus, and 45 (69% male) completed [11C]CURB PET and hippocampal volumetry. Higher FAAH activity was associated with greater concentration of hippocampal Glx (F1,36.36 = 9.17, p = 0.0045; Cohen's f = 0.30, medium effect size) and smaller hippocampal volume (F1,44.70 = 5.94, p = 0.019, Cohen's f = 0.26, medium effect size). These effects did not differ between psychosis and healthy control groups (no group interaction). This multimodal imaging study provides the first in vivo evidence linking hippocampal Glx and hippocampus volume with endocannabinoid metabolism in the human brain.
Collapse
Affiliation(s)
- Jeremy J Watts
- Research Centre, CHU Sainte-Justine, Montreal, QC, Canada
- Department of Psychiatry, Université de Montréal, Montreal, QC, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Elisa Guma
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, QC, Canada
- Developmental Neurogenomics Unit, Human Genetics Branch, National Institute of Mental Health, Bethesda, MD, USA
| | - Sofia Chavez
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Rachel F Tyndale
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Ruth A Ross
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Sylvain Houle
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Alan A Wilson
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Mallar Chakravarty
- Computational Brain Anatomy (CoBrA) Laboratory, Cerebral Imaging Centre, Douglas Mental Health University Institute, Montreal, QC, Canada
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Pablo M Rusjan
- Department of Psychiatry, McGill University, Montreal, QC, Canada
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Romina Mizrahi
- Department of Psychiatry, McGill University, Montreal, QC, Canada.
- Douglas Mental Health University Institute, Montreal, QC, Canada.
| |
Collapse
|
8
|
Dynamic Changes in the Endocannabinoid System during the Aging Process: Focus on the Middle-Age Crisis. Int J Mol Sci 2022; 23:ijms231810254. [PMID: 36142165 PMCID: PMC9499672 DOI: 10.3390/ijms231810254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Endocannabinoid (eCB) signaling is markedly decreased in the hippocampus (Hip) of aged mice, and the genetic deletion of the cannabinoid receptor type 1 (CB1) leads to an early onset of cognitive decline and age-related histological changes in the brain. Thus, it is hypothesized that cognitive aging is modulated by eCB signaling through CB1. In the present study, we detailed the changes in the eCB system during the aging process using different complementary techniques in mouse brains of five different age groups, ranging from adolescence to old age. Our findings indicate that the eCB system is most strongly affected in middle-aged mice (between 9 and 12 months of age) in a brain region-specific manner. We show that 2-arachidonoylglycerol (2-AG) was prominently decreased in the Hip and moderately in caudate putamen (CPu), whereas anandamide (AEA) was decreased in both CPu and medial prefrontal cortex along with cingulate cortex (mPFC+Cg), starting from 6 months until 12 months. Consistent with the changes in 2-AG, the 2-AG synthesizing enzyme diacylglycerol lipase α (DAGLα) was also prominently decreased across the sub-regions of the Hip. Interestingly, we found a transient increase in CB1 immunoreactivity across the sub-regions of the Hip at 9 months, a plausible compensation for reduced 2-AG, which ultimately decreased strongly at 12 months. Furthermore, quantitative autoradiography of CB1 revealed that [3H]CP55940 binding markedly increased in the Hip at 9 months. However, unlike the protein levels, CB1 binding density did not drop strongly at 12 months and at old age. Furthermore, [3H]CP55940 binding was significantly increased in the lateral entorhinal cortex (LEnt), starting from the middle age until the old age. Altogether, our findings clearly indicate a middle-age crisis in the eCB system, which could be a potential time window for therapeutic interventions to abrogate the course of cognitive aging.
Collapse
|
9
|
Razi O, Tartibian B, Laher I, Govindasamy K, Zamani N, Rocha-Rodrigues S, Suzuki K, Zouhal H. Multimodal Benefits of Exercise in Patients With Multiple Sclerosis and COVID-19. Front Physiol 2022; 13:783251. [PMID: 35492581 PMCID: PMC9048028 DOI: 10.3389/fphys.2022.783251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/31/2022] [Indexed: 11/24/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease characterized by plaque formation and neuroinflammation. The plaques can present in various locations, causing a variety of clinical symptoms in patients with MS. Coronavirus disease-2019 (COVID-19) is also associated with systemic inflammation and a cytokine storm which can cause plaque formation in several areas of the brain. These concurring events could exacerbate the disease burden of MS. We review the neuro-invasive properties of SARS-CoV-2 and the possible pathways for the entry of the virus into the central nervous system (CNS). Complications due to this viral infection are similar to those occurring in patients with MS. Conditions related to MS which make patients more susceptible to viral infection include inflammatory status, blood-brain barrier (BBB) permeability, function of CNS cells, and plaque formation. There are also psychoneurological and mood disorders associated with both MS and COVID-19 infections. Finally, we discuss the effects of exercise on peripheral and central inflammation, BBB integrity, glia and neural cells, and remyelination. We conclude that moderate exercise training prior or after infection with SARS-CoV-2 can produce health benefits in patients with MS patients, including reduced mortality and improved physical and mental health of patients with MS.
Collapse
Affiliation(s)
- Omid Razi
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Razi University, Kermanshah, Iran
| | - Bakhtyar Tartibian
- Department of Sports Injuries, Faculty of Physical Education and Sports Sciences, Allameh Tabataba’i University, Tehran, Iran
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Karuppasamy Govindasamy
- Department of Physical Education & Sports Science, SRM Institute of Science and Technology, Kattankulathur, India
| | - Nastaran Zamani
- Department of Biology, Faculty of Science, Payame-Noor University, Tehran, Iran
| | - Silvia Rocha-Rodrigues
- Escola Superior de Desporto e Lazer, Instituto Politécnico de Viana do Castelo, Viana do Castelo, Portugal
- Research Centre in Sports Sciences, Health Sciences and Human Development (CIDESD), Quinta de Prados, Edifício Ciências de Desporto, Vila Real, Portugal
- Tumor & Microenvironment Interactions Group, i3S, Porto, Portugal
| | | | - Hassane Zouhal
- Laboratoire Mouvement, Sport, Santé, University of Rennes, Rennes, France
- Institut International des Sciences du Sport (2I2S), Irodouer, France
| |
Collapse
|
10
|
The Endocannabinoid System and Physical Activity—A Robust Duo in the Novel Therapeutic Approach against Metabolic Disorders. Int J Mol Sci 2022; 23:ijms23063083. [PMID: 35328503 PMCID: PMC8948925 DOI: 10.3390/ijms23063083] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/09/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Rapidly increasing worldwide prevalence of obesity and related pathologies encompassing coronary heart disease, hypertension, metabolic syndrome, or type 2 diabetes constitute serious threats to global health and are associated with a significantly elevated risk of premature death. Considering the enormous burden of these pathologies, novel therapeutic and preventive patterns are indispensable. Dysregulation of one of the most complex biological systems in the human body namely, the endocannabinoid system (ECS) may result in metabolic imbalance and development of insulin resistance, type 2 diabetes, or non-alcoholic fatty liver disease. Furthermore, many studies showed that physical exercises, depending on their type, intensity, and frequency, exert various alterations within the ECS. Emerging evidence suggests that targeting the ECS via physical activity may produce robust beneficial effects on the course of metabolic pathologies. However, the data showing a direct correlation between the ECS and physical activity in the aspect of metabolic health are very scarce. Therefore, the aim of this review was to provide the most up-to-date state of knowledge about the interplay between the ECS activity and physical exercises in the novel therapeutic and preventive approach toward metabolic pathologies. We believe that this paper, at least in part, will fulfill the existing gap in knowledge and encourage researchers to further explore this very complex yet interesting link between the ECS, its action in physical activity, and subsequent positive outcomes for metabolic health.
Collapse
|
11
|
de Melo Reis RA, Isaac AR, Freitas HR, de Almeida MM, Schuck PF, Ferreira GC, Andrade-da-Costa BLDS, Trevenzoli IH. Quality of Life and a Surveillant Endocannabinoid System. Front Neurosci 2021; 15:747229. [PMID: 34776851 PMCID: PMC8581450 DOI: 10.3389/fnins.2021.747229] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 10/01/2021] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system (ECS) is an important brain modulatory network. ECS regulates brain homeostasis throughout development, from progenitor fate decision to neuro- and gliogenesis, synaptogenesis, brain plasticity and circuit repair, up to learning, memory, fear, protection, and death. It is a major player in the hypothalamic-peripheral system-adipose tissue in the regulation of food intake, energy storage, nutritional status, and adipose tissue mass, consequently affecting obesity. Loss of ECS control might affect mood disorders (anxiety, hyperactivity, psychosis, and depression), lead to drug abuse, and impact neurodegenerative (Alzheimer's, Parkinson, Huntington, Multiple, and Amyotrophic Lateral Sclerosis) and neurodevelopmental (autism spectrum) disorders. Practice of regular physical and/or mind-body mindfulness and meditative activities have been shown to modulate endocannabinoid (eCB) levels, in addition to other players as brain-derived neurotrophic factor (BDNF). ECS is involved in pain, inflammation, metabolic and cardiovascular dysfunctions, general immune responses (asthma, allergy, and arthritis) and tumor expansion, both/either in the brain and/or in the periphery. The reason for such a vast impact is the fact that arachidonic acid, a precursor of eCBs, is present in every membrane cell of the body and on demand eCBs synthesis is regulated by electrical activity and calcium shifts. Novel lipid (lipoxins and resolvins) or peptide (hemopressin) players of the ECS also operate as regulators of physiological allostasis. Indeed, the presence of cannabinoid receptors in intracellular organelles as mitochondria or lysosomes, or in nuclear targets as PPARγ might impact energy consumption, metabolism and cell death. To live a better life implies in a vigilant ECS, through healthy diet selection (based on a balanced omega-3 and -6 polyunsaturated fatty acids), weekly exercises and meditation therapy, all of which regulating eCBs levels, surrounded by a constructive social network. Cannabidiol, a diet supplement has been a major player with anti-inflammatory, anxiolytic, antidepressant, and antioxidant activities. Cognitive challenges and emotional intelligence might strengthen the ECS, which is built on a variety of synapses that modify human behavior. As therapeutically concerned, the ECS is essential for maintaining homeostasis and cannabinoids are promising tools to control innumerous targets.
Collapse
Affiliation(s)
- Ricardo Augusto de Melo Reis
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alinny Rosendo Isaac
- Laboratory of Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hércules Rezende Freitas
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana Macedo de Almeida
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Fernanda Schuck
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Costa Ferreira
- Laboratory of Neuroenergetics and Inborn Errors of Metabolism, Institute of Medical Biochemistry Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Isis Hara Trevenzoli
- Laboratory of Molecular Endocrinology, Institute of Biophysics Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
12
|
Marin Bosch B, Bringard A, Logrieco MG, Lauer E, Imobersteg N, Thomas A, Ferretti G, Schwartz S, Igloi K. A single session of moderate intensity exercise influences memory, endocannabinoids and brain derived neurotrophic factor levels in men. Sci Rep 2021; 11:14371. [PMID: 34257382 PMCID: PMC8277796 DOI: 10.1038/s41598-021-93813-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 06/24/2021] [Indexed: 11/15/2022] Open
Abstract
Regular physical exercise enhances memory functions, synaptic plasticity in the hippocampus, and brain derived neurotrophic factor (BDNF) levels. Likewise, short periods of exercise, or acute exercise, benefit hippocampal plasticity in rodents, via increased endocannabinoids (especially anandamide, AEA) and BDNF release. Yet, it remains unknown whether acute exercise has similar effects on BDNF and AEA levels in humans, with parallel influences on memory performance. Here we combined blood biomarkers, behavioral, and fMRI measurements to assess the impact of a single session of physical exercise on associative memory and underlying neurophysiological mechanisms in healthy male volunteers. For each participant, memory was tested after three conditions: rest, moderate or high intensity exercise. A long-term memory retest took place 3 months later. At both test and retest, memory performance after moderate intensity exercise was increased compared to rest. Memory after moderate intensity exercise correlated with exercise-induced increases in both AEA and BNDF levels: while AEA was associated with hippocampal activity during memory recall, BDNF enhanced hippocampal memory representations and long-term performance. These findings demonstrate that acute moderate intensity exercise benefits consolidation of hippocampal memory representations, and that endocannabinoids and BNDF signaling may contribute to the synergic modulation of underlying neural plasticity mechanisms.
Collapse
Affiliation(s)
- Blanca Marin Bosch
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Aurélien Bringard
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, 1205, Geneva, Switzerland.,Pulmonology Division, Geneva University Hospital, Geneva, Switzerland
| | - Maria G Logrieco
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Estelle Lauer
- Unit of Toxicology, CURML, Lausanne University Hospital and Geneva University Hospitals, Geneva, Switzerland
| | - Nathalie Imobersteg
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, 1205, Geneva, Switzerland
| | - Aurélien Thomas
- Unit of Toxicology, CURML, Lausanne University Hospital and Geneva University Hospitals, Geneva, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Chemin Vulliette 4, 1000, Lausanne, Switzerland
| | - Guido Ferretti
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, 1205, Geneva, Switzerland
| | - Sophie Schwartz
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Affective Sciences, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland
| | - Kinga Igloi
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland. .,Swiss Center for Affective Sciences, University of Geneva, Geneva, Switzerland. .,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
13
|
Leschik J, Lutz B, Gentile A. Stress-Related Dysfunction of Adult Hippocampal Neurogenesis-An Attempt for Understanding Resilience? Int J Mol Sci 2021; 22:7339. [PMID: 34298958 PMCID: PMC8305135 DOI: 10.3390/ijms22147339] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Newborn neurons in the adult hippocampus are regulated by many intrinsic and extrinsic cues. It is well accepted that elevated glucocorticoid levels lead to downregulation of adult neurogenesis, which this review discusses as one reason why psychiatric diseases, such as major depression, develop after long-term stress exposure. In reverse, adult neurogenesis has been suggested to protect against stress-induced major depression, and hence, could serve as a resilience mechanism. In this review, we will summarize current knowledge about the functional relation of adult neurogenesis and stress in health and disease. A special focus will lie on the mechanisms underlying the cascades of events from prolonged high glucocorticoid concentrations to reduced numbers of newborn neurons. In addition to neurotransmitter and neurotrophic factor dysregulation, these mechanisms include immunomodulatory pathways, as well as microbiota changes influencing the gut-brain axis. Finally, we discuss recent findings delineating the role of adult neurogenesis in stress resilience.
Collapse
Affiliation(s)
- Julia Leschik
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
| | - Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany;
- Leibniz Institute for Resilience Research (LIR), 55122 Mainz, Germany
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy;
| |
Collapse
|
14
|
Parihar VK, Syage A, Flores L, Lilagan A, Allen BD, Angulo MC, Song J, Smith SM, Arechavala RJ, Giedzinski E, Limoli CL. The Cannabinoid Receptor 1 Reverse Agonist AM251 Ameliorates Radiation-Induced Cognitive Decrements. Front Cell Neurosci 2021; 15:668286. [PMID: 34262437 PMCID: PMC8273551 DOI: 10.3389/fncel.2021.668286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/26/2021] [Indexed: 11/13/2022] Open
Abstract
Despite advancements in the radiotherapeutic management of brain malignancies, resultant sequelae include persistent cognitive dysfunction in the majority of survivors. Defining the precise causes of normal tissue toxicity has proven challenging, but the use of preclinical rodent models has suggested that reductions in neurogenesis and microvascular integrity, impaired synaptic plasticity, increased inflammation, and alterations in neuronal structure are contributory if not causal. As such, strategies to reverse these persistent radiotherapy-induced neurological disorders represent an unmet medical need. AM251, a cannabinoid receptor 1 reverse agonist known to facilitate adult neurogenesis and synaptic plasticity, may help to ameliorate radiation-induced CNS impairments. To test this hypothesis, three treatment paradigms were used to evaluate the efficacy of AM251 to ameliorate radiation-induced learning and memory deficits along with disruptions in mood at 4 and 12 weeks postirradiation. Results demonstrated that acute (four weekly injections) and chronic (16 weekly injections) AM251 treatments (1 mg/kg) effectively alleviated cognitive and mood dysfunction in cranially irradiated mice. The beneficial effects of AM251 were exemplified by improved hippocampal- and cortical-dependent memory function on the novel object recognition and object in place tasks, while similar benefits on mood were shown by reductions in depressive- and anxiety-like behaviors on the forced swim test and elevated plus maze. The foregoing neurocognitive benefits were associated with significant increases in newly born (doublecortin+) neurons (1.7-fold), hippocampal neurogenesis (BrdU+/NeuN+mature neurons, 2.5-fold), and reduced expression of the inflammatory mediator HMGB (1.2-fold) in the hippocampus of irradiated mice. Collectively, these findings indicate that AM251 ameliorates the effects of clinically relevant cranial irradiation where overall neurological benefits in memory and mood coincided with increased hippocampal cell proliferation, neurogenesis, and reduced expression of proinflammatory markers.
Collapse
Affiliation(s)
- Vipan K Parihar
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Amber Syage
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Lidia Flores
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Angelica Lilagan
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Maria C Angulo
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Joseph Song
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Sarah M Smith
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Rebecca J Arechavala
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
15
|
Neurobiological Processes Induced by Aerobic Exercise through the Endocannabinoidome. Cells 2021; 10:cells10040938. [PMID: 33920695 PMCID: PMC8072750 DOI: 10.3390/cells10040938] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/31/2021] [Accepted: 04/13/2021] [Indexed: 12/23/2022] Open
Abstract
Evidence suggesting the triangulation of the endocannabinoid system, exercise, and neurological health is emerging. In addition to the endocannabinoids N-arachidonoylethanolamine (anandamide; AEA) and 2-arachidonoylglycerol (2-AG), the expanded endocannabinoid system, known as the endocannabinoidome (eCBome), appears to be an important player in this relationship. The eCBome includes several endocannabinoid-like mediators such as N-acylethanolamines and 2-monoacylglycerols, the enzymes involved in their biosynthesis and degradation, and the receptors they affect. This review aims to relate the functional interactions between aerobic exercise, and the molecular and cellular pathways related to endocannabinoids, in the hypothalamus, hippocampus, and the periphery, with special attention given to associations with emotional state, cognition, and mental health. Given the well-documented roles of many eCBome members in regulating stress and neurological processes, we posit that the eCBome is an important effector of exercise-induced central and peripheral adaptive mechanisms that benefit mental health. Gut microbiota imbalance, affecting the gut-brain axis and metabolism, also influences certain eCBome-modulated inflammation pathways. The integrity of the gut microbiota could thus be crucial in the onset of neuroinflammation and mental conditions. Further studies on how the modulation by exercise of the peripheral eCBome affects brain functions could reveal to be key elements in the prevention and treatment of neuropsychological disorders.
Collapse
|
16
|
Marin Bosch B, Bringard A, Logrieco MG, Lauer E, Imobersteg N, Thomas A, Ferretti G, Schwartz S, Igloi K. Effect of acute physical exercise on motor sequence memory. Sci Rep 2020; 10:15322. [PMID: 32948800 PMCID: PMC7501852 DOI: 10.1038/s41598-020-72108-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/26/2020] [Indexed: 12/19/2022] Open
Abstract
Acute physical exercise improves memory functions by increasing neural plasticity in the hippocampus. In animals, a single session of physical exercise has been shown to boost anandamide (AEA), an endocannabinoid known to promote hippocampal plasticity. Hippocampal neuronal networks encode episodic memory representations, including the temporal organization of elements, and can thus benefit motor sequence learning. While previous work established that acute physical exercise has positive effects on declarative memory linked to hippocampal plasticity mechanisms, its influence on memory for motor sequences, and especially on neural mechanisms underlying possible effects, has been less investigated. Here we studied the impact of acute physical exercise on motor sequence learning, and its underlying neurophysiological mechanisms in humans, using a cross-over randomized within-subjects design. We measured behavior, fMRI activity, and circulating AEA levels in fifteen healthy participants while they performed a serial reaction time task before and after a short period of exercise (moderate or high intensity) or rest. We show that exercise enhanced motor sequence memory, significantly for high intensity exercise and tending towards significance for moderate intensity exercise. This enhancement correlated with AEA increase, and dovetailed with local increases in caudate nucleus and hippocampus activity. These findings demonstrate that acute physical exercise promotes sequence learning, thus attesting the overarching benefit of exercise to hippocampus-related memory functions.
Collapse
Affiliation(s)
- Blanca Marin Bosch
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| | - Aurélien Bringard
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, Geneva, Switzerland
| | - Maria Grazia Logrieco
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Estelle Lauer
- Unit of Toxicology, CURML, Lausanne University Hospital and Geneva University Hospitals, Geneva, Switzerland
| | - Nathalie Imobersteg
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, Geneva, Switzerland
| | - Aurélien Thomas
- Unit of Toxicology, CURML, Lausanne University Hospital and Geneva University Hospitals, Geneva, Switzerland.,Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Guido Ferretti
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Anesthesiology, Pharmacology and Intensive Care, Geneva University Hospitals, Geneva, Switzerland
| | - Sophie Schwartz
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Affective Sciences, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland
| | - Kinga Igloi
- Department of Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss Center for Affective Sciences, University of Geneva, Geneva, Switzerland.,Geneva Neuroscience Center, University of Geneva, Geneva, Switzerland
| |
Collapse
|
17
|
Abstract
The endocannabinoid system (ECS) is a highly versatile signaling system within the nervous system. Despite its widespread localization, its functions within the context of distinct neural processes are very well discernable and specific. This is remarkable, and the question remains as to how such specificity is achieved. One key player in the ECS is the cannabinoid type 1 receptor (CB1), a G protein-coupled receptor characterized by the complexity of its cell-specific expression, cellular and subcellular localization, and its adaptable regulation of intracellular signaling cascades. CB1 receptors are involved in different synaptic and cellular plasticity processes and in the brain's bioenergetics in a context-specific manner. CB2 receptors are also important in several processes in neurons, glial cells, and immune cells of the brain. As polymorphisms in ECS components, as well as external impacts such as stress and metabolic challenges, can both lead to dysregulated ECS activity and subsequently to possible neuropsychiatric disorders, pharmacological intervention targeting the ECS is a promising therapeutic approach. Understanding the neurobiology of cannabinoid receptor signaling in depth will aid optimal design of therapeutic interventions, minimizing unwanted side effects.
.
Collapse
Affiliation(s)
- Beat Lutz
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
18
|
Charytoniuk T, Zywno H, Konstantynowicz-Nowicka K, Berk K, Bzdega W, Chabowski A. Can Physical Activity Support the Endocannabinoid System in the Preventive and Therapeutic Approach to Neurological Disorders? Int J Mol Sci 2020; 21:E4221. [PMID: 32545780 PMCID: PMC7352563 DOI: 10.3390/ijms21124221] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/07/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022] Open
Abstract
The worldwide prevalence of neurological and neurodegenerative disorders, such as depression or Alzheimer's disease, has spread extensively throughout the last decades, becoming an enormous health issue. Numerous data indicate a distinct correlation between the altered endocannabinoid signaling and different aspects of brain physiology, such as memory or neurogenesis. Moreover, the endocannabinoid system is widely regarded as a crucial factor in the development of neuropathologies. Thus, targeting those disorders via synthetic cannabinoids, as well as phytocannabinoids, becomes a widespread research issue. Over the last decade, the endocannabinoid system has been extensively studied for its correlation with physical activity. Recent data showed that physical activity correlates with elevated endocannabinoid serum concentrations and increased cannabinoid receptor type 1 (CB1R) expression in the brain, which results in positive neurological effects including antidepressant effect, ameliorated memory, neuroplasticity development, and reduced neuroinflammation. However, none of the prior reviews presented a comprehensive correlation between physical activity, the endocannabinoid system, and neuropathologies. Thus, our review provides a current state of knowledge of the endocannabinoid system, its action in physical activity, as well as neuropathologies and a possible correlation between all those fields. We believe that this might contribute to finding a new preventive and therapeutic approach to both neurological and neurodegenerative disorders.
Collapse
Affiliation(s)
- Tomasz Charytoniuk
- Department of Physiology, Medical University of Bialystok, 15-089 Białystok, Poland; (H.Z.); (K.K.-N.); (K.B.); (W.B.); (A.C.)
| | | | | | | | | | | |
Collapse
|
19
|
Recovery of BDNF and CB1R in the Prefrontal Cortex Underlying Improvement of Working Memory in Prenatal DEHP-Exposed Male Rats after Aerobic Exercise. Int J Mol Sci 2020; 21:ijms21113867. [PMID: 32485872 PMCID: PMC7312003 DOI: 10.3390/ijms21113867] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/21/2020] [Accepted: 05/28/2020] [Indexed: 01/02/2023] Open
Abstract
Early-life exposure to di-(2-ethylhexyl)-phthalate (DEHP) has been suggested to relate to hyperactivity, lack of attention, and working memory deficits in school-age children. Brain-derived neurotrophic factor (BDNF) and endocannabinoids are induced by aerobic exercises to provide beneficial effects on brain functions. This study investigated the mechanisms underlying working memory impairment and the protective role of exercise in prenatal DEHP-exposed male rats. Sprague Dawley dams were fed with vehicle or DEHP during gestation. The male offspring were trained to exercise on a treadmill for 5 weeks, which was followed by an assessment of their working memory with a T-maze delayed non-match-to-sample task. The expressions of BDNF, dopamine D1 receptor (D1R), cannabinoid receptor 1 (CB1R), and fatty acid amide hydrolase (FAAH) in the prefrontal cortex were detected by Western blot. The results showed that DEHP-exposed rats exhibited working memory impairments without significant alterations in locomotor activities. The reduced expressions of prefrontal BDNF and CB1R were obtained in the DEHP-exposed rats, while D1R and FAAH were barely affected. Importantly, aerobic exercise during childhood-adolescence prevented the impairment of working memory in the DEHP-exposed rats by recovering the BDNF and CB1R expressions in the prefrontal cortex. These findings suggest that exercise may provide beneficial effects in ameliorating the impairment of working memory in the prenatal DEHP-exposed male rats at late adolescence.
Collapse
|
20
|
Sullivan RM, Wallace AL, Wade NE, Swartz AM, Lisdahl KM. Assessing the Role of Cannabis Use on Cortical Surface Structure in Adolescents and Young Adults: Exploring Gender and Aerobic Fitness as Potential Moderators. Brain Sci 2020; 10:E117. [PMID: 32098300 PMCID: PMC7071505 DOI: 10.3390/brainsci10020117] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/05/2022] Open
Abstract
Cannabis use in adolescents and young adults is linked with aberrant brain structure, although findings to date are inconsistent. We examined whether aerobic fitness moderated the effects of cannabis on cortical surface structure and whether gender may play a moderating role. Seventy-four adolescents and young adults completed three-weeks of monitored abstinence, aerobic fitness testing, and structural magnetic resonance imaging (sMRI). Whole-sample linear regressions examined the effects of gender, VO2 max, cannabis use, and their interactions on the surface area (SA) and local gyrification index (LGI). Cannabis use was associated with greater cuneus SA. Gender-by-cannabis predicted precuneus and frontal SA, and precentral, supramarginal, and frontal LGI; female cannabis users demonstrated greater LGI, whereas male cannabis users demonstrated decreased LGI compared to non-users. Aerobic fitness was positively associated with various SA and LGI regions. Cannabis-by-aerobic fitness predicted cuneus SA and occipital LGI. These findings demonstrate that aerobic fitness moderates the impact of cannabis on cortical surface structure, and gender differences are evident. These moderating factors may help explain inconsistencies in the literature and warrant further investigation. Present findings and aerobic fitness literature jointly suggest aerobic intervention may be a low-cost avenue for improving cortical surface structure, although the impact may be gender-specific.
Collapse
Affiliation(s)
- Ryan M. Sullivan
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA; (R.M.S.); (A.L.W.)
| | - Alexander L. Wallace
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA; (R.M.S.); (A.L.W.)
| | - Natasha E. Wade
- Department of Psychiatry, University of California, San Diego, La Jolla, CA 92093, USA;
| | - Ann M. Swartz
- Department of Kinesiology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA;
| | - Krista M. Lisdahl
- Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA; (R.M.S.); (A.L.W.)
| |
Collapse
|
21
|
Di Marzo V, Silvestri C. Lifestyle and Metabolic Syndrome: Contribution of the Endocannabinoidome. Nutrients 2019; 11:nu11081956. [PMID: 31434293 PMCID: PMC6722643 DOI: 10.3390/nu11081956] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022] Open
Abstract
Lifestyle is a well-known environmental factor that plays a major role in facilitating the development of metabolic syndrome or eventually exacerbating its consequences. Various lifestyle factors, especially changes in dietary habits, extreme temperatures, unusual light-dark cycles, substance abuse, and other stressful factors, are also established modifiers of the endocannabinoid system and its extended version, the endocannabinoidome. The endocannabinoidome is a complex lipid signaling system composed of a plethora (>100) of fatty acid-derived mediators and their receptors and anabolic and catabolic enzymes (>50 proteins) which are deeply involved in the control of energy metabolism and its pathological deviations. A strong link between the endocannabinoidome and another major player in metabolism and dysmetabolism, the gut microbiome, is also emerging. Here, we review several examples of how lifestyle modifications (westernized diets, lack or presence of certain nutritional factors, physical exercise, and the use of cannabis) can modulate the propensity to develop metabolic syndrome by modifying the crosstalk between the endocannabinoidome and the gut microbiome and, hence, how lifestyle interventions can provide new therapies against cardiometabolic risk by ensuring correct functioning of both these systems.
Collapse
Affiliation(s)
- Vincenzo Di Marzo
- École de nutrition, Université Laval, Québec, QC G1V 0A6, Canada
- Institut sur la nutrition et les aliments fonctionnels, Université Laval, Québec, QC G1V 0A6, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec, QC G1V 0A6, Canada
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC G1V 4G5, Canada
- Department de médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche, 80078 Pozzuoli, Italy
| | - Cristoforo Silvestri
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec, QC G1V 0A6, Canada.
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec, Québec, QC G1V 4G5, Canada.
- Department de médecine, Université Laval, Québec, QC G1V 0A6, Canada.
| |
Collapse
|
22
|
Loprinzi PD, Zou L, Li H. The Endocannabinoid System as a Potential Mechanism through which Exercise Influences Episodic Memory Function. Brain Sci 2019; 9:E112. [PMID: 31100856 PMCID: PMC6562547 DOI: 10.3390/brainsci9050112] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 05/13/2019] [Accepted: 05/14/2019] [Indexed: 12/17/2022] Open
Abstract
Emerging research demonstrates that exercise, including both acute and chronic exercise, may influence episodic memory function. To date, mechanistic explanations of this effect are often attributed to alterations in long-term potentiation, neurotrophic production, angiogenesis, and neurogenesis. Herein, we discuss a complementary mechanistic model, suggesting that the endocannabinoid system may, in part, influence the effects of exercise on memory function. We discuss the role of the endocannabinoid system on memory function as well as the effects of exercise on endocannabinoid alterations. This is an exciting line of inquiry that should help delineate new insights into the mechanistic role of exercise on memory function.
Collapse
Affiliation(s)
- Paul D Loprinzi
- Exercise & Memory Laboratory, Department of Health, Exercise Science and Recreation Management, The University of Mississippi, Oxford, MS 38677, USA.
| | - Liye Zou
- Lifestyle (Mind-Body Movement) Research Center, College of Psychology and Sociology, Shenzhen University, Shenzhen 518060, China.
| | - Hong Li
- Shenzhen Key Laboratory of Affective and Social Cognitive Science, College of Psychology and Sociology, Shenzhen University, Shenzhen 518060, China.
- Research Centre of Brain Function and Psychological Science, Shenzhen University, Shenzhen 518060, China.
- Shenzhen Institute of Neuroscience, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
23
|
Rodrigues RS, Lourenço DM, Paulo SL, Mateus JM, Ferreira MF, Mouro FM, Moreira JB, Ribeiro FF, Sebastião AM, Xapelli S. Cannabinoid Actions on Neural Stem Cells: Implications for Pathophysiology. Molecules 2019; 24:E1350. [PMID: 30959794 PMCID: PMC6480122 DOI: 10.3390/molecules24071350] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/01/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023] Open
Abstract
With the increase of life expectancy, neurodegenerative disorders are becoming not only a health but also a social burden worldwide. However, due to the multitude of pathophysiological disease states, current treatments fail to meet the desired outcomes. Therefore, there is a need for new therapeutic strategies focusing on more integrated, personalized and effective approaches. The prospect of using neural stem cells (NSC) as regenerative therapies is very promising, however several issues still need to be addressed. In particular, the potential actions of pharmacological agents used to modulate NSC activity are highly relevant. With the ongoing discussion of cannabinoid usage for medical purposes and reports drawing attention to the effects of cannabinoids on NSC regulation, there is an enormous, and yet, uncovered potential for cannabinoids as treatment options for several neurological disorders, specifically when combined with stem cell therapy. In this manuscript, we review in detail how cannabinoids act as potent regulators of NSC biology and their potential to modulate several neurogenic features in the context of pathophysiology.
Collapse
Affiliation(s)
- Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Diogo M Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Sara L Paulo
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Joana M Mateus
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Miguel F Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Francisco M Mouro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - João B Moreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Filipa F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, 1649-028 Lisboa, Portugal.
| |
Collapse
|
24
|
Cuccurazzu B, Zamberletti E, Nazzaro C, Prini P, Trusel M, Grilli M, Parolaro D, Tonini R, Rubino T. Adult Cellular Neuroadaptations Induced by Adolescent THC Exposure in Female Rats Are Rescued by Enhancing Anandamide Signaling. Int J Neuropsychopharmacol 2018; 21:1014-1024. [PMID: 29982505 PMCID: PMC6209859 DOI: 10.1093/ijnp/pyy057] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 06/27/2018] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND In rodent models, chronic exposure to cannabis' psychoactive ingredient, Δ9-tetrahydrocannabinol, during adolescence leads to abnormal behavior in adulthood. In female rats, this maladaptive behavior is characterized by endophenotypes for depressive-like and psychotic-like disorders as well as cognitive deficits. We recently reported that most depressive-like behaviors triggered by adolescent Δ9-tetrahydrocannabinol exposure can be rescued by manipulating endocannabinoid signaling in adulthood with the anandamide-inactivating enzyme FAAH inhibitor, URB597. However, the molecular mechanisms underlying URB597's antidepressant-like properties remain to be established. METHODS Here we examined the impact of adult URB597 treatment on the cellular and functional neuroadaptations that occurred in the prefrontal cortex and dentate gyrus of the hippocampus upon Δ9-tetrahydrocannabinol during adolescence through biochemical, morphofunctional, and electrophysiological studies. RESULTS We found that the positive action of URB597 is associated with the rescue of Δ9-tetrahydrocannabinol-induced deficits in endocannabinoid-mediated signaling and synaptic plasticity in the prefrontal cortex and the recovery of functional neurogenesis in the dentate gyrus of the hippocampus. Moreover, the rescue property of URB597 on depressive-like behavior requires the activity of the CB1 cannabinoid receptor. CONCLUSIONS By providing novel insights into the cellular and molecular mechanisms of URB597 at defined cortical and hippocampal circuits, our results highlight that positive modulation of endocannabinoid-signaling could be a strategy for treating mood alterations secondary to adolescent cannabis use.
Collapse
Affiliation(s)
- Bruna Cuccurazzu
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro,” Novara, Italy
| | - Erica Zamberletti
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy
| | - Cristiano Nazzaro
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Pamela Prini
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy
| | - Massimo Trusel
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Mariagrazia Grilli
- Department of Pharmaceutical Sciences, University of Piemonte Orientale “A. Avogadro,” Novara, Italy
| | - Daniela Parolaro
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy,Zardi Gori Foundation, Milan, Italy
| | - Raffaella Tonini
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia (IIT), Genova, Italy
| | - Tiziana Rubino
- Department of Biotechnology and Life Sciences, and Neuroscience Center, University of Insubria, Busto Arsizio VA, Italy,Correspondence: Tiziana Rubino, PhD, Department of Biotechnology and Life Sciences and Neuroscience Center, University of Insubria, via Manara 7, 21052 Busto Arsizio VA, Italy ()
| |
Collapse
|
25
|
Hamedinia M, Sharifi M, Hosseini-Kakhak A. The Effect of Eight Weeks of Aerobic, Anaerobic and Resistance Training on some factor of Endocannabinoid System, Serotonin, Beta-Endorphin and BDNF in Young Men. ACTA ACUST UNITED AC 2017. [DOI: 10.13005/bbra/2562] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
ABSTRACT: The aim of this study was investigated the effect of eight weeks of aerobic, anaerobic and resistance training on some endocannabinoid, serotonin, beta-endorphin and BDNF agents of young men. Thirty-two young men (19 to 25 years old) who did not have regular physical activity were randomly divided into four groups, and each of them were participated in various sports exercises for eight weeks. A group for aerobic exercises (two exercises with 65-70 maximum heart rate 3 sessions per week), an anaerobic exercise group (two exercises with a maximum intensity of 3 sessions per week), a group for circular resistance exercises (6-8 Station Which is repeated 8-12 times, and for three times a week) and finally a group was selected as a control. ELISA method was used to measure endocannabinoid system, serotonin, beta-endorphin and BDNF factors. The results showed that eight weeks aerobic training significantly increased serotonin levels and eight weeks aerobic and anaerobic exercise significantly increased BDNF. Aerobic, anaerobic, and resistive exercises have no significant effect on arachidonoyl glycerol (2-AG), anandamide(AEA) and beta-endorphin. There was a significant increase in happiness in all three training groups compared to the control group.The results of this study indicated an increase in mediation associated with pleasure and happiness in humans. Concerning the particular effect of long-term exercise on the endocannabinoid system, it is difficult to conclude.
Collapse
Affiliation(s)
- Mohammadreza Hamedinia
- Department of Sport Physiology, Faculty of Physical Education and Sports Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Moslem Sharifi
- Department of Sport Physiology, Faculty of Physical Education and Sports Science, Hakim Sabzevari University, Sabzevar, Iran
| | - Alireza Hosseini-Kakhak
- Department of Sport Physiology, Faculty of Physical Education and Sports Science, Hakim Sabzevari University, Sabzevar, Iran
| |
Collapse
|
26
|
Rodrigues RS, Ribeiro FF, Ferreira F, Vaz SH, Sebastião AM, Xapelli S. Interaction between Cannabinoid Type 1 and Type 2 Receptors in the Modulation of Subventricular Zone and Dentate Gyrus Neurogenesis. Front Pharmacol 2017; 8:516. [PMID: 28848435 PMCID: PMC5554396 DOI: 10.3389/fphar.2017.00516] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 07/24/2017] [Indexed: 01/13/2023] Open
Abstract
Neurogenesis in the adult mammalian brain occurs mainly in two neurogenic niches, the subventricular zone (SVZ) and the subgranular zone (SGZ) of the dentate gyrus (DG). Cannabinoid type 1 and 2 receptors (CB1R and CB2R) have been shown to differently modulate neurogenesis. However, low attention has been given to the interaction between CB1R and CB2R in modulating postnatal neurogenesis (proliferation, neuronal differentiation and maturation). We focused on a putative crosstalk between CB1R and CB2R to modulate neurogenesis and cultured SVZ and DG stem/progenitor cells from early postnatal (P1-3) Sprague-Dawley rats. Data showed that the non-selective cannabinoid receptor agonist WIN55,212-2 promotes DG cell proliferation (measured by BrdU staining), an effect blocked by either CB1R or CB2R selective antagonists. Experiments with selective agonists showed that facilitation of DG cell proliferation requires co-activation of both CB1R and CB2R. Cell proliferation in the SVZ was not affected by the non-selective receptor agonist, but it was enhanced by CB1R selective activation. However, either CB1R or CB2R selective antagonists abolished the effect of the CB1R agonist in SVZ cell proliferation. Neuronal differentiation (measured by immunocytochemistry against neuronal markers of different stages and calcium imaging) was facilitated by WIN55,212-2 at both SVZ and DG. This effect was mimicked by either CB1R or CB2R selective agonists and blocked by either CB1R or CB2R selective antagonists, cross-antagonism being evident. In summary, our findings indicate a tight interaction between CB1R and CB2R to modulate neurogenesis in the two major neurogenic niches, thus contributing to further unraveling the mechanisms behind the action of endocannabinoids in the brain.
Collapse
Affiliation(s)
- Rui S Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal
| | - Filipa F Ribeiro
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal
| | - Filipa Ferreira
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal
| | - Sandra H Vaz
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal.,Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de LisboaLisboa, Portugal
| |
Collapse
|
27
|
Alkadhi KA. Exercise as a Positive Modulator of Brain Function. Mol Neurobiol 2017; 55:3112-3130. [PMID: 28466271 DOI: 10.1007/s12035-017-0516-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 04/04/2017] [Indexed: 12/24/2022]
Abstract
Various forms of exercise have been shown to prevent, restore, or ameliorate a variety of brain disorders including dementias, Parkinson's disease, chronic stress, thyroid disorders, and sleep deprivation, some of which are discussed here. In this review, the effects on brain function of various forms of exercise and exercise mimetics in humans and animal experiments are compared and discussed. Possible mechanisms of the beneficial effects of exercise including the role of neurotrophic factors and others are also discussed.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, 77204, USA.
| |
Collapse
|
28
|
Hueston CM, Cryan JF, Nolan YM. Stress and adolescent hippocampal neurogenesis: diet and exercise as cognitive modulators. Transl Psychiatry 2017; 7:e1081. [PMID: 28375209 PMCID: PMC5416690 DOI: 10.1038/tp.2017.48] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 01/04/2017] [Accepted: 02/01/2017] [Indexed: 02/08/2023] Open
Abstract
Adolescence is a critical period for brain maturation. Deciphering how disturbances to the central nervous system at this time affect structure, function and behavioural outputs is important to better understand any long-lasting effects. Hippocampal neurogenesis occurs during development and continues throughout life. In adulthood, integration of these new cells into the hippocampus is important for emotional behaviour, cognitive function and neural plasticity. During the adolescent period, maturation of the hippocampus and heightened levels of hippocampal neurogenesis are observed, making alterations to neurogenesis at this time particularly consequential. As stress negatively affects hippocampal neurogenesis, and adolescence is a particularly stressful time of life, it is important to investigate the impact of stressor exposure at this time on hippocampal neurogenesis and cognitive function. Adolescence may represent not only a time for which stress can have long-lasting effects, but is also a critical period during which interventions, such as exercise and diet, could ameliorate stress-induced changes to hippocampal function. In addition, intervention at this time may also promote life-long behavioural changes that would aid in fostering increased hippocampal neurogenesis and cognitive function. This review addresses both the acute and long-term stress-induced alterations to hippocampal neurogenesis and cognition during the adolescent period, as well as changes to the stress response and pubertal hormones at this time which may result in differential effects than are observed in adulthood. We hypothesise that adolescence may represent an optimal time for healthy lifestyle changes to have a positive and long-lasting impact on hippocampal neurogenesis, and to protect against stress-induced deficits. We conclude that future research into the mechanisms underlying the susceptibility of the adolescent hippocampus to stress, exercise and diet and the consequent effect on cognition may provide insight into why adolescence may be a vital period for correct conditioning of future hippocampal function.
Collapse
Affiliation(s)
- C M Hueston
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - J F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Y M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
29
|
Exercise training and high-fat diet elicit endocannabinoid system modifications in the rat hypothalamus and hippocampus. J Physiol Biochem 2017; 73:335-347. [PMID: 28283967 DOI: 10.1007/s13105-017-0557-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 02/23/2017] [Indexed: 01/03/2023]
Abstract
The purpose of the present study was to examine the effect of chronic exercise on the hypothalamus and hippocampus levels of the endocannabinoids (eCBs) anandamide (AEA) and 2-arachidonoylglycerol (2-AG) and of two AEA congeners and on the expression of genes coding for CB1, CB2 receptors (Cnr1 and Cnr2, respectively), and the enzymes responsible for eCB biosynthesis and degradation, in rats fed with a standard or high-fat diet. Male Wistar rats (n = 28) were placed on a 12-week high-fat (HFD) or standard diet period, followed by 12 weeks of exercise training for half of each group. Tissue levels of eCBs and related lipids were measured by liquid chromatography mass spectrometry, and expression of genes coding for CB1 and CB2 receptors and eCB metabolic enzymes was measured by quantitative real-time polymerase chain reaction (qPCR). HFD induced a significant increase in 2-AG (p < 0.01) in hypothalamus. High-fat diet paired with exercise training had no effect on AEA, 2-AG, and AEA congener levels in the hypothalamus and hippocampus. Cnr1 expression levels were significantly increased in the hippocampus in response to HFD, exercise, and the combination of both (p < 0.05). Our results indicate that eCB signaling in the CNS is sensitive to diet and/or exercise.
Collapse
|
30
|
Thompson Z, Argueta D, Garland T, DiPatrizio N. Circulating levels of endocannabinoids respond acutely to voluntary exercise, are altered in mice selectively bred for high voluntary wheel running, and differ between the sexes. Physiol Behav 2016; 170:141-150. [PMID: 28017680 DOI: 10.1016/j.physbeh.2016.11.041] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/07/2016] [Accepted: 11/22/2016] [Indexed: 11/25/2022]
Abstract
The endocannabinoid system serves many physiological roles, including in the regulation of energy balance, food reward, and voluntary locomotion. Signaling at the cannabinoid type 1 receptor has been specifically implicated in motivation for rodent voluntary exercise on wheels. We studied four replicate lines of high runner (HR) mice that have been selectively bred for 81 generations based on average number of wheel revolutions on days five and six of a six-day period of wheel access. Four additional replicate lines are bred without regard to wheel running, and serve as controls (C) for random genetic effects that may cause divergence among lines. On average, mice from HR lines voluntarily run on wheels three times more than C mice on a daily basis. We tested the general hypothesis that circulating levels of endocannabinoids (i.e., 2-arachidonoylglycerol [2-AG] and anandamide [AEA]) differ between HR and C mice in a sex-specific manner. Fifty male and 50 female mice were allowed access to wheels for six days, while another 50 males and 50 females were kept without access to wheels (half HR, half C for all groups). Blood was collected by cardiac puncture during the time of peak running on the sixth night of wheel access or no wheel access, and later analyzed for 2-AG and AEA content by ultra-performance liquid chromatography coupled to tandem mass spectrometry. We observed a significant three-way interaction among sex, linetype, and wheel access for 2-AG concentrations, with females generally having lower levels than males and wheel access lowering 2-AG levels in some but not all subgroups. The number of wheel revolutions in the minutes or hours immediately prior to sampling did not quantitatively predict plasma 2-AG levels within groups. We also observed a trend for a linetype-by-wheel access interaction for AEA levels, with wheel access lowering plasma concentrations of AEA in HR mice, while raising them in C mice. In addition, females tended to have higher AEA concentrations than males. For mice housed with wheels, the amount of running during the 30min before sampling was a significant positive predictor of plasma AEA within groups, and HR mice had significantly lower levels of AEA than C mice. Our results suggest that voluntary exercise alters circulating levels of endocannabinoids, and further demonstrate that selective breeding for voluntary exercise is associated with evolutionary changes in the endocannabinoid system.
Collapse
Affiliation(s)
- Zoe Thompson
- Neuroscience Graduate Program, University of California, Riverside, CA 92521, USA
| | - Donovan Argueta
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| | - Theodore Garland
- Department of Biology, University of California, Riverside, CA 92521, USA.
| | - Nicholas DiPatrizio
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, CA 92521, USA
| |
Collapse
|
31
|
Brellenthin AG, Koltyn KF. Exercise as an adjunctive treatment for cannabis use disorder. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2016; 42:481-489. [PMID: 27314543 PMCID: PMC5055462 DOI: 10.1080/00952990.2016.1185434] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 04/22/2016] [Accepted: 04/29/2016] [Indexed: 01/19/2023]
Abstract
BACKGROUND Despite cannabis being the most widely used illicit substance in the United States, individuals diagnosed with cannabis use disorder (CUD) have few well-researched, affordable treatment options available to them. Although found to be effective for improving treatment outcomes in other drug populations, exercise is an affordable and highly accessible treatment approach that has not been routinely investigated in cannabis users. OBJECTIVES The aim of this paper is to inform the topic regarding exercise's potential as an adjunctive treatment for individuals with CUD. METHODS We reviewed the evidence surrounding cannabis use and its current treatment in the United States, explored the rationale for including exercise in the treatment of substance use disorders (SUDs), and in particular, proposed a biological mechanism (i.e., endocannabinoids (eCBs)) that should be examined when utilizing exercise for the treatment of CUD. RESULTS Cannabis use is widespread and increasing in the United States. Chronic, heavy cannabis use may dysregulate the endogenous cannabinoid system, which has implications for several psychobiological processes that interact with the eCB system such as reward processing and the stress response. Given that exercise is a potent activator of the eCB system, it is mechanistically plausible that exercise could be an optimal method to supplement cessation efforts by reducing psychophysical withdrawal, managing stress, and attenuating drug cravings. CONCLUSION We suggest there is a strong behavioral and physiological rationale to design studies which specifically assess the efficacy of exercise, in combination with other therapies, in treating CUD. Moreover, it will be especially important to include the investigation of psychobiological mechanisms (e.g., eCBs, hippocampal volume), which have been associated with both exercise and SUDs, to examine the broader impact of exercise on behavioral and physiological responses to treatment.
Collapse
Affiliation(s)
| | - Kelli F Koltyn
- a Department of Kinesiology , University of Wisconsin-Madison , Madison , WI , USA
| |
Collapse
|
32
|
Gillman AS, Hutchison KE, Bryan AD. Cannabis and Exercise Science: A Commentary on Existing Studies and Suggestions for Future Directions. Sports Med 2016; 45:1357-63. [PMID: 26178329 DOI: 10.1007/s40279-015-0362-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Policies regarding cannabis use are rapidly changing, yet public officials have limited access to scientific information that might inform the creation of these policies. One important area in which to begin investigations is the link between recreational cannabis use and health, specifically exercise. There are common anecdotal reports that cannabis decreases motivation, including motivation to exercise. On the other hand, there are also anecdotal reports that cannabis is used prior to athletic activity. In fact, the World Anti-Doping Agency includes cannabis as a prohibited substance in sport, partly because it is believed that it may enhance sports performance. At the current time, there is limited scientific evidence to support either one of these opposing lay perspectives. Given recent political, cultural, and legal trends, and the growing acceptance of recreational cannabis use, it is important to develop a more nuanced understanding of the relationship between cannabis and exercise, specifically the potential effects of use on exercise performance, motivation, and recovery.
Collapse
Affiliation(s)
- Arielle S Gillman
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244, 345 UCB, Boulder, CO, 80309-0345, USA.
| | - Kent E Hutchison
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244, 345 UCB, Boulder, CO, 80309-0345, USA
| | - Angela D Bryan
- Department of Psychology and Neuroscience, University of Colorado Boulder, Muenzinger D244, 345 UCB, Boulder, CO, 80309-0345, USA
| |
Collapse
|
33
|
Diamond A, Ling DS. Conclusions about interventions, programs, and approaches for improving executive functions that appear justified and those that, despite much hype, do not. Dev Cogn Neurosci 2016; 18:34-48. [PMID: 26749076 PMCID: PMC5108631 DOI: 10.1016/j.dcn.2015.11.005] [Citation(s) in RCA: 451] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/26/2015] [Accepted: 11/23/2015] [Indexed: 12/26/2022] Open
Abstract
The 'Executive Functions' (EFs) of inhibitory control, working memory, and cognitive flexibility enable us to think before we act, resist temptations or impulsive reactions, stay focused, reason, problem-solve, flexibly adjust to changed demands or priorities, and see things from new and different perspectives. These skills are critical for success in all life's aspects and are sometimes more predictive than even IQ or socioeconomic status. Understandably, there is great interest in improving EFs. It's now clear they can be improved at any age through training and practice, much as physical exercise hones physical fitness. However, despite claims to the contrary, wide transfer does not seem to occur and 'mindless' aerobic exercise does little to improve EFs. Important questions remain: How much can EFs be improved (are benefits only superficial) and how long can benefits be sustained? What are the best methods for improving EFs? What about an approach accounts for its success? Do the answers to these differ by individual characteristics such as age or gender? Since stress, sadness, loneliness, or poor health impair EFs, and the reverse enhances EFs, we predict that besides directly train EFs, the most successful approaches for improving EFs will also address emotional, social, and physical needs.
Collapse
Affiliation(s)
- Adele Diamond
- Program in Developmental Cognitive Neuroscience, Department of Psychiatry, UBC, 2255 Wesbrook Mall, Vancouver, BC, Canada V6T 2A1.
| | - Daphne S Ling
- Program in Developmental Cognitive Neuroscience, Department of Psychiatry, UBC, 2255 Wesbrook Mall, Vancouver, BC, Canada V6T 2A1
| |
Collapse
|
34
|
Cassilhas RC, Tufik S, de Mello MT. Physical exercise, neuroplasticity, spatial learning and memory. Cell Mol Life Sci 2016; 73:975-83. [PMID: 26646070 PMCID: PMC11108521 DOI: 10.1007/s00018-015-2102-0] [Citation(s) in RCA: 175] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/21/2015] [Accepted: 11/23/2015] [Indexed: 02/07/2023]
Abstract
There has long been discussion regarding the positive effects of physical exercise on brain activity. However, physical exercise has only recently begun to receive the attention of the scientific community, with major interest in its effects on the cognitive functions, spatial learning and memory, as a non-drug method of maintaining brain health and treating neurodegenerative and/or psychiatric conditions. In humans, several studies have shown the beneficial effects of aerobic and resistance exercises in adult and geriatric populations. More recently, studies employing animal models have attempted to elucidate the mechanisms underlying neuroplasticity related to physical exercise-induced spatial learning and memory improvement, even under neurodegenerative conditions. In an attempt to clarify these issues, the present review aims to discuss the role of physical exercise in the improvement of spatial learning and memory and the cellular and molecular mechanisms involved in neuroplasticity.
Collapse
Affiliation(s)
- Ricardo C Cassilhas
- Department of Physical Education, Universidade Federal dos Vales do Jequitinhonha e Mucuri (UFVJM), Diamantina, MG, Brazil.
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
| | - Sergio Tufik
- Department of Psychobiology, Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Marco Túlio de Mello
- School of Physical Education, Physiotherapy and Occupational Therapy (EEFFTO), Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil.
| |
Collapse
|
35
|
Heijnen S, Hommel B, Kibele A, Colzato LS. Neuromodulation of Aerobic Exercise-A Review. Front Psychol 2016; 6:1890. [PMID: 26779053 PMCID: PMC4703784 DOI: 10.3389/fpsyg.2015.01890] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/23/2015] [Indexed: 12/31/2022] Open
Abstract
Running, and aerobic exercise in general, is a physical activity that increasingly many people engage in but that also has become popular as a topic for scientific research. Here we review the available studies investigating whether and to which degree aerobic exercise modulates hormones, amino acids, and neurotransmitters levels. In general, it seems that factors such as genes, gender, training status, and hormonal status need to be taken into account to gain a better understanding of the neuromodular underpinnings of aerobic exercise. More research using longitudinal studies and considering individual differences is necessary to determine actual benefits. We suggest that, in order to succeed, aerobic exercise programs should include optimal periodization, prevent overtraining and be tailored to interindividual differences, including neuro-developmental and genetically-based factors.
Collapse
Affiliation(s)
- Saskia Heijnen
- Cognitive Psychology Unit, Leiden UniversityLeiden, Netherlands; Leiden Institute for Brain and Cognition, Leiden UniversityLeiden, Netherlands
| | - Bernhard Hommel
- Cognitive Psychology Unit, Leiden UniversityLeiden, Netherlands; Leiden Institute for Brain and Cognition, Leiden UniversityLeiden, Netherlands
| | - Armin Kibele
- Institute for Sports and Sport Science, University of Kassel Kassel, Germany
| | - Lorenza S Colzato
- Cognitive Psychology Unit, Leiden UniversityLeiden, Netherlands; Leiden Institute for Brain and Cognition, Leiden UniversityLeiden, Netherlands
| |
Collapse
|
36
|
Sex differences in drug addiction and response to exercise intervention: From human to animal studies. Front Neuroendocrinol 2016; 40:24-41. [PMID: 26182835 PMCID: PMC4712120 DOI: 10.1016/j.yfrne.2015.07.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 06/08/2015] [Accepted: 07/10/2015] [Indexed: 02/08/2023]
Abstract
Accumulated research supports the idea that exercise could be an option of potential prevention and treatment for drug addiction. During the past few years, there has been increased interest in investigating of sex differences in exercise and drug addiction. This demonstrates that sex-specific exercise intervention strategies may be important for preventing and treating drug addiction in men and women. However, little is known about how and why sex differences are found when doing exercise-induced interventions for drug addiction. In this review, we included both animal and human that pulled subjects from a varied age demographic, as well as neurobiological mechanisms that may highlight the sex-related differences in these potential to assess the impact of sex-specific roles in drug addiction and exercise therapies.
Collapse
|
37
|
Abstract
Exercise is rewarding, and long-distance runners have described a runner's high as a sudden pleasant feeling of euphoria, anxiolysis, sedation, and analgesia. A popular belief has been that endogenous endorphins mediate these beneficial effects. However, running exercise increases blood levels of both β-endorphin (an opioid) and anandamide (an endocannabinoid). Using a combination of pharmacologic, molecular genetic, and behavioral studies in mice, we demonstrate that cannabinoid receptors mediate acute anxiolysis and analgesia after running. We show that anxiolysis depends on intact cannabinoid receptor 1 (CB1) receptors on forebrain GABAergic neurons and pain reduction on activation of peripheral CB1 and CB2 receptors. We thus demonstrate that the endocannabinoid system is crucial for two main aspects of a runner's high. Sedation, in contrast, was not influenced by cannabinoid or opioid receptor blockage, and euphoria cannot be studied in mouse models.
Collapse
|
38
|
Mechanisms of exercise-induced hypoalgesia. THE JOURNAL OF PAIN 2015; 15:1294-1304. [PMID: 25261342 DOI: 10.1016/j.jpain.2014.09.006] [Citation(s) in RCA: 217] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 07/17/2014] [Accepted: 09/12/2014] [Indexed: 11/17/2022]
Abstract
UNLABELLED The purpose of this study was to examine opioid and endocannabinoid mechanisms of exercise-induced hypoalgesia (EIH). Fifty-eight men and women (mean age = 21 years) completed 3 sessions. During the first session, participants were familiarized with the temporal summation of heat pain and pressure pain protocols. In the exercise sessions, following double-blind administration of either an opioid antagonist (50 mg naltrexone) or placebo, participants rated the intensity of heat pulses and indicated their pressure pain thresholds and pressure pain ratings before and after 3 minutes of submaximal isometric exercise. Blood was drawn before and after exercise. Results indicated that circulating concentrations of 2 endocannabinoids, N-arachidonylethanolamine and 2-arachidonoylglycerol, as well as related lipids oleoylethanolamide, palmitoylethanolamide, N-docosahexaenoylethanolamine, and 2-oleoylglycerol, increased significantly (P < .05) following exercise. Pressure pain thresholds increased significantly (P < .05), whereas pressure pain ratings decreased significantly (P < .05) following exercise. Also, temporal summation ratings were significantly lower (P < .05) following exercise. These changes in pain responses did not differ between the placebo and naltrexone conditions (P > .05). A significant association was found between EIH and docosahexaenoylethanolamine. These results suggest involvement of a nonopioid mechanism in EIH following isometric exercise. PERSPECTIVE Currently, the mechanisms responsible for EIH are unknown. This study provides support for a potential endocannabinoid mechanism of EIH following isometric exercise.
Collapse
|
39
|
Prenderville JA, Kelly ÁM, Downer EJ. The role of cannabinoids in adult neurogenesis. Br J Pharmacol 2015; 172:3950-63. [PMID: 25951750 DOI: 10.1111/bph.13186] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 04/17/2015] [Accepted: 04/22/2015] [Indexed: 12/17/2022] Open
Abstract
The processes underpinning post-developmental neurogenesis in the mammalian brain continue to be defined. Such processes involve the proliferation of neural stem cells and neural progenitor cells (NPCs), neuronal migration, differentiation and integration into a network of functional synapses within the brain. Both intrinsic (cell signalling cascades) and extrinsic (neurotrophins, neurotransmitters, cytokines, hormones) signalling molecules are intimately associated with adult neurogenesis and largely dictate the proliferative activity and differentiation capacity of neural cells. Cannabinoids are a unique class of chemical compounds incorporating plant-derived cannabinoids (the active components of Cannabis sativa), the endogenous cannabinoids and synthetic cannabinoid ligands, and these compounds are becoming increasingly recognized for their roles in neural developmental processes. Indeed, cannabinoids have clear modulatory roles in adult neurogenesis, probably through activation of both CB1 and CB2 receptors. In recent years, a large body of literature has deciphered the signalling networks involved in cannabinoid-mediated regulation of neurogenesis. This timely review summarizes the evidence that the cannabinoid system is intricately associated with neuronal differentiation and maturation of NPCs and highlights intrinsic/extrinsic signalling mechanisms that are cannabinoid targets. Overall, these findings identify the central role of the cannabinoid system in adult neurogenesis in the hippocampus and the lateral ventricles and hence provide insight into the processes underlying post-developmental neurogenesis in the mammalian brain.
Collapse
Affiliation(s)
- Jack A Prenderville
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland.,Trinity College Institute of Neuroscience, University of Dublin, Trinity College, Dublin, Ireland
| | - Áine M Kelly
- Department of Physiology, School of Medicine, Trinity College, Dublin, Ireland.,Trinity College Institute of Neuroscience, University of Dublin, Trinity College, Dublin, Ireland
| | - Eric J Downer
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
40
|
Geil CR, Hayes DM, McClain JA, Liput DJ, Marshall SA, Chen KY, Nixon K. Alcohol and adult hippocampal neurogenesis: promiscuous drug, wanton effects. Prog Neuropsychopharmacol Biol Psychiatry 2014; 54:103-13. [PMID: 24842804 PMCID: PMC4134968 DOI: 10.1016/j.pnpbp.2014.05.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 05/01/2014] [Accepted: 05/08/2014] [Indexed: 01/29/2023]
Abstract
Adult neurogenesis is now widely accepted as an important contributor to hippocampal integrity and function but also dysfunction when adult neurogenesis is affected in neuropsychiatric diseases such as alcohol use disorders. Excessive alcohol consumption, the defining characteristic of alcohol use disorders, results in a variety of cognitive and behavioral impairments related wholly or in part to hippocampal structure and function. Recent preclinical work has shown that adult neurogenesis may be one route by which alcohol produces hippocampal neuropathology. Alcohol is a pharmacologically promiscuous drug capable of interfering with adult neurogenesis through multiple mechanisms. This review will discuss the primary mechanisms underlying alcohol-induced changes in adult hippocampal neurogenesis including alcohol's effects on neurotransmitters, CREB and its downstream effectors, and the neurogenic niche.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Kimberly Nixon
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY 40536, United States.
| |
Collapse
|
41
|
Zhang Z, Wang W, Zhong P, Liu SJ, Long JZ, Zhao L, Gao HQ, Cravatt BF, Liu QS. Blockade of 2-arachidonoylglycerol hydrolysis produces antidepressant-like effects and enhances adult hippocampal neurogenesis and synaptic plasticity. Hippocampus 2014; 25:16-26. [PMID: 25131612 DOI: 10.1002/hipo.22344] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/05/2014] [Indexed: 11/11/2022]
Abstract
The endocannabinoid ligand 2-arachidonoylglycerol (2-AG) is inactivated primarily by monoacylglycerol lipase (MAGL). We have shown recently that chronic treatments with MAGL inhibitor JZL184 produce antidepressant- and anxiolytic-like effects in a chronic unpredictable stress (CUS) model of depression in mice. However, the underlying mechanisms remain poorly understood. Adult hippocampal neurogenesis has been implicated in animal models of anxiety and depression and behavioral effects of antidepressants. We tested whether CUS and chronic JZL184 treatments affected adult neurogenesis and synaptic plasticity in the dentate gyrus (DG) of mouse hippocampus. We report that CUS induced depressive-like behaviors and decreased the number of bromodeoxyuridine-labeled neural progenitor cells and doublecortin-positive immature neurons in the DG, while chronic JZL184 treatments prevented these behavioral and cellular deficits. We also investigated the effects of CUS and chronic JZL184 on a form long-term potentiation (LTP) in the DG known to be neurogenesis-dependent. CUS impaired LTP induction, whereas chronic JZL184 treatments restored LTP in CUS-exposed mice. These results suggest that enhanced adult neurogenesis and long-term synaptic plasticity in the DG of the hippocampus might contribute to antidepressant- and anxiolytic-like behavioral effects of JZL184.
Collapse
Affiliation(s)
- Zhen Zhang
- Department of Geriatrics, Qi-Lu Hospital of Shandong University, Key Laboratory of Proteomics of Shandong Province, Jinan Shandong, China.,Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Wei Wang
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Peng Zhong
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Sarah J Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jonathan Z Long
- The Skaggs Institute for Chemical Biology, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Li Zhao
- Department of Sports Physiology, Beijing Sport University, Beijing 100084, China
| | - Hai-Qing Gao
- Department of Geriatrics, Qi-Lu Hospital of Shandong University, Key Laboratory of Proteomics of Shandong Province, Jinan Shandong, China
| | - Benjamin F Cravatt
- The Skaggs Institute for Chemical Biology, Department of Chemical Physiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Qing-Song Liu
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
42
|
McPartland JM, Guy GW, Di Marzo V. Care and feeding of the endocannabinoid system: a systematic review of potential clinical interventions that upregulate the endocannabinoid system. PLoS One 2014; 9:e89566. [PMID: 24622769 PMCID: PMC3951193 DOI: 10.1371/journal.pone.0089566] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 01/21/2014] [Indexed: 12/31/2022] Open
Abstract
Background The “classic” endocannabinoid (eCB) system includes the cannabinoid receptors CB1 and CB2, the eCB ligands anandamide (AEA) and 2-arachidonoylglycerol (2-AG), and their metabolic enzymes. An emerging literature documents the “eCB deficiency syndrome” as an etiology in migraine, fibromyalgia, irritable bowel syndrome, psychological disorders, and other conditions. We performed a systematic review of clinical interventions that enhance the eCB system—ways to upregulate cannabinoid receptors, increase ligand synthesis, or inhibit ligand degradation. Methodology/Principal Findings We searched PubMed for clinical trials, observational studies, and preclinical research. Data synthesis was qualitative. Exclusion criteria limited the results to 184 in vitro studies, 102 in vivo animal studies, and 36 human studies. Evidence indicates that several classes of pharmaceuticals upregulate the eCB system, including analgesics (acetaminophen, non-steroidal anti-inflammatory drugs, opioids, glucocorticoids), antidepressants, antipsychotics, anxiolytics, and anticonvulsants. Clinical interventions characterized as “complementary and alternative medicine” also upregulate the eCB system: massage and manipulation, acupuncture, dietary supplements, and herbal medicines. Lifestyle modification (diet, weight control, exercise, and the use of psychoactive substances—alcohol, tobacco, coffee, cannabis) also modulate the eCB system. Conclusions/Significance Few clinical trials have assessed interventions that upregulate the eCB system. Many preclinical studies point to other potential approaches; human trials are needed to explore these promising interventions.
Collapse
Affiliation(s)
- John M. McPartland
- GW Pharmaceuticals, Porton Down Science Park, Salisbury, Wiltshire, United Kingdom
- Department of Family Medicine, University of Vermont, Burlington, Vermont, United States of America
- * E-mail:
| | - Geoffrey W. Guy
- GW Pharmaceuticals, Porton Down Science Park, Salisbury, Wiltshire, United Kingdom
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomoleculare, CNR, Via Campi Flegrei, Pozzuoli, Napoli, Italy
| |
Collapse
|
43
|
Blanco-Calvo E, Rivera P, Arrabal S, Vargas A, Pavón FJ, Serrano A, Castilla-Ortega E, Galeano P, Rubio L, Suárez J, Rodriguez de Fonseca F. Pharmacological blockade of either cannabinoid CB1 or CB2 receptors prevents both cocaine-induced conditioned locomotion and cocaine-induced reduction of cell proliferation in the hippocampus of adult male rat. Front Integr Neurosci 2014; 7:106. [PMID: 24409127 PMCID: PMC3884150 DOI: 10.3389/fnint.2013.00106] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2013] [Accepted: 12/18/2013] [Indexed: 11/14/2022] Open
Abstract
Addiction to major drugs of abuse, such as cocaine, has recently been linked to alterations in adult neurogenesis in the hippocampus. The endogenous cannabinoid system modulates this proliferative response as demonstrated by the finding that pharmacological activation/blockade of cannabinoid CB1 and CB2 receptors not only modulates neurogenesis but also modulates cell death in the brain. In the present study, we evaluated whether the endogenous cannabinoid system affects cocaine-induced alterations in cell proliferation. To this end, we examined whether pharmacological blockade of either CB1 (Rimonabant, 3 mg/kg) or CB2 receptors (AM630, 3 mg/kg) would affect cell proliferation [the cells were labeled with 5-bromo-2′-deoxyuridine (BrdU)] in the subventricular zone (SVZ) of the lateral ventricle and the dentate subgranular zone (SGZ). Additionally, we measured cell apoptosis (as monitored by the expression of cleaved caspase-3) and glial activation [by analyzing the expression of glial fibrillary acidic protein (GFAP) and Iba-1] in the striatum and hippocampus during acute and repeated (4 days) cocaine administration (20 mg/kg). The results showed that acute cocaine exposure decreased the number of BrdU-immunoreactive (ir) cells in the SVZ and SGZ. In contrast, repeated cocaine exposure reduced the number of BrdU-ir cells only in the SVZ. Both acute and repeated cocaine exposure increased the number of cleaved caspase-3-, GFAP- and Iba1-ir cells in the hippocampus, and this effect was counteracted by AM630 or Rimonabant, which increased the number of BrdU-, GFAP-, and Iba1-ir cells in the hippocampus. These results indicate that the changes in neurogenic, apoptotic and gliotic processes that were produced by repeated cocaine administration were normalized by pharmacological blockade of CB1 and CB2. The restorative effects of cannabinoid receptor blockade on hippocampal cell proliferation were associated with the prevention of the induction of conditioned locomotion but not with the prevention of cocaine-induced sensitization.
Collapse
Affiliation(s)
- Eduardo Blanco-Calvo
- Departament de Pedagogia i Psicologia, Facultat de Ciències de l'Educació, Universitat de Lleida Lleida, Spain ; Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Patricia Rivera
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Sergio Arrabal
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Antonio Vargas
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Francisco Javier Pavón
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Antonia Serrano
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Estela Castilla-Ortega
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Pablo Galeano
- Instituto de Investigaciones Cardiológicas Prof. Dr. Alberto C. Taquini, Universidad de Buenos Aires-CONICET Ciudad de Buenos Aires, Argentina
| | - Leticia Rubio
- Departamento de Anatomía y Medicina Legal y Forense, Facultad de Medicina, Universidad de Málaga Málaga, Spain
| | - Juan Suárez
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| | - Fernando Rodriguez de Fonseca
- Laboratorio de Investigación-UGC de Salud Mental, Instituto de Investigación Biomédica de Málaga, Universidad de Málaga, Hospital Regional Universitario de Málaga Málaga, Spain
| |
Collapse
|
44
|
Hillard CJ, Liu QS. Endocannabinoid signaling in the etiology and treatment of major depressive illness. Curr Pharm Des 2014; 20:3795-811. [PMID: 24180398 PMCID: PMC4002665 DOI: 10.2174/13816128113196660735] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/23/2013] [Indexed: 12/28/2022]
Abstract
The purpose of this review is to examine human and preclinical data that are relevant to the following hypotheses. The first hypothesis is that deficient CB1R-mediated signaling results in symptoms that mimic those seen in depression. The second hypothesis is that activation of CB1R-mediated signaling results in behavioral, endocrine and other effects that are similar to those produced by currently used antidepressants. The third hypothesis is that conventional antidepressant therapies act through enhanced CB1R mediated signaling. Together the available data indicate that activators of CB1R signaling, particularly inhibitors of fatty acid amide hydrolase, should be considered for clinical trials for the treatment of depression.
Collapse
MESH Headings
- Animals
- Antidepressive Agents/pharmacology
- Antidepressive Agents/therapeutic use
- Brain/drug effects
- Brain/enzymology
- Brain/metabolism
- Brain/pathology
- Cannabis
- Depressive Disorder, Major/drug therapy
- Depressive Disorder, Major/etiology
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/pathology
- Disease Models, Animal
- Endocannabinoids/genetics
- Endocannabinoids/metabolism
- Humans
- Magnetic Resonance Imaging
- Neurogenesis/drug effects
- Polymorphism, Genetic
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
| | - Qing-song Liu
- Neuroscience Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226.
| |
Collapse
|
45
|
Fogaça MV, Galve-Roperh I, Guimarães FS, Campos AC. Cannabinoids, Neurogenesis and Antidepressant Drugs: Is there a Link? Curr Neuropharmacol 2013; 11:263-75. [PMID: 24179463 PMCID: PMC3648779 DOI: 10.2174/1570159x11311030003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/14/2012] [Accepted: 01/04/2013] [Indexed: 11/22/2022] Open
Abstract
Similar to clinically used antidepressants, cannabinoids can also regulate anxiety and depressive symptoms. Although the mechanisms of these effects are not completely understood, recent evidence suggests that changes in endocannabinoid system could be involved in some actions of antidepressants. Chronic antidepressant treatment modifies the expression of CB1 receptors and endocannabinoid (EC) content in brain regions related to mood and anxiety control. Moreover, both antidepressant and cannabinoids activate mitogen-activated protein (MAP) kinase and phosphoinositide 3-kinase(PI3-K)/Akt or PKB signaling, intracellular pathways that regulate cell proliferation and neural cell survival. Facilitation of hippocampal neurogenesis is proposed as a common effect of chronic antidepressant treatment. Genetic or pharmacological manipulations of cannabinoid receptors (CB1 and CB2) or enzymes responsible for endocannabinoid-metabolism have also been shown to control proliferation and neurogenesis in the hippocampus. In the present paper we reviewed the studies that have investigated the potential contribution of cannabinoids and neurogenesisto antidepressant effects. Considering the widespread brain distribution of the EC system, a better understanding of this possible interaction could contribute to the development of therapeutic alternatives to mood and anxiety disorders.
Collapse
Affiliation(s)
- Manoela Viar Fogaça
- Department of Pharmacology; School of Medicine of RibeirãoPreto- University of São Paulo, Brazil ; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, Brazil
| | | | | | | |
Collapse
|
46
|
Voss MW, Vivar C, Kramer AF, van Praag H. Bridging animal and human models of exercise-induced brain plasticity. Trends Cogn Sci 2013; 17:525-44. [PMID: 24029446 DOI: 10.1016/j.tics.2013.08.001] [Citation(s) in RCA: 631] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 12/20/2022]
Abstract
Significant progress has been made in understanding the neurobiological mechanisms through which exercise protects and restores the brain. In this feature review, we integrate animal and human research, examining physical activity effects across multiple levels of description (neurons up to inter-regional pathways). We evaluate the influence of exercise on hippocampal structure and function, addressing common themes such as spatial memory and pattern separation, brain structure and plasticity, neurotrophic factors, and vasculature. Areas of research focused more within species, such as hippocampal neurogenesis in rodents, also provide crucial insight into the protective role of physical activity. Overall, converging evidence suggests exercise benefits brain function and cognition across the mammalian lifespan, which may translate into reduced risk for Alzheimer's disease (AD) in humans.
Collapse
Affiliation(s)
- Michelle W Voss
- Department of Psychology, The University of Iowa, Iowa City, IA, USA; Aging Mind and Brain Initiative (AMBI), The University of Iowa, Iowa City, IA, USA.
| | | | | | | |
Collapse
|
47
|
Casteels C, Gérard N, van Kuyck K, Pottel L, Nuttin B, Bormans G, Van Laere K. Small animal PET imaging of the type 1 cannabinoid receptor in a rodent model for anorexia nervosa. Eur J Nucl Med Mol Imaging 2013; 41:308-21. [PMID: 24006151 DOI: 10.1007/s00259-013-2522-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 07/11/2013] [Indexed: 11/29/2022]
Abstract
PURPOSE Several lines of evidence strongly implicate a dysfunctional endocannabinoid system (ECS) in eating disorders. Using [(18)F]MK-9470 and small animal positron emission tomography (PET), we investigated for the first time cerebral changes in type 1 cannabinoid (CB1) receptor binding in vivo in the activity-based rat model of anorexia (ABA), in comparison to distinct motor- and food-related control conditions and in relation to gender and behavioural variables. METHODS In total, experiments were conducted on 80 Wistar rats (23 male and 57 female). Male rats were assigned to the cross-sectional conditions: ABA (n = 12) and CONTROL (n = 11), whereas female rats were divided between two settings: (1) a cross-sectional design using ABA (n = 13), CONTROL (n = 9), and two extra control conditions for each of the variables manipulated in ABA, i.e. DIET (n = 8) and WHEEL (n = 9), and (2) a longitudinal one using ABA (n = 10) and CONTROL (n = 8) studied at baseline, during the model and upon recovery. The ABA group was subjected to food restriction in the presence of a running wheel, the DIET group to food restriction without wheel, the WHEEL group to a normal diet with wheel and CONTROL animals had a normal diet and no running wheel. Parametric CB1 receptor images of each group were spatially normalized to Paxinos space and analysed voxel-wise. RESULTS In the ABA model, absolute [(18)F]MK-9470 binding was significantly increased in all cortical and subcortical brain areas as compared to control conditions (male +67 %; female >51%, all p cluster < 6.3×10(-6)) that normalized towards baseline values after weight gain. Additionally, relative [(18)F]MK-9470 binding was increased in the hippocampus, inferior colliculus and entorhinal cortex of female ABA (+4.6%; p cluster < 1.3×10(-6)), whereas no regional differences were observed in male subjects. Again, relative [(18)F]MK-9470 binding values normalized upon weight gain. CONCLUSION These data point to a widespread transient disturbance of the endocannabinoid transmission, specifically for CB1 receptors in the ABA model. Our data also suggest (1) gender effects on regional CB1 receptor binding in the hippocampus and (2) add further proof to the validity of the ABA model to mimic aspects of human disease.
Collapse
Affiliation(s)
- Cindy Casteels
- Division of Nuclear Medicine, University Hospital and KU Leuven, Herestraat 49 bus 7003, 3000, Leuven, Belgium,
| | | | | | | | | | | | | |
Collapse
|
48
|
Ota KT, Duman RS. Environmental and pharmacological modulations of cellular plasticity: role in the pathophysiology and treatment of depression. Neurobiol Dis 2013; 57:28-37. [PMID: 22691453 PMCID: PMC3458126 DOI: 10.1016/j.nbd.2012.05.022] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 04/05/2012] [Accepted: 05/31/2012] [Indexed: 01/09/2023] Open
Abstract
Atrophy of neurons and gross structural alterations of limbic brain regions, including the prefrontal cortex (PFC) and hippocampus, have been reported in brain imaging and postmortem studies of depressed patients. Preclinical findings have suggested that prolonged negative stress can induce changes comparable to those seen in major depressive disorder (MDD), through dendritic retraction and decreased spine density in PFC and hippocampal CA3 pyramidal neurons. Interestingly, recent studies have suggested that environmental and pharmacological manipulations, including antidepressant medication, exercise, and diet, can block or even reverse many of the molecular changes induced by stress, providing a clear link between these factors and susceptibility to MDD. In this review, we will discuss the environmental and pharmacological factors, as well as the contribution of genetic polymorphisms, involved in the regulation of neuronal morphology and plasticity in MDD and preclinical stress models. In particular, we will highlight the pro-depressive changes incurred by stress and the reversal of these changes by antidepressants, exercise, and diet.
Collapse
Affiliation(s)
- Kristie T Ota
- Division of Molecular Psychiatry, Abraham Ribicoff Research Facilities, Connecticut Mental Health Center, Yale University School of Medicine, New Haven, CT 06508, USA
| | | |
Collapse
|
49
|
Abstract
Voluntary wheel running has long been known to induce precursor cell proliferation in adult hippocampal neurogenesis in rodents. However, mechanisms that couple activity with the promitotic effect are not yet fully understood. Using tryptophan hydroxylase (TPH) 2 deficient (Tph2-deficient) mice that lack brain serotonin, we explored the relationship between serotonin signaling and exercise-induced neurogenesis. Surprisingly, Tph2-deficient mice exhibit normal baseline hippocampal neurogenesis but impaired activity-induced proliferation. Our data demonstrate that the proproliferative effect of running requires the release of central serotonin in young-adult and aged mice. Lack of brain serotonin further results in alterations at the stage of Sox2-positive precursor cells, suggesting physiological adaptations to changes in serotonin supply to maintain homeostasis in the neurogenic niche. We conclude that serotonin plays a direct and acute regulatory role in activity-dependent hippocampal neurogenesis. The understanding of exercise-induced neurogenesis might offer preventive but also therapeutic opportunities in depression and age-related cognitive decline.
Collapse
|
50
|
Xapelli S, Agasse F, Sardà-Arroyo L, Bernardino L, Santos T, Ribeiro FF, Valero J, Bragança J, Schitine C, de Melo Reis RA, Sebastião AM, Malva JO. Activation of type 1 cannabinoid receptor (CB1R) promotes neurogenesis in murine subventricular zone cell cultures. PLoS One 2013; 8:e63529. [PMID: 23704915 PMCID: PMC3660454 DOI: 10.1371/journal.pone.0063529] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Accepted: 04/06/2013] [Indexed: 11/18/2022] Open
Abstract
The endocannabinoid system has been implicated in the modulation of adult neurogenesis. Here, we describe the effect of type 1 cannabinoid receptor (CB1R) activation on self-renewal, proliferation and neuronal differentiation in mouse neonatal subventricular zone (SVZ) stem/progenitor cell cultures. Expression of CB1R was detected in SVZ-derived immature cells (Nestin-positive), neurons and astrocytes. Stimulation of the CB1R by (R)-(+)-Methanandamide (R-m-AEA) increased self-renewal of SVZ cells, as assessed by counting the number of secondary neurospheres and the number of Sox2+/+ cell pairs, an effect blocked by Notch pathway inhibition. Moreover, R-m-AEA treatment for 48 h, increased proliferation as assessed by BrdU incorporation assay, an effect mediated by activation of MAPK-ERK and AKT pathways. Surprisingly, stimulation of CB1R by R-m-AEA also promoted neuronal differentiation (without affecting glial differentiation), at 7 days, as shown by counting the number of NeuN-positive neurons in the cultures. Moreover, by monitoring intracellular calcium concentrations ([Ca2+]i) in single cells following KCl and histamine stimuli, a method that allows the functional evaluation of neuronal differentiation, we observed an increase in neuronal-like cells. This proneurogenic effect was blocked when SVZ cells were co-incubated with R-m-AEA and the CB1R antagonist AM 251, for 7 days, thus indicating that this effect involves CB1R activation. In accordance with an effect on neuronal differentiation and maturation, R-m-AEA also increased neurite growth, as evaluated by quantifying and measuring the number of MAP2-positive processes. Taken together, these results demonstrate that CB1R activation induces proliferation, self-renewal and neuronal differentiation from mouse neonatal SVZ cell cultures.
Collapse
Affiliation(s)
- Sara Xapelli
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisboa, Portugal
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisboa, Portugal
| | - Fabienne Agasse
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
- * E-mail: (JOM); (FA)
| | - Laura Sardà-Arroyo
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Liliana Bernardino
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
- Health Sciences Research Center, University of Beira Interior, Covilhã, Portugal
| | - Tiago Santos
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
| | - Filipa F. Ribeiro
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisboa, Portugal
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisboa, Portugal
| | - Jorge Valero
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
| | - José Bragança
- Institute for Biotechnology and Bioengineering, Centre for Molecular and Structural Biomedicine, University of Algarve, Faro, Portugal
| | - Clarissa Schitine
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
- Neurochemistry Laboratory, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo A. de Melo Reis
- Center for Neuroscience and Cell Biology of Coimbra, University of Coimbra, Coimbra, Portugal
- Neurochemistry Laboratory, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana M. Sebastião
- Institute of Pharmacology and Neurosciences, Faculty of Medicine, University of Lisbon, Lisboa, Portugal
- Unit of Neurosciences, Instituto de Medicina Molecular, University of Lisbon, Lisboa, Portugal
| | - João O. Malva
- Center for Research on Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine (polo 3), University of Coimbra, Coimbra, Portugal
- * E-mail: (JOM); (FA)
| |
Collapse
|