1
|
Gom RC, Wickramarachchi P, George AG, Lightfoot SHM, Newton-Gunderson D, Hill MN, Teskey GC, Colangeli R. Phytocannabinoids restore seizure-induced alterations in emotional behaviour in male rats. Neuropsychopharmacology 2024:10.1038/s41386-024-02005-y. [PMID: 39433952 DOI: 10.1038/s41386-024-02005-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/23/2024]
Abstract
Epilepsy often presents with severe emotional comorbidities including anxiety and abnormal fear responses which impose a significant burden on, and reduce, quality of life in people living with the disease. Our lab has recently shown that kindled seizures lead to changes in emotional processing resulting from the downregulation of anandamide signalling within the amygdala. Phytocannabinoids derived from the Cannabis sativa plant have attracted a lot of interest as a new class of drugs with potential anticonvulsant effects. Among the wide number of compounds occurring in Cannabis sativa, Δ9- tetrahydrocannabinol (THC), the one responsible for its main psychoactive effects, and the nonpsychoactive cannabidiol (CBD) have been extensively examined under pre-clinical and clinical contexts to control seizures, however, neither have been assessed in the context of the management of emotional comorbidities associated with seizure activity. We used two behavioural procedures to assess anxiety- and fear-like responding in adult male Long-Evans rats: elevated plus maze and auditory fear conditioning. In agreement with previous reports, we found seizure-induced increases in anxiety- and fear-like responding. These effects were reversed by either CBD (vaporized) or THC (oral). We also found that antagonism of serotonin 1 A receptors prior to CBD exposure prevented its protective effects. Phytocannabinoids offer a novel and reliable opportunity to treat seizure induced comorbid emotional alterations.
Collapse
Affiliation(s)
- Renaud C Gom
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada.
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada.
| | - Pasindu Wickramarachchi
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
| | - Antis G George
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
| | - Savannah H M Lightfoot
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Mathison Centre for Mental Health Research and Education; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Dana Newton-Gunderson
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
| | - Matthew N Hill
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
- Mathison Centre for Mental Health Research and Education; University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - G Campbell Teskey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
| | - Roberto Colangeli
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Cell Biology and Anatomy, University of Calgary, Calgary, Calgary, AB, Canada
- Department of Experimental and Clinical Medicine; Università Politecnica delle Marche, 60126, Ancona, Italy
| |
Collapse
|
2
|
Gao JH, Liu YY, Xu HX, Wu K, Zhang LL, Cheng P, Peng XH, Cao JL, Hua R, Zhang YM. Divergent input patterns to the central lateral amygdala play a duet in fear memory formation. iScience 2024; 27:110886. [PMID: 39319272 PMCID: PMC11421289 DOI: 10.1016/j.isci.2024.110886] [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: 01/15/2024] [Revised: 04/18/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
Somatostatin (SOM)-expressing neurons in the central lateral amygdala (CeL) are responsible for fear memory learning, but the circuit and molecular mechanisms underlying this biology remain elusive. Here, we found that glutamatergic neurons in the lateral parabrachial nucleus (LPB) directly dominated the activity of CeLSOM neurons, and that selectively inhibiting the LPBGlu→CeLSOM pathway suppressed fear memory acquisition. By contrast, inhibiting CeL-projecting glutamatergic neurons in the paraventricular thalamic nucleus (PVT) interfered with consolidation-related processes. Notably, CeLSOM-innervating neurons in the LPB were modulated by presynaptic cannabinoid receptor 1 (CB1R), and knock down of CB1Rs in LPB glutamatergic neurons enhanced excitatory transmission to the CeL and partially rescued the impairment in fear memory induced by CB1R activation in the CeL. Overall, our study reveals the mechanisms by which CeLSOM neurons mediate the formation of fear memories during fear conditioning in mice, which may provide a new direction for the clinical research of fear-related disorders.
Collapse
Affiliation(s)
- Jing-Hua Gao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Department of Anesthesiology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng 224008, Jiangsu, China
| | - Yue-Ying Liu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Hui-Xiang Xu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Ke Wu
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Le-le Zhang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Peng Cheng
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Xiao-Han Peng
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Jun-Li Cao
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| | - Rong Hua
- Department of Emergency, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yong-Mei Zhang
- NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| |
Collapse
|
3
|
Zabik NL, Iadipaolo A, Peters CA, Baglot SL, Hill MN, Rabinak CA. Dose-dependent effect of acute THC on extinction memory recall and fear renewal: a randomized, double-blind, placebo-controlled study. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06702-w. [PMID: 39412674 DOI: 10.1007/s00213-024-06702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/03/2024] [Indexed: 11/06/2024]
Abstract
RATIONALE Prior work from our lab and others demonstrates that the endocannabinoid system is a promising avenue for improving fear memory deficits in posttraumatic stress disorder (PTSD). Specifically, 7.5 mg of delta-9-tetrahydrocannabinol (THC) decreases fear responding in healthy adults and increases prefrontal cortex activation during extinction learning and fear renewal in adults with PTSD. OBJECTIVES The present study will determine whether there is a dose-dependent effect of THC on short-term (24 h) and long-term (one week) fear learning and memory in adults with PTSD. METHODS Using a randomized, double-blind, placebo-controlled design, N = 36 adults with PTSD completed the study and were randomized to receive placebo (PBO, n = 11), 5 mg of THC (n = 11), or 10 mg of THC (n = 14) prior to fear extinction learning. Participants completed a Pavlovian conditioning paradigm with extinction recall and fear renewal occurring 24 h and one week later, where we measured concurrent functional imaging and behavioral responses. RESULTS Twenty-four hours after drug administration, individuals with PTSD given 5 mg of THC exhibited greater anterior cingulate cortex and prefrontal cortex activation during early fear renewal. One week later, individuals given 10 mg of THC exhibited greater hippocampus activation during extinction recall and prefrontal cortex activation during fear renewal. CONCLUSIONS These data suggest that dosing and timing are critical for facilitating fear memory processes in PTSD, and that low-dose oral THC prior to extinction learning can affect brain indices of fear learning and memory both acutely and one week after administration.
Collapse
Affiliation(s)
- Nicole L Zabik
- Translational Neuroscience Program, Wayne State University School of Medicine, Tolan Park Medical Building, Detroit, MI, 48201, USA
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Tolan Park Medical Building, Detroit, MI, 48201, USA
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Allesandra Iadipaolo
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Craig A Peters
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA
| | - Samantha L Baglot
- Department of Cell Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
- Department of Anatomy & Psychiatry, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Matthew N Hill
- Department of Cell Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
- Department of Anatomy & Psychiatry, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N4N1, Canada
| | - Christine A Rabinak
- Translational Neuroscience Program, Wayne State University School of Medicine, Tolan Park Medical Building, Detroit, MI, 48201, USA.
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Tolan Park Medical Building, Detroit, MI, 48201, USA.
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA.
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, 48201, USA.
| |
Collapse
|
4
|
Iqbal J, Huang GD, Xue YX, Yang M, Jia XJ. The neural circuits and molecular mechanisms underlying fear dysregulation in posttraumatic stress disorder. Front Neurosci 2023; 17:1281401. [PMID: 38116070 PMCID: PMC10728304 DOI: 10.3389/fnins.2023.1281401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 10/13/2023] [Indexed: 12/21/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a stress-associated complex and debilitating psychiatric disorder due to an imbalance of neurotransmitters in response to traumatic events or fear. PTSD is characterized by re-experiencing, avoidance behavior, hyperarousal, negative emotions, insomnia, personality changes, and memory problems following exposure to severe trauma. However, the biological mechanisms and symptomatology underlying this disorder are still largely unknown or poorly understood. Considerable evidence shows that PTSD results from a dysfunction in highly conserved brain systems involved in regulating stress, anxiety, fear, and reward circuitry. This review provides a contemporary update about PTSD, including new data from the clinical and preclinical literature on stress, PTSD, and fear memory consolidation and extinction processes. First, we present an overview of well-established laboratory models of PTSD and discuss their clinical translational value for finding various treatments for PTSD. We then highlight the research progress on the neural circuits of fear and extinction-related behavior, including the prefrontal cortex, hippocampus, and amygdala. We further describe different molecular mechanisms, including GABAergic, glutamatergic, cholinergic, and neurotropic signaling, responsible for the structural and functional changes during fear acquisition and fear extinction processes in PTSD.
Collapse
Affiliation(s)
- Javed Iqbal
- Shenzhen Graduate School, Peking University Shenzhen, Guangdong, China
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital and Shenzhen Mental Health Center; Clinical College of Mental Health, Shenzhen University Health Science Center; Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Geng-Di Huang
- Shenzhen Graduate School, Peking University Shenzhen, Guangdong, China
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital and Shenzhen Mental Health Center; Clinical College of Mental Health, Shenzhen University Health Science Center; Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yan-Xue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Mei Yang
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital and Shenzhen Mental Health Center; Clinical College of Mental Health, Shenzhen University Health Science Center; Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Xiao-Jian Jia
- Department of Addiction Medicine, Shenzhen Engineering Research Center for Precision Psychiatric Technology, Shenzhen Clinical Research Center for Mental Disorders, Shenzhen Kangning Hospital and Shenzhen Mental Health Center; Clinical College of Mental Health, Shenzhen University Health Science Center; Affiliated Mental Health Center, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
5
|
Gunduz-Cinar O, Castillo LI, Xia M, Van Leer E, Brockway ET, Pollack GA, Yasmin F, Bukalo O, Limoges A, Oreizi-Esfahani S, Kondev V, Báldi R, Dong A, Harvey-White J, Cinar R, Kunos G, Li Y, Zweifel LS, Patel S, Holmes A. A cortico-amygdala neural substrate for endocannabinoid modulation of fear extinction. Neuron 2023; 111:3053-3067.e10. [PMID: 37480845 PMCID: PMC10592324 DOI: 10.1016/j.neuron.2023.06.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 07/24/2023]
Abstract
Preclinical and clinical studies implicate endocannabinoids (eCBs) in fear extinction, but the underlying neural circuit basis of these actions is unclear. Here, we employed in vivo optogenetics, eCB biosensor imaging, ex vivo electrophysiology, and CRISPR-Cas9 gene editing in mice to examine whether basolateral amygdala (BLA)-projecting medial prefrontal cortex (mPFC) neurons represent a neural substrate for the effects of eCBs on extinction. We found that photoexcitation of mPFC axons in BLA during extinction mobilizes BLA eCBs. eCB biosensor imaging showed that eCBs exhibit a dynamic stimulus-specific pattern of activity at mPFC→BLA neurons that tracks extinction learning. Furthermore, using CRISPR-Cas9-mediated gene editing, we demonstrated that extinction memory formation involves eCB activity at cannabinoid CB1 receptors expressed at vmPFC→BLA synapses. Our findings reveal the temporal characteristics and a neural circuit basis of eCBs' effects on fear extinction and inform efforts to target the eCB system as a therapeutic approach in extinction-deficient neuropsychiatric disorders.
Collapse
Affiliation(s)
- Ozge Gunduz-Cinar
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA.
| | - Laura I Castillo
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Maya Xia
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Elise Van Leer
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Emma T Brockway
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Gabrielle A Pollack
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Farhana Yasmin
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olena Bukalo
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Aaron Limoges
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Sarvar Oreizi-Esfahani
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Veronika Kondev
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37232, USA
| | - Rita Báldi
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ao Dong
- Peking University School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Judy Harvey-White
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Resat Cinar
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA; Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA
| | - Yulong Li
- Peking University School of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Larry S Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Sachin Patel
- Northwestern Center for Psychiatric Neuroscience, Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
6
|
Li Y, Zhi W, Qi B, Wang L, Hu X. Update on neurobiological mechanisms of fear: illuminating the direction of mechanism exploration and treatment development of trauma and fear-related disorders. Front Behav Neurosci 2023; 17:1216524. [PMID: 37600761 PMCID: PMC10433239 DOI: 10.3389/fnbeh.2023.1216524] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Fear refers to an adaptive response in the face of danger, and the formed fear memory acts as a warning when the individual faces a dangerous situation again, which is of great significance to the survival of humans and animals. Excessive fear response caused by abnormal fear memory can lead to neuropsychiatric disorders. Fear memory has been studied for a long time, which is of a certain guiding effect on the treatment of fear-related disorders. With continuous technological innovations, the study of fear has gradually shifted from the level of brain regions to deeper neural (micro) circuits between brain regions and even within single brain regions, as well as molecular mechanisms. This article briefly outlines the basic knowledge of fear memory and reviews the neurobiological mechanisms of fear extinction and relapse, which aims to provide new insights for future basic research on fear emotions and new ideas for treating trauma and fear-related disorders.
Collapse
Affiliation(s)
- Ying Li
- College of Education, Hebei University, Baoding, China
- Laboratory of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Weijia Zhi
- Laboratory of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Bing Qi
- College of Education, Hebei University, Baoding, China
| | - Lifeng Wang
- Laboratory of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| | - Xiangjun Hu
- College of Education, Hebei University, Baoding, China
- Laboratory of Experimental Pathology, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
7
|
The Effects of the Inhalant Toluene on Cognitive Function and Behavioral Flexibility: A Review of Recent Findings. ADDICTION NEUROSCIENCE 2023; 5:100059. [PMID: 36798693 PMCID: PMC9928149 DOI: 10.1016/j.addicn.2022.100059] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Substance use disorder (SUD) is characterized, in part, by lack of control over drug seeking and taking. The prefrontal cortex (PFC) is highly involved in control of behavior and deficits in PFC structure and function have been demonstrated in clinical and preclinical studies of SUD. Of the various classes of drugs associated with the development of SUD, inhalants are among the least studied despite their widespread use among adolescents and children. In this work, we review what is currently known regarding the sites and mechanisms of action of inhalants with a focus on the volatile solvent toluene that is contained in a wide variety of legal and easily obtained products. We then describe how inhalants including toluene affect various behaviors with an emphasis on those associated with PFC function and how chronic use of inhalants alters brain structure and neuronal signaling. Findings from these studies highlight advances made in recent years that have expanded our understanding of the effects of inhalants on brain structure and reinforce the need for continued work in this field.
Collapse
|
8
|
Niloy N, Hediyal TA, Vichitra C, Sonali S, Chidambaram SB, Gorantla VR, Mahalakshmi AM. Effect of Cannabis on Memory Consolidation, Learning and Retrieval and Its Current Legal Status in India: A Review. Biomolecules 2023; 13:biom13010162. [PMID: 36671547 PMCID: PMC9855787 DOI: 10.3390/biom13010162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/31/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Cannabis is one of the oldest crops grown, traditionally held religious attachments in various cultures for its medicinal use much before its introduction to Western medicine. Multiple preclinical and clinical investigations have explored the beneficial effects of cannabis in various neurocognitive and neurodegenerative diseases affecting the cognitive domains. Tetrahydrocannabinol (THC), the major psychoactive component, is responsible for cognition-related deficits, while cannabidiol (CBD), a non-psychoactive phytocannabinoid, has been shown to elicit neuroprotective activity. In the present integrative review, the authors focus on the effects of cannabis on the different cognitive domains, including learning, consolidation, and retrieval. The present study is the first attempt in which significant focus has been imparted on all three aspects of cognition, thus linking to its usage. Furthermore, the investigators have also depicted the current legal position of cannabis in India and the requirement for reforms.
Collapse
Affiliation(s)
- Nandi Niloy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Tousif Ahmed Hediyal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Chandrasekaran Vichitra
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Sharma Sonali
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
| | - Vasavi Rakesh Gorantla
- Department of Anatomical Science, St. George’s University, University Centre, St. Georges FZ818, Grenada
- Correspondence: (V.R.G.); (A.M.M.)
| | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, SS Nagar, Mysore 570015, Karnataka, India
- Correspondence: (V.R.G.); (A.M.M.)
| |
Collapse
|
9
|
Kumar Kalvala A, Bagde A, Arthur P, Kumar Surapaneni S, Ramesh N, Nathani A, Singh M. Role of Cannabidiol and Tetrahydrocannabivarin on Paclitaxel-induced neuropathic pain in rodents. Int Immunopharmacol 2022; 107:108693. [PMID: 35303507 PMCID: PMC10791145 DOI: 10.1016/j.intimp.2022.108693] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/26/2022] [Accepted: 03/07/2022] [Indexed: 01/13/2023]
Abstract
The purpose of this study was to evaluate if phytocannabinoids, synthetic cannabidiol (CBD), and tetrahydrocannabivarin (THCV), and their combination, could protect mice from Paclitaxel-induced peripheral neuropathy (PIPN). Six groups of C57BL/6J mice (n = 6) were used in this study. The mice were given paclitaxel (PTX) (8 mg/kg/day, i.p.) on days 1, 3, 5, and 7 to induce neuropathy. Mice were evaluated for behavioral parameters, and dorsal root ganglions (DRG) were collected from the animals and subjected to RNA sequencing and westernblot analysis at the end of the study. On cultured DRGs derived from adult male rats, immunocytochemistry and mitochondrial functional assays were also performed. When compared to individual treatments, the combination of CBD and THCV improved thermal and mechanical neurobehavioral symptoms in mice by twofold. Targets for CBD and THCV therapy were identified by KEGG (RNA sequencing). PTX reduced the expression of p-AMPK, SIRT1, NRF2, HO1, SOD2, and catalase while increasing the expression of PI3K, p-AKT, p-P38 MAP kinase, BAX, TGF-β, NLRP3 inflammasome, and caspase 3 in DRG homogenates of mice. Combination therapy outperformed monotherapy in reversing these protein expressions. The addition of CBD and THCV to DRG primary cultures reduced mitochondrial superoxides while increasing mitochondrial membrane potentials. WAY100135 and rimonabant altered the neuroprotective effects of CBD and THCV respectively by blocking 5-HT1A and CB1 receptors in mice and DRG primary cultures. The entourage effect of CBD and THCV against PIPN appears to protect neurons in mice via 5HT1A and CB1 receptors respectively.
Collapse
Affiliation(s)
- Anil Kumar Kalvala
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Arvind Bagde
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Peggy Arthur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Sunil Kumar Surapaneni
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Nimma Ramesh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, 32307, USA.
| |
Collapse
|
10
|
Braunscheidel KM, Okas MP, Floresco SB, Woodward JJ. Cannabinoid receptor type 1 antagonists alter aspects of risk/reward decision making independent of toluene-mediated effects. Psychopharmacology (Berl) 2022; 239:1337-1347. [PMID: 34291308 PMCID: PMC9885490 DOI: 10.1007/s00213-021-05914-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/25/2021] [Indexed: 02/02/2023]
Abstract
Drugs of abuse including cannabis and inhalants impair risk/reward decision making. Cannabis use is often concurrent with inhalant intoxication; yet, preclinical studies investigating the role of endocannabinoids in inhalant misuse are limited. To address this gap in the literature, we used the well-validated probabilistic discounting task to assess risk/reward decision making in rodents following combinations of toluene vapor (a common inhalant) and manipulations of cannabinoid receptor type 1 (CB1R) signaling. As reported previously, acute exposure to toluene vapor disrupted behavioral flexibility during probabilistic discounting. Systemic administration of the CB1R inverse agonist AM281 did not prevent toluene-induced alterations in risky choices, but did independently reduce win-stay behavior, increase choice latency, and increase omissions. Toluene-induced deficits in probabilistic discounting are thought to involve impaired medial prefrontal cortex (mPFC) activity. As we previously reported that some of toluene's inhibitory effects on glutamatergic signaling in the mPFC are endocannabinoid-dependent, we tested the hypothesis that mPFC CB1R activity mediates toluene-induced deficits in discounting. However, bilateral injection of the CB1R inverse agonist AM251 prior to toluene vapor exposure had no effect on toluene-induced changes in risk behavior. In a final set of experiments, we injected the CB1R inverse agonist AM251 (5 and 50 ng), the CB1R agonist WIN55,212-2 (50 ng and 500 ng), or vehicle into the mPFC prior to testing. While mPFC CB1R stimulation did not affect any of the measures tested, the CB1R inverse agonist caused a dose-dependent reduction in win-stay behavior without altering any other measures. Together, these studies indicate that toluene-induced deficits in probabilistic discounting are largely distinct from CB1R-dependent effects that include decreased effectiveness of positive reinforcement (mPFC CB1Rs), decision making speed, and task engagement (non-mPFC CB1Rs).
Collapse
Affiliation(s)
- Kevin M Braunscheidel
- Department of Neuroscience, Medical University of South Carolina, MSC 861, 30 Courtenay Drive, Charleston, SC, 29425-5712, USA
| | - Michael P Okas
- Department of Neuroscience, Medical University of South Carolina, MSC 861, 30 Courtenay Drive, Charleston, SC, 29425-5712, USA
| | - Stan B Floresco
- Department of Psychology and Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Colombia, V6T 1Z4, Canada
| | - John J Woodward
- Department of Neuroscience, Medical University of South Carolina, MSC 861, 30 Courtenay Drive, Charleston, SC, 29425-5712, USA.
| |
Collapse
|
11
|
Higginbotham JA, Jones NM, Wang R, Christian RJ, Ritchie JL, McLaughlin RJ, Fuchs RA. Basolateral amygdala CB1 receptors gate HPA axis activation and context-cocaine memory strength during reconsolidation. Neuropsychopharmacology 2021; 46:1554-1564. [PMID: 33452429 PMCID: PMC8280224 DOI: 10.1038/s41386-020-00919-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/24/2020] [Accepted: 11/15/2020] [Indexed: 11/09/2022]
Abstract
Re-exposure to a cocaine-associated context triggers craving and relapse through the retrieval of salient context-drug memories. Upon retrieval, context-drug memories become labile and temporarily sensitive to modification before they are reconsolidated into long-term memory stores. The effects of systemic cannabinoid type 1 receptor (CB1R) antagonism indicate that CB1R signaling is necessary for cocaine-memory reconsolidation and associated glutamatergic plasticity in the basolateral amygdala (BLA); however, the contribution of BLA CB1R signaling to cocaine-memory reconsolidation is unknown. Here, we assessed whether intra-BLA CB1R manipulations immediately after cocaine-memory retrieval alter cocaine-memory strength indexed by subsequent drug context-induced cocaine-seeking behavior in an instrumental rodent model of drug relapse. Administration of the CB1R antagonist, AM251 (0.3 µg/hemisphere) into the BLA increased subsequent drug context-induced cocaine-seeking behavior in a memory retrieval-dependent and anatomically selective manner. Conversely, the CB1R agonist, WIN55,212-2 (0.5 or 5 µg/hemisphere) failed to alter this behavior. In follow-up experiments, cocaine-memory retrieval elicited robust hypothalamic-pituitary-adrenal axis activation, as indicated by a rise in serum corticosterone concentrations. Intra-BLA AM251 administration during memory reconsolidation selectively increased this cocaine-memory retrieval-induced corticosterone response. Intra-BLA corticosterone administration (3 or 10 ng/hemisphere) during memory reconsolidation did not augment subsequent cocaine-seeking behavior, suggesting that CB1R-dependent effects of corticosterone on memory strength, if any, are mediated outside of the BLA. Together, these findings suggest that CB1R signaling in the BLA gates cocaine-memory strength, possibly by diminishing the impact of cue-induced arousal on the integrity of the reconsolidating memory trace or on the efficacy of the memory reconsolidation process.
Collapse
Affiliation(s)
- Jessica A. Higginbotham
- grid.30064.310000 0001 2157 6568Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA USA
| | - Nicole M. Jones
- grid.30064.310000 0001 2157 6568Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA USA
| | - Rong Wang
- grid.30064.310000 0001 2157 6568Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA USA
| | - Robert J. Christian
- grid.30064.310000 0001 2157 6568Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA USA
| | - Jobe L. Ritchie
- grid.30064.310000 0001 2157 6568Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA USA
| | - Ryan J. McLaughlin
- grid.30064.310000 0001 2157 6568Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA USA ,grid.30064.310000 0001 2157 6568Washington State University Alcohol and Drug Abuse Research Program, Pullman, WA USA ,grid.30064.310000 0001 2157 6568Translational Addiction Research Collaborative, Washington State University, Pullman, WA USA
| | - Rita A. Fuchs
- grid.30064.310000 0001 2157 6568Department of Integrative Physiology and Neuroscience, Washington State University College of Veterinary Medicine, Pullman, WA USA ,grid.30064.310000 0001 2157 6568Washington State University Alcohol and Drug Abuse Research Program, Pullman, WA USA ,grid.30064.310000 0001 2157 6568Translational Addiction Research Collaborative, Washington State University, Pullman, WA USA
| |
Collapse
|
12
|
Mizuno I, Matsuda S. The role of endocannabinoids in consolidation, retrieval, reconsolidation, and extinction of fear memory. Pharmacol Rep 2021; 73:984-1003. [PMID: 33954935 DOI: 10.1007/s43440-021-00246-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/26/2021] [Accepted: 03/07/2021] [Indexed: 12/19/2022]
Abstract
Endocannabinoids are involved in various physiological functions, including synaptic plasticity and memory, and some psychiatric disorders, such as posttraumatic stress disorder (PTSD), through the activation of cannabinoid (CB) receptors. Patients with PTSD often show excessive fear memory and impairment of fear extinction (FE). It has been reported that the stability of acquired fear memory is altered through multiple memory stages, such as consolidation and reconsolidation. FE also affects the stability of fear memory. Each stage of fear memory formation and FE are regulated by different molecular mechanisms, including the CB system. However, to the best of our knowledge, no review summarizes the role of the CB system during each stage of fear memory formation and FE. In this review, we summarize the roles of endocannabinoids in fear memory formation and FE. Moreover, based on the summary, we propose a new hypothesis for the role of endocannabinoids in fear regulation, and discuss treatment for PTSD using CB system-related drugs.
Collapse
Affiliation(s)
- Ikumi Mizuno
- Department of Pharmacotherapeutics, Showa Pharmaceutical University, 3-3165, Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan
| | - Shingo Matsuda
- Department of Pharmacotherapeutics, Showa Pharmaceutical University, 3-3165, Higashi-Tamagawagakuen, Machida, Tokyo, 194-8543, Japan. .,Department of Cognitive Behavioral Physiology, Chiba University Graduate School of Medicine, 1-8-1 Inohana, Chiba, Chiba, 260-8670, Japan. .,Department of Ultrastructural Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawahigashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
13
|
Warren WG, Papagianni EP, Stevenson CW, Stubbendorff C. In it together? The case for endocannabinoid-noradrenergic interactions in fear extinction. Eur J Neurosci 2021; 55:952-970. [PMID: 33759226 DOI: 10.1111/ejn.15200] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 01/26/2021] [Accepted: 03/17/2021] [Indexed: 12/18/2022]
Abstract
Anxiety and trauma-related disorders, such as post-traumatic stress disorder (PTSD), are debilitating mental illnesses with great personal and socioeconomic costs. Examining memory formation and relevant behavioural responding associated with aversive stimuli may improve our understanding of the neurobiology underlying fear memory processing and PTSD treatment. The neurocircuitry underpinning learned fear and its inhibition through extinction is complex, involving synergistic interactions between different neurotransmitter systems in inter-connected brain areas. Endocannabinoid and noradrenergic transmission have both been implicated separately in fear memory processing and PTSD, but potential interactions between these systems in relation to fear extinction have received little attention to date. Their receptors are expressed together in brain areas crucial for fear extinction, which is enhanced by both cannabinoid and noradrenergic receptor activation in these areas. Moreover, cannabinoid signalling modulates the activity of locus coeruleus noradrenaline (NA) neurons and the release of NA in the medial prefrontal cortex, a brain area that is crucial for fear extinction. Interestingly, endocannabinoid-noradrenergic system interactions have been shown to regulate the encoding and retrieval of fear memory. Thus, noradrenergic regulation of fear extinction may also be driven indirectly in part via cannabinoid receptor signalling. In this perspective paper, we collate the available relevant literature and propose a synergistic role for the endocannabinoid and noradrenergic systems in regulating fear extinction, the study of which may further our understanding of the neurobiological substrates of PTSD and its treatment.
Collapse
Affiliation(s)
- William G Warren
- School of Biosciences, University of Nottingham, Loughborough, UK
| | | | - Carl W Stevenson
- School of Biosciences, University of Nottingham, Loughborough, UK
| | - Christine Stubbendorff
- School of Biosciences, University of Nottingham, Loughborough, UK.,Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
14
|
Gidyk DC, Diwan M, Gouveia FV, Giacobbe P, Lipsman N, Hamani C. Investigating the role of CB1 endocannabinoid transmission in the anti-fear and anxiolytic-like effects of ventromedial prefrontal cortex deep brain stimulation. J Psychiatr Res 2021; 135:264-269. [PMID: 33513472 DOI: 10.1016/j.jpsychires.2021.01.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 01/16/2021] [Accepted: 01/18/2021] [Indexed: 10/22/2022]
Abstract
Deep brain stimulation (DBS) delivered to the ventromedial prefrontal cortex (vmPFC) of rats induces anti-fear and anxiolytic-like behaviours, while reducing principal cell firing in the basolateral amygdala (BLA). In parallel, the endocannabinoid system, particularly in the vmPFC and BLA, has emerged as a target for the amelioration of fear and stress-related behaviours. We tested whether DBS-related improvements in fear and anxiety-type behaviour are mediated by endocannabinoid signalling. First, we examined type-1 cannabinoid (CB1) receptor and fatty acid amide hydrolase (FAAH) expression in the vmPFC and BLA and found reduced CB1 expression in both loci in rats treated with DBS. Next, we conducted pharmacological experiments to test whether the inverse CB1 agonist AM251 could mitigate the behavioural effects of stimulation. Chronic vmPFC DBS was delivered to rats following conditioning and extinction. Animals were then tested for extinction recall and anxiety-type behaviour following the systemic administration of AM251 or vehicle. We found that DBS reduced freezing and induced anxiolytic-type effects in defensive burying and novelty supressed feeding paradigms. These responses were not countered by CB1 antagonism, suggesting that other mechanisms may be involved in the anti-fear and anxiolytic effects of DBS.
Collapse
Affiliation(s)
- Darryl C Gidyk
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Mustansir Diwan
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Flavia Venetucci Gouveia
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Peter Giacobbe
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Clement Hamani
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada.
| |
Collapse
|
15
|
Inhibitory neurotransmission drives endocannabinoid degradation to promote memory consolidation. Nat Commun 2020; 11:6407. [PMID: 33335094 PMCID: PMC7747732 DOI: 10.1038/s41467-020-20121-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/12/2020] [Indexed: 11/08/2022] Open
Abstract
Endocannabinoids retrogradely regulate synaptic transmission and their abundance is controlled by the fine balance between endocannabinoid synthesis and degradation. While the common assumption is that “on-demand” release determines endocannabinoid signaling, their rapid degradation is expected to control the temporal profile of endocannabinoid action and may impact neuronal signaling. Here we show that memory formation through fear conditioning selectively accelerates the degradation of endocannabinoids in the cerebellum. Learning induced a lasting increase in GABA release and this was responsible for driving the change in endocannabinoid degradation. Conversely, Gq-DREADD activation of cerebellar Purkinje cells enhanced endocannabinoid signaling and impaired memory consolidation. Our findings identify a previously unappreciated reciprocal interaction between GABA and the endocannabinoid system in which GABA signaling accelerates endocannabinoid degradation, and triggers a form of learning-induced metaplasticity. Endocannabinoid levels are controlled by the fine balance between their synthesis and degradation. Here, the authors show that memory formation through fear conditioning selectively accelerates the degradation of endocannabinoids in the cerebellum via a lasting increase in GABA release.
Collapse
|
16
|
Lunardi P, de Souza LW, dos Santos B, Popik B, de Oliveira Alvares L. Effect of the Endocannabinoid System in Memory Updating and Forgetting. Neuroscience 2020; 444:33-42. [DOI: 10.1016/j.neuroscience.2020.07.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/31/2022]
|
17
|
Developmental differences in the effects of CB1/2R agonist WIN55212-2 on extinction of learned fear. Prog Neuropsychopharmacol Biol Psychiatry 2020; 99:109834. [PMID: 31830508 DOI: 10.1016/j.pnpbp.2019.109834] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/18/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
Abstract
Adolescence is characterised by substantial changes in emotion regulation and, in particular, impaired extinction consolidation and retention. In this study, we replicated the well-established finding that increasing the activation of cannabinoid receptor 1 (CB1R) via the agonist WIN55212-2 improves fear extinction in adult rodents before examining whether this adjunct would also rescue the extinction retention deficit seen in adolescent rodents. Contrary to the effects in adults, we found that WIN55212-2 impaired within-session acquisition of extinction in adolescent rats with no effect on extinction retention. The same effects of WIN55212-2 were observed for juvenile rats, and did not vary as a function of drug dose. Increased fear expression observed during extinction training was not a result of altered locomotor or anxiety-like behaviour in adolescent rats, as assessed by the open field test. Lastly, we observed a linear decrease in CB1R protein expression across age (i.e., from juveniles, to adolescents, and adults) in both the medial prefrontal cortex and amygdala, two regions implicated in fear expression and extinction, suggesting that there is continued refinement of the endocannabinoid system across development in two regions involved in extinction. Our findings suggest that the expression and extinction of fear in developing rats is differentially affected by CB1R agonism due to an immature endocannabinoid system.
Collapse
|
18
|
Vimalanathan A, Gidyk DC, Diwan M, Gouveia FV, Lipsman N, Giacobbe P, Nobrega JN, Hamani C. Endocannabinoid modulating drugs improve anxiety but not the expression of conditioned fear in a rodent model of post-traumatic stress disorder. Neuropharmacology 2020; 166:107965. [PMID: 31962287 DOI: 10.1016/j.neuropharm.2020.107965] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 01/07/2020] [Accepted: 01/12/2020] [Indexed: 12/23/2022]
Abstract
The endocannabinoid (eCB) system is a potential target for the treatment of symptoms of post-traumatic stress disorder (PTSD). Similar to clinical PTSD, approximately 25-30% of rats that undergo cued fear conditioning exhibit impaired extinction learning. In addition to extinction-resistant fear, these "weak extinction" (WE) rats show persistent anxiety-like behaviors. The goal of the present study was to test the hypothesis that behavioural differences between WE animals and those presenting normal extinction patterns (strong extinction; SE) could be mediated by the eCB system. Rats undergoing fear conditioning/extinction and fear recall sessions were initially segregated in weak and strong-extinction groups. Two weeks later, animals underwent a fear recall session followed by a novelty-suppressed feeding (NSF) test. In acute experiments, WE rats were injected with either the fatty acid amide hydrolase (FAAH) inhibitor URB597 or the CB1 agonist WIN55,212-2 1 h prior to long-term recall and NSF testing. SE animals were injected with the inverse CB1 receptor agonist AM251. In chronic experiments, WE and SE rats were given daily injections of URB597 or AM251 between short and long-term recall sessions. We found that acute administration of WIN55,212-2 but not URB597 reduced anxiety-like behaviour in WE rats. In contrast, AM251 was anxiogenic in SE animals. Neither treatment was effective in altering freezing expression during fear recall. The chronic administration of AM251 to SE or URB597 to WE did not alter fear or anxiety-like behaviour or changed the expression of FAAH and CB1. Together, these results suggest that systemic manipulations of the eCB system may alter anxiety-like behaviour but not the behavioural expression of an extinction-resistant associative fear memory.
Collapse
Affiliation(s)
- Akshayan Vimalanathan
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada; Behavioural Neurobiology Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Darryl C Gidyk
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Mustansir Diwan
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Flavia V Gouveia
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Nir Lipsman
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - Peter Giacobbe
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada
| | - José N Nobrega
- Behavioural Neurobiology Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada
| | - Clement Hamani
- Harquail Centre for Neuromodulation, Sunnybrook Health Sciences Centre, Sunnybrook Research Institute, Toronto, ON, M4N 3M5, Canada; Behavioural Neurobiology Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, 250 College Street, Toronto, ON, M5T 1R8, Canada.
| |
Collapse
|
19
|
Maymon N, Mizrachi Zer-Aviv T, Sabban EL, Akirav I. Neuropeptide Y and cannabinoids interaction in the amygdala after exposure to shock and reminders model of PTSD. Neuropharmacology 2019; 162:107804. [PMID: 31622603 DOI: 10.1016/j.neuropharm.2019.107804] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 12/14/2022]
Abstract
Modulation of cannabinoid and neuropeptide Y (NPY) receptors may offer therapeutic benefits for post-traumatic stress disorder (PTSD). In this study, we aimed to investigate the functional interaction between these systems in the basolateral amygdala (BLA) in a rat model of PTSD. Rats were exposed to the shock and reminders model of PTSD and tested for hyper arousal/PTSD- and depression-like behaviors 3 weeks later. Immediately after shock exposure rats were microinjected into the BLA with URB597, a selective inhibitor of fatty acid amide hydrolase (FAAH) that increases the levels of the endocannabinoid anandamide or with the NPY1 receptor agonist Leu31,Pro34-NPY (Leu). Intra-BLA URB597 prevented the shock/reminders-induced PTSD- behaviors (extinction, startle) and depression-behaviors (despair, social impairments). These preventing effects of URB597 on PTSD- and depression-like behaviors were shown to be mostly mediated by cannabinoid CB1 and NPY1 receptors, as they were blocked when URB597 was co-administered with a low dose of a CB1 or NPY1 receptor antagonist. Similarly, intra-BLA Leu prevented development of all the behaviors. Interestingly, a CB1 antagonist prevented the effects of Leu on despair and social behavior, but not the effects on extinction and startle. Moreover, exposure to shock and reminders upregulated CB1 and NPY1 receptors in the BLA and infralimbic prefrontal cortex and this upregulation was restored to normal with intra-BLA URB597 or Leu. The findings suggest that the functional interaction between the eCB and NPY1 systems is complex and provide a rationale for exploring novel therapeutic strategies that target the cannabinoid and NPY systems for stress-related diseases.
Collapse
Affiliation(s)
- Neta Maymon
- Department of Psychology, University of Haifa, Haifa, 3498838, Israel
| | | | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College Valhalla, New York, 10595, USA
| | - Irit Akirav
- Department of Psychology, University of Haifa, Haifa, 3498838, Israel.
| |
Collapse
|
20
|
Abizaid A, Merali Z, Anisman H. Cannabis: A potential efficacious intervention for PTSD or simply snake oil? J Psychiatry Neurosci 2019; 44:75-78. [PMID: 30810022 PMCID: PMC6397040 DOI: 10.1503/jpn.190021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Alfonso Abizaid
- From the Department of Neuroscience, Carleton University (Abizaid, Anisman); and the Royal’s Institute of Mental Health (Merali), Ottawa, Ont., Canada
| | - Zul Merali
- From the Department of Neuroscience, Carleton University (Abizaid, Anisman); and the Royal’s Institute of Mental Health (Merali), Ottawa, Ont., Canada
| | - Hymie Anisman
- From the Department of Neuroscience, Carleton University (Abizaid, Anisman); and the Royal’s Institute of Mental Health (Merali), Ottawa, Ont., Canada
| |
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Anxiety- and trauma-related disorders are prevalent and debilitating mental illnesses associated with a significant socioeconomic burden. Current treatment approaches often have inadequate therapeutic responses, leading to symptom relapse. Here we review recent preclinical and clinical findings on the potential of cannabinoids as novel therapeutics for regulating fear and anxiety. RECENT FINDINGS Evidence from preclinical studies has shown that the non-psychotropic phytocannabinoid cannabidiol and the endocannabinoid anandamide have acute anxiolytic effects and also regulate learned fear by dampening its expression, enhancing its extinction and disrupting its reconsolidation. The findings from the relevant clinical literature are still very preliminary but are nonetheless encouraging. Based on this preclinical evidence, larger-scale placebo-controlled clinical studies are warranted to investigate the effects of cannabidiol in particular as an adjunct to psychological therapy or medication to determine its potential utility for treating anxiety-related disorders in the future.
Collapse
Affiliation(s)
- Eleni P. Papagianni
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD UK
| | - Carl W. Stevenson
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD UK
| |
Collapse
|
22
|
Lisboa SF, Vila-Verde C, Rosa J, Uliana DL, Stern CAJ, Bertoglio LJ, Resstel LB, Guimaraes FS. Tempering aversive/traumatic memories with cannabinoids: a review of evidence from animal and human studies. Psychopharmacology (Berl) 2019; 236:201-226. [PMID: 30604182 DOI: 10.1007/s00213-018-5127-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/14/2018] [Indexed: 01/08/2023]
Abstract
RATIONALE Aversive learning and memory are essential to cope with dangerous and stressful stimuli present in an ever-changing environment. When this process is dysfunctional, however, it is associated with posttraumatic stress disorder (PTSD). The endocannabinoid (eCB) system has been implicated in synaptic plasticity associated with physiological and pathological aversive learning and memory. OBJECTIVE AND METHODS The objective of this study was to review and discuss evidence on how and where in the brain genetic or pharmacological interventions targeting the eCB system would attenuate aversive/traumatic memories through extinction facilitation in laboratory animals and humans. The effect size of the experimental intervention under investigation was also calculated. RESULTS Currently available data indicate that direct or indirect activation of cannabinoid type-1 (CB1) receptor facilitates the extinction of aversive/traumatic memories. Activating CB1 receptors around the formation of aversive/traumatic memories or their reminders can potentiate their subsequent extinction. In most cases, the effect size has been large (Cohen's d ≥ 1.0). The brain areas responsible for the abovementioned effects include the medial prefrontal cortex, amygdala, and/or hippocampus. The potential role of cannabinoid type-2 (CB2) receptors in extinction learning is now under investigation. CONCLUSION Drugs augmenting the brain eCB activity can temper the impact of aversive/traumatic experiences by diverse mechanisms depending on the moment of their administration. Considering the pivotal role the extinction process plays in PTSD, the therapeutic potential of these drugs is evident. The sparse number of clinical trials testing these compounds in stress-related disorders is a gap in the literature that needs to be addressed.
Collapse
Affiliation(s)
- Sabrina F Lisboa
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil.
| | - C Vila-Verde
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - J Rosa
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - D L Uliana
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - C A J Stern
- Department of Pharmacology, Federal University of Parana, Curitiba, PR, Brazil
| | - L J Bertoglio
- Department of Pharmacology, Federal University of Santa Catarina, Florianopolis, SC, Brazil
| | - L B Resstel
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - F S Guimaraes
- Department of Pharmacology, Medical School of Ribeirão Preto, University of São Paulo (FMRP/USP), Av Bandeirantes 3900, Monte Alegre, 14049900, Ribeirão Preto, São Paulo, Brazil.,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), Medical School of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil
| |
Collapse
|
23
|
Augustin SM, Lovinger DM. Functional Relevance of Endocannabinoid-Dependent Synaptic Plasticity in the Central Nervous System. ACS Chem Neurosci 2018; 9:2146-2161. [PMID: 29400439 DOI: 10.1021/acschemneuro.7b00508] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The endocannabinoid (eCB) signaling system plays a key role in short-term and long-term synaptic plasticity in brain regions involved in various neural functions ranging from action selection to appetite control. This review will explore the role of eCBs in shaping neural circuit function to regulate behaviors. In particular, we will discuss the behavioral consequences of eCB mediated long-term synaptic plasticity in different brain regions. This review brings together evidence from in vitro and ex vivo studies and points out the need for more in vivo studies.
Collapse
Affiliation(s)
- Shana M. Augustin
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852, United States
| | - David M. Lovinger
- Laboratory for Integrative Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Rockville, Maryland 20852, United States
| |
Collapse
|
24
|
Prefrontal Cortex Deep Brain Stimulation Improves Fear and Anxiety-Like Behavior and Reduces Basolateral Amygdala Activity in a Preclinical Model of Posttraumatic Stress Disorder. Neuropsychopharmacology 2018; 43:1099-1106. [PMID: 28862251 PMCID: PMC5854795 DOI: 10.1038/npp.2017.207] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 08/06/2017] [Accepted: 08/29/2017] [Indexed: 11/09/2022]
Abstract
Deep brain stimulation (DBS) is being investigated for a number of psychiatric indications, including posttraumatic stress disorder (PTSD). Preclinical studies continue to be a cornerstone for the development of new DBS applications. We investigate whether DBS delivered to the infralimbic cortex (IL), a region involved in mechanisms of stress resiliency, may counter behavioral abnormalities in rats that present persistent extinction deficits and long-term anxiety after exposure to fear conditioning. Rats undergoing fear conditioning/extinction were segregated into weak and strong extinction groups (WE >70% or SE <30% of freezing during extinction). Following 2 weeks of DBS, animals were exposed to novel recall sessions and tested in the open field, novelty-suppressed feeding, and elevated plus maze. zif268 expression was measured in structures involved in mechanisms of fear and stress. In vivo electrophysiology was used to record activity from the basolateral amygdala (BLA). We found that DBS improved extinction deficits and anxiety-like behavior in WE animals, having no significant effects in SE rats. No major differences in absolute zif268 levels were recorded across groups. However, correlation between zif268 expression in the IL and BLA was disrupted in WE animals, a deficit that was countered by DBS treatment. Electrophysiology experiments have shown that DBS reduced BLA firing of both putative principal cells and interneurons in WE rats, with no significant differences being detected between SE and SE DBS animals. In summary, IL DBS mitigated fear, partially improved anxiety-like behavior, reversed neurocircuitry abnormalities, and reduced BLA cell firing in a preclinical model of PTSD.
Collapse
|
25
|
Psychedelics and reconsolidation of traumatic and appetitive maladaptive memories: focus on cannabinoids and ketamine. Psychopharmacology (Berl) 2018; 235:433-445. [PMID: 29178010 DOI: 10.1007/s00213-017-4793-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 11/14/2017] [Indexed: 12/22/2022]
Abstract
RATIONALE Clinical data with 3,4-methylenedioxymethamphetamine (MDMA) in post-traumatic stress disorder (PTSD) patients recently stimulated interest on the potential therapeutic use of psychedelics in disorders characterized by maladaptive memories, including substance use disorders (SUD). The rationale for the use of MDMA in PTSD and SUD is being extended to a broader beneficial "psychedelic effect," which is supporting further clinical investigations, in spite of the lack of mechanistic hypothesis. Considering that the retrieval of emotional memories reactivates specific brain mechanisms vulnerable to inhibition, interference, or strengthening (i.e., the reconsolidation process), it was proposed that the ability to retrieve and change these maladaptive memories might be a novel intervention for PTSD and SUD. The mechanisms underlying MDMA effects indicate memory reconsolidation modulation as a hypothetical process underlying its efficacy. OBJECTIVE Mechanistic and clinical studies with other two classes of psychedelic substances, namely cannabinoids and ketamine, are providing data in support of a potential use in PTSD and SUD based on the modulation of traumatic and appetitive memory reconsolidation, respectively. Here, we review preclinical and clinical data on cannabinoids and ketamine effects on biobehavioral processes related to the reconsolidation of maladaptive memories. RESULTS We report the findings supporting (or not) the working hypothesis linking the potential therapeutic effect of these substances to the underlying reconsolidation process. We also proposed possible approaches for testing the use of these two classes of drugs within the current paradigm of reconsolidation memory inhibition. CONCLUSIONS Metaplasticity may be the process in common between cannabinoids and ketamine/ketamine-like substance effects on the mediation and potential manipulation of maladaptive memories.
Collapse
|
26
|
Stern CA, de Carvalho CR, Bertoglio LJ, Takahashi RN. Effects of Cannabinoid Drugs on Aversive or Rewarding Drug-Associated Memory Extinction and Reconsolidation. Neuroscience 2018; 370:62-80. [DOI: 10.1016/j.neuroscience.2017.07.018] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 06/23/2017] [Accepted: 07/09/2017] [Indexed: 12/22/2022]
|
27
|
Integrating Endocannabinoid Signaling and Cannabinoids into the Biology and Treatment of Posttraumatic Stress Disorder. Neuropsychopharmacology 2018; 43:80-102. [PMID: 28745306 PMCID: PMC5719095 DOI: 10.1038/npp.2017.162] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/17/2017] [Accepted: 07/20/2017] [Indexed: 01/21/2023]
Abstract
Exposure to stress is an undeniable, but in most cases surmountable, part of life. However, in certain individuals, exposure to severe or cumulative stressors can lead to an array of pathological conditions including posttraumatic stress disorder (PTSD), characterized by debilitating trauma-related intrusive thoughts, avoidance behaviors, hyperarousal, as well as depressed mood and anxiety. In the context of the rapidly changing political and legal landscape surrounding use of cannabis products in the USA, there has been a surge of public and research interest in the role of cannabinoids in the regulation of stress-related biological processes and in their potential therapeutic application for stress-related psychopathology. Here we review the current state of knowledge regarding the effects of cannabis and cannabinoids in PTSD and the preclinical and clinical literature on the effects of cannabinoids and endogenous cannabinoid signaling systems in the regulation of biological processes related to the pathogenesis of PTSD. Potential therapeutic implications of the reviewed literature are also discussed. Finally, we propose that a state of endocannabinoid deficiency could represent a stress susceptibility endophenotype predisposing to the development of trauma-related psychopathology and provide biologically plausible support for the self-medication hypotheses used to explain high rates of cannabis use in patients with trauma-related disorders.
Collapse
|
28
|
Fattore L, Piva A, Zanda MT, Fumagalli G, Chiamulera C. Psychedelics and reconsolidation of traumatic and appetitive maladaptive memories: focus on cannabinoids and ketamine. Psychopharmacology (Berl) 2017. [PMID: 29178010 DOI: 10.1007/s00213-017-4793-4.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
RATIONALE Clinical data with 3,4-methylenedioxymethamphetamine (MDMA) in post-traumatic stress disorder (PTSD) patients recently stimulated interest on the potential therapeutic use of psychedelics in disorders characterized by maladaptive memories, including substance use disorders (SUD). The rationale for the use of MDMA in PTSD and SUD is being extended to a broader beneficial "psychedelic effect," which is supporting further clinical investigations, in spite of the lack of mechanistic hypothesis. Considering that the retrieval of emotional memories reactivates specific brain mechanisms vulnerable to inhibition, interference, or strengthening (i.e., the reconsolidation process), it was proposed that the ability to retrieve and change these maladaptive memories might be a novel intervention for PTSD and SUD. The mechanisms underlying MDMA effects indicate memory reconsolidation modulation as a hypothetical process underlying its efficacy. OBJECTIVE Mechanistic and clinical studies with other two classes of psychedelic substances, namely cannabinoids and ketamine, are providing data in support of a potential use in PTSD and SUD based on the modulation of traumatic and appetitive memory reconsolidation, respectively. Here, we review preclinical and clinical data on cannabinoids and ketamine effects on biobehavioral processes related to the reconsolidation of maladaptive memories. RESULTS We report the findings supporting (or not) the working hypothesis linking the potential therapeutic effect of these substances to the underlying reconsolidation process. We also proposed possible approaches for testing the use of these two classes of drugs within the current paradigm of reconsolidation memory inhibition. CONCLUSIONS Metaplasticity may be the process in common between cannabinoids and ketamine/ketamine-like substance effects on the mediation and potential manipulation of maladaptive memories.
Collapse
Affiliation(s)
- Liana Fattore
- National Research Council of Italy, Institute of Neuroscience-Cagliari, Cagliari, Italy
| | - Alessandro Piva
- Sezione Farmacologia, Dipt. Diagnostica e Sanità Pubblica, Università degli Studi di Verona, Policlinico Borgo Roma, P.le Scuro 10, 37134, Verona, Italy
| | - Mary Tresa Zanda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Cittadella Universitaria di Monserrato, SP 8, Km 0.700, 09042, Monserrato, Italy
| | - Guido Fumagalli
- Sezione Farmacologia, Dipt. Diagnostica e Sanità Pubblica, Università degli Studi di Verona, Policlinico Borgo Roma, P.le Scuro 10, 37134, Verona, Italy
| | - Cristiano Chiamulera
- Sezione Farmacologia, Dipt. Diagnostica e Sanità Pubblica, Università degli Studi di Verona, Policlinico Borgo Roma, P.le Scuro 10, 37134, Verona, Italy.
| |
Collapse
|
29
|
Stern CA, da Silva TR, Raymundi AM, de Souza CP, Hiroaki-Sato VA, Kato L, Guimarães FS, Andreatini R, Takahashi RN, Bertoglio LJ. Cannabidiol disrupts the consolidation of specific and generalized fear memories via dorsal hippocampus CB 1 and CB 2 receptors. Neuropharmacology 2017; 125:220-230. [DOI: 10.1016/j.neuropharm.2017.07.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/17/2017] [Accepted: 07/22/2017] [Indexed: 11/28/2022]
|
30
|
Aisenberg N, Serova L, Sabban EL, Akirav I. The effects of enhancing endocannabinoid signaling and blocking corticotrophin releasing factor receptor in the amygdala and hippocampus on the consolidation of a stressful event. Eur Neuropsychopharmacol 2017; 27:913-927. [PMID: 28663121 DOI: 10.1016/j.euroneuro.2017.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 04/20/2017] [Accepted: 06/10/2017] [Indexed: 11/18/2022]
Abstract
Current clinical and pre-clinical data suggest that both cannabinoid agents and blockage of CRF through corticotrophin releasing factor receptor type 1 (CRFr1) may offer therapeutic benefits for post-traumatic stress disorder (PTSD). Here we aim to determine whether they are more effective when combined when microinjected into the basolateral amygdala (BLA) or CA1 area of the hippocampus after exposure to a stressful event in the shock/reminders rat model for PTSD. Injection of the fatty acid amide hydrolase (FAAH) inhibitor URB597 after the shock into either the BLA or CA1 facilitated extinction, and attenuated startle response and anxiety-like behavior. These preventive effects of URB597 were found to be mediated by the CB1 receptor. Intra-BLA and intra-CA1 microinjection of the CRFr1 antagonist, CP-154,526 attenuated startle response. When microinjected into the BLA, CP-154,526 also attenuated freezing behavior during exposure to the first reminder and decreased anxiety-like behavior. The combined treatment of URB597 and CP-154,526 was not more effective than the separate treatments. Finally, mRNA levels of CRF, CRFr1 and CB1r were significantly higher in the BLA of rats exposed to shock and reminders compared to non-shocked rats almost one month after the shock. Taken together, the results show that enhancing endocannabinoid signaling in the amygdala and hippocampus produced a more favorable spectrum of effects than those caused by the CRFr1 antagonist. The findings suggest that FAAH inhibitors may be used as a novel treatment for stress-related anxiety disorders.
Collapse
MESH Headings
- Amidohydrolases/antagonists & inhibitors
- Amidohydrolases/metabolism
- Animals
- Anxiety/drug therapy
- Anxiety/metabolism
- Basolateral Nuclear Complex/drug effects
- Basolateral Nuclear Complex/metabolism
- Benzamides/pharmacology
- CA1 Region, Hippocampal/drug effects
- CA1 Region, Hippocampal/metabolism
- Carbamates/pharmacology
- Disease Models, Animal
- Endocannabinoids/metabolism
- Male
- Memory Consolidation/drug effects
- Memory Consolidation/physiology
- Nootropic Agents/pharmacology
- Pyrimidines/pharmacology
- Pyrroles/pharmacology
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Reflex, Startle/drug effects
- Reflex, Startle/physiology
- Stress Disorders, Post-Traumatic/drug therapy
- Stress Disorders, Post-Traumatic/metabolism
- Stress, Psychological/drug therapy
- Stress, Psychological/metabolism
Collapse
Affiliation(s)
- Nurit Aisenberg
- Department of Psychology, University of Haifa, Haifa 3498838, Israel
| | - Lidia Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | - Irit Akirav
- Department of Psychology, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
31
|
Shoshan N, Akirav I. The effects of cannabinoid receptors activation and glucocorticoid receptors deactivation in the amygdala and hippocampus on the consolidation of a traumatic event. Neurobiol Learn Mem 2017; 144:248-258. [PMID: 28818702 DOI: 10.1016/j.nlm.2017.08.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 08/10/2017] [Accepted: 08/13/2017] [Indexed: 12/26/2022]
Abstract
Ample evidence demonstrates that fear learning contributes significantly to many anxiety pathologies including post-traumatic stress disorder (PTSD). The endocannabinoid (eCB) system may offer therapeutic benefits for PTSD and it is a modulator of the hypothalamic pituitary adrenal (HPA) axis. Here we compared the separated and combined effects of blocking glucocorticoid receptors (GRs) using the GR antagonist RU486 and enhancing CB1r signaling using the CB1/2 receptor agonist WIN55,212-2 in the CA1 and basolateral amygdala (BLA) on the consolidation of traumatic memory. Traumatic memory was formed by exposure to a severe footshock in an inhibitory avoidance apparatus followed by exposure to trauma reminders. Intra-BLA RU486 (10ng/side) and WIN55,212-2 (5μg/side) administered immediately after shock exposure dampened the consolidation of the memory about the traumatic event and attenuated the increase in acoustic startle response in rats exposed to shock and reminders. In the CA1, WIN55,212-2 impaired consolidation and attenuated the increase in acoustic startle response whereas RU486 had no effect. The effects of WIN55,212-2 were found to be mediated by CB1 receptors, but not by GRs. Moreover, post-shock systemic WIN55,212-2 (0.5mg/kg) administration prevented the increase in GRs and CB1 receptor levels in the CA1 and BLA in rats exposed to shock and reminders. The findings suggest that the BLA is a locus of action of cannabinoids and glucocorticoids in modulating consolidation of traumatic memory in a rat model of PTSD. Also, the findings highlight novel targets for the treatment of emotional disorders and PTSD in particular.
Collapse
Affiliation(s)
- Noa Shoshan
- Department of Psychology, University of Haifa, Haifa 3498838, Israel
| | - Irit Akirav
- Department of Psychology, University of Haifa, Haifa 3498838, Israel.
| |
Collapse
|
32
|
Shoshan N, Segev A, Abush H, Mizrachi Zer-Aviv T, Akirav I. Cannabinoids prevent the differential long-term effects of exposure to severe stress on hippocampal- and amygdala-dependent memory and plasticity. Hippocampus 2017; 27:1093-1109. [DOI: 10.1002/hipo.22755] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/22/2017] [Accepted: 06/20/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Noa Shoshan
- Department of Psychology; University of Haifa; Haifa 3498838 Israel
| | - Amir Segev
- Department of Psychology; University of Haifa; Haifa 3498838 Israel
| | - Hila Abush
- Department of Psychology; University of Haifa; Haifa 3498838 Israel
| | | | - Irit Akirav
- Department of Psychology; University of Haifa; Haifa 3498838 Israel
| |
Collapse
|
33
|
Endocannabinoid signaling and memory dynamics: A synaptic perspective. Neurobiol Learn Mem 2017; 138:62-77. [DOI: 10.1016/j.nlm.2016.07.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/21/2016] [Accepted: 07/29/2016] [Indexed: 01/26/2023]
|
34
|
|
35
|
Cannabinoid modulation of zebrafish fear learning and its functional analysis investigated by c-Fos expression. Pharmacol Biochem Behav 2016; 153:18-31. [PMID: 27965084 DOI: 10.1016/j.pbb.2016.12.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/05/2016] [Accepted: 12/09/2016] [Indexed: 12/31/2022]
Abstract
It has been shown that zebrafish fear learning proceeds in the same way as reported for rodents. However, in zebrafish fear learning it is possible to substitute the use of electric shocks as unconditioned stimulus and utilize the inborn fear responses to the alarm substance Schreckstoff, instead. The skin extract Schreckstoff elicits typical fear reactions such as preferred bottom dwelling, swimming in a tighter shoal, erratic movements and freezing. This natural fear behavior can be transferred from Schreckstoff to any other sensory stimulus by associative conditioning (fear learning). We presented Schreckstoff simultaneously with a red light stimulus and tested the effectiveness of fear learning during memory retrieval. The two brain regions known to be relevant for learning in zebrafish are the medial and the lateral pallium of the dorsal telencephalon, both containing rich expressions of the endocannabinoid receptor CB1. To test the influence of the zebrafish endocannabinoid system on fear acquisition learning, an experimental group of ten fish was pretreated with the CB1 receptor agonist THC (Δ9-tetrahydrocannabinol; 100nM for 1h). We found that CB1 activation significantly inhibited acquisition of fear learning, possibly by impairing stimulus encoding processes in pallial areas. This was supported by analyzes of c-Fos expression in the brains of experimental animals. Schreckstoff exposure during fear acquisition learning and memory retrieval during red light presentation increased the number of labelled cells in pallial structures, but in no other brain region investigated (e.g. striatum, thalamus, and habenula). THC administration before fear conditioning significantly decreased c-Fos expression in these structures to a level similar to the control group without Schreckstoff experience, suggesting that Schreckstoff induced fear learning requires brain circuits restricted mainly to pallial regions of the dorsal telencephalon.
Collapse
|
36
|
Bennett MR, Arnold J, Hatton SN, Lagopoulos J. Regulation of fear extinction by long-term depression: The roles of endocannabinoids and brain derived neurotrophic factor. Behav Brain Res 2016; 319:148-164. [PMID: 27867101 DOI: 10.1016/j.bbr.2016.11.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 11/10/2016] [Accepted: 11/15/2016] [Indexed: 11/19/2022]
Abstract
The extinction of a conditioned fear response is of great interest in the search for a means of ameliorating adverse neurobiological changes resulting from stress. The discovery that endocannibinoid (EC) levels are inversely related to the extent of such stress, and that the amygdala is a primary site mediating stress, suggests that ECs in this brain region might play a major role in extinction. Supporting this are the observations that the basolateral complex of the amygdala shows an increase in ECs only during extinction and that early clinical trials indicate that cannabinoid-like agents, when taken orally by patients suffering from post traumatic stress disorder (PTSD), reduce insomnia and nightmares. In order to optimize the potential of these agents to ameliorate symptoms of PTSD four important questions need to be answered: first, what is the identity of the cells that release ECs in the amygdala during extinction; second, what are their sites of action; third, what roles do the ECs play in the alleviation of long- depression (LTD), a process central to extinction; and finally, to what extent does brain derived neurotrophic factor (BDNF) facilitate the release of ECs? A review of the relevant literature is presented in an attempt to answer these questions. It is suggested that the principal cell involved in EC synthesis and release during extinction is the so-called excitatory extinction neuron in the basal nucleus of the amygdala. Furthermore that the main site of action of the ECs is the adjacent calcitonin gene-related peptide inhibitory interneurons, whose normal role of blocking the excitatory neurons is greatly diminished. The molecular pathways leading (during extinction trials) to the synthesis and release of ECs from synaptic spines of extinction neurons, that is potentiated by BDNF, are also delineated in this review. Finally, consideration is given to how the autocrine action of BDNF, linked to the release of ECs, can lead to the sustained release of these, so maintaining extinction over long times.
Collapse
Affiliation(s)
- Maxwell R Bennett
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia.
| | - Jonathon Arnold
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Sean N Hatton
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia
| | - Jim Lagopoulos
- The Brain and Mind Centre, University of Sydney, 94 Mallett Street, Camperdown, NSW, Australia; The Sunshine Coast Mind and Neuroscience, Thompson Institute, The University of the Sunshine Coast, QLD, Australia
| |
Collapse
|
37
|
The endocannabinoid system and Post Traumatic Stress Disorder (PTSD): From preclinical findings to innovative therapeutic approaches in clinical settings. Pharmacol Res 2016; 111:668-678. [DOI: 10.1016/j.phrs.2016.07.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/30/2016] [Accepted: 07/21/2016] [Indexed: 02/01/2023]
|
38
|
Interplay between serotonin and cannabinoid function in the amygdala in fear conditioning. Brain Res 2016; 1636:142-151. [DOI: 10.1016/j.brainres.2016.01.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 12/14/2015] [Accepted: 01/20/2016] [Indexed: 12/26/2022]
|
39
|
An Overview of Translationally Informed Treatments for Posttraumatic Stress Disorder: Animal Models of Pavlovian Fear Conditioning to Human Clinical Trials. Biol Psychiatry 2015; 78:E15-27. [PMID: 26238379 PMCID: PMC4527085 DOI: 10.1016/j.biopsych.2015.06.008] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 05/07/2015] [Accepted: 06/02/2015] [Indexed: 01/13/2023]
Abstract
Posttraumatic stress disorder manifests after exposure to a traumatic event and is characterized by avoidance/numbing, intrusive symptoms and flashbacks, mood and cognitive disruptions, and hyperarousal/reactivity symptoms. These symptoms reflect dysregulation of the fear system likely caused by poor fear inhibition/extinction, increased generalization, and/or enhanced consolidation or acquisition of fear. These phenotypes can be modeled in animal subjects using Pavlovian fear conditioning, allowing investigation of the underlying neurobiology of normative and pathological fear. Preclinical studies reveal a number of neurotransmitter systems and circuits critical for aversive learning and memory that have informed the development of therapies used in human clinical trials. In this review, we discuss the evidence for a number of established and emerging pharmacotherapies and device-based treatments for posttraumatic stress disorder that have been developed via a bench to bedside translational model.
Collapse
|
40
|
Busquets-Garcia A, Desprez T, Metna-Laurent M, Bellocchio L, Marsicano G, Soria-Gomez E. Dissecting the cannabinergic control of behavior: Thewherematters. Bioessays 2015; 37:1215-25. [DOI: 10.1002/bies.201500046] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Arnau Busquets-Garcia
- Group “Endocannabinoids and Neuroadaptation,” NeuroCentre Magendie, INSERM U862; University of Bordeaux; Bordeaux France
| | - Tifany Desprez
- Group “Endocannabinoids and Neuroadaptation,” NeuroCentre Magendie, INSERM U862; University of Bordeaux; Bordeaux France
| | - Mathilde Metna-Laurent
- Group “Endocannabinoids and Neuroadaptation,” NeuroCentre Magendie, INSERM U862; University of Bordeaux; Bordeaux France
| | - Luigi Bellocchio
- Group “Endocannabinoids and Neuroadaptation,” NeuroCentre Magendie, INSERM U862; University of Bordeaux; Bordeaux France
| | - Giovanni Marsicano
- Group “Endocannabinoids and Neuroadaptation,” NeuroCentre Magendie, INSERM U862; University of Bordeaux; Bordeaux France
| | - Edgar Soria-Gomez
- Group “Endocannabinoids and Neuroadaptation,” NeuroCentre Magendie, INSERM U862; University of Bordeaux; Bordeaux France
| |
Collapse
|
41
|
Kishimoto Y, Cagniard B, Yamazaki M, Nakayama J, Sakimura K, Kirino Y, Kano M. Task-specific enhancement of hippocampus-dependent learning in mice deficient in monoacylglycerol lipase, the major hydrolyzing enzyme of the endocannabinoid 2-arachidonoylglycerol. Front Behav Neurosci 2015; 9:134. [PMID: 26082696 PMCID: PMC4451424 DOI: 10.3389/fnbeh.2015.00134] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 05/11/2015] [Indexed: 12/03/2022] Open
Abstract
Growing evidence indicates that the endocannabinoid system is important for the acquisition and/or extinction of learning and memory. However, it is unclear which endocannabinoid(s) play(s) a crucial role in these cognitive functions, especially memory extinction. To elucidate the physiological role of 2-arachidonoylglycerol (2-AG), a major endocannabinoid, in behavioral and cognitive functions, we conducted a comprehensive behavioral test battery in knockout (KO) mice deficient in monoacylglycerol lipase (MGL), the major hydrolyzing enzyme of 2-AG. We found age-dependent increases in spontaneous physical activity (SPA) in MGL KO mice. Next, we tested the MGL KO mice using 5 hippocampus-dependent learning paradigms (i.e., Morris water maze (MWM), contextual fear conditioning, novel object recognition test, trace eyeblink conditioning, and water-finding test). In the MWM, MGL KO mice showed normal acquisition of reference memory, but exhibited significantly faster extinction of the learned behavior. Moreover, they showed faster memory acquisition on the reversal-learning task of the MWM. In contrast, in the contextual fear conditioning, MGL KO mice tended to show slower memory extinction. In the novel object recognition and water-finding tests, MGL KO mice exhibited enhanced memory acquisition. Trace eyeblink conditioning was not altered in MGL KO mice throughout the acquisition and extinction phases. These results indicate that 2-AG signaling is important for hippocampus-dependent learning and memory, but its contribution is highly task-dependent.
Collapse
Affiliation(s)
- Yasushi Kishimoto
- Laboratory of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University Sanuki, Kagawa, Japan
| | - Barbara Cagniard
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo Bunkyo-ku, Tokyo, Japan
| | - Maya Yamazaki
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University Niigata, Japan
| | - Junko Nakayama
- Laboratory of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University Sanuki, Kagawa, Japan
| | - Kenji Sakimura
- Department of Cellular Neurobiology, Brain Research Institute, Niigata University Niigata, Japan
| | - Yutaka Kirino
- Laboratory of Neurobiophysics, Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University Sanuki, Kagawa, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
42
|
Baldi E, Bucherelli C. Brain sites involved in fear memory reconsolidation and extinction of rodents. Neurosci Biobehav Rev 2015; 53:160-90. [DOI: 10.1016/j.neubiorev.2015.04.003] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 03/30/2015] [Accepted: 04/06/2015] [Indexed: 12/21/2022]
|
43
|
Bowers ME, Ressler KJ. Interaction between the cholecystokinin and endogenous cannabinoid systems in cued fear expression and extinction retention. Neuropsychopharmacology 2015; 40:688-700. [PMID: 25176168 PMCID: PMC4289957 DOI: 10.1038/npp.2014.225] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/10/2014] [Accepted: 08/02/2014] [Indexed: 01/29/2023]
Abstract
Post-traumatic stress disorder (PTSD) is thought to develop, in part, from improper inhibition of fear. Accordingly, one of the most effective treatment strategies for PTSD is exposure-based psychotherapy. Ideally, neuroscience would inform adjunct therapies that target the neurotransmitter systems involved in extinction processes. Separate studies have implicated the cholecystokinin (CCK) and endocannabinoid systems in fear; however, there is a high degree of anatomical colocalization between the cannabinoid 1 receptor (Cnr1) and CCK in the basolateral amygdala (BLA), a brain region critical for emotion regulation. Although most research has focused on GABA and GABAergic plasticity as the mechanism by which Cnr1 mediates fear inhibition, we hypothesize that a functional interaction between Cnr1 and CCKB receptor (CCKBR) is critical for fear extinction processes. In this study, systemic pharmacological manipulation of the cannabinoid system modulated cued fear expression in C57BL/6J mice after consolidation of auditory fear conditioning. Knockout of the CCKBR, however, had no effect on fear- or anxiety-like behaviors. Nonetheless, administration of a Cnr1 antagonist increased freezing behavior during a cued fear expression test in wild-type subjects, but had no effect on freezing behavior in CCKBR knockout littermates. In addition, we found that Cnr1-positive fibers form perisomatic clusters around CCKBR-positive cell bodies in the BLA. These CCKBR-positive cells comprise a molecularly heterogenous population of excitatory and inhibitory neurons. These findings provide novel evidence that Cnr1 contributes to cued fear expression via an interaction with the CCK system. Dysfunctional Cnr1-CCKBR interactions might contribute to the etiology of, or result from, fear-related psychiatric disease.
Collapse
Affiliation(s)
- Mallory E Bowers
- Behavioral Neuroscience, Department of Psychiatry and Behavioral Sciences, Howard Hughes Medical Institute, Emory University, Yerkes Research Center, Atlanta, GA, USA
| | - Kerry J Ressler
- Behavioral Neuroscience, Department of Psychiatry and Behavioral Sciences, Howard Hughes Medical Institute, Emory University, Yerkes Research Center, Atlanta, GA, USA,Howard Hughes Medical Institute, Emory University, Atlanta, GA, USA,Behavioral Neuroscience, Department of Psychiatry and Behavioral Sciences, Howard Hughes Medical Institute, Emory University, Yerkes Research Center, 954 Gatewood Dr, NE Atlanta, GA 30329, USA, Tel: +1 404 727 7739, Fax: +1 404 727 8070, E-mail:
| |
Collapse
|
44
|
Singewald N, Schmuckermair C, Whittle N, Holmes A, Ressler KJ. Pharmacology of cognitive enhancers for exposure-based therapy of fear, anxiety and trauma-related disorders. Pharmacol Ther 2014; 149:150-90. [PMID: 25550231 PMCID: PMC4380664 DOI: 10.1016/j.pharmthera.2014.12.004] [Citation(s) in RCA: 275] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 12/24/2014] [Indexed: 12/20/2022]
Abstract
Pathological fear and anxiety are highly debilitating and, despite considerable advances in psychotherapy and pharmacotherapy they remain insufficiently treated in many patients with PTSD, phobias, panic and other anxiety disorders. Increasing preclinical and clinical evidence indicates that pharmacological treatments including cognitive enhancers, when given as adjuncts to psychotherapeutic approaches [cognitive behavioral therapy including extinction-based exposure therapy] enhance treatment efficacy, while using anxiolytics such as benzodiazepines as adjuncts can undermine long-term treatment success. The purpose of this review is to outline the literature showing how pharmacological interventions targeting neurotransmitter systems including serotonin, dopamine, noradrenaline, histamine, glutamate, GABA, cannabinoids, neuropeptides (oxytocin, neuropeptides Y and S, opioids) and other targets (neurotrophins BDNF and FGF2, glucocorticoids, L-type-calcium channels, epigenetic modifications) as well as their downstream signaling pathways, can augment fear extinction and strengthen extinction memory persistently in preclinical models. Particularly promising approaches are discussed in regard to their effects on specific aspects of fear extinction namely, acquisition, consolidation and retrieval, including long-term protection from return of fear (relapse) phenomena like spontaneous recovery, reinstatement and renewal of fear. We also highlight the promising translational value of the preclinial research and the clinical potential of targeting certain neurochemical systems with, for example d-cycloserine, yohimbine, cortisol, and L-DOPA. The current body of research reveals important new insights into the neurobiology and neurochemistry of fear extinction and holds significant promise for pharmacologically-augmented psychotherapy as an improved approach to treat trauma and anxiety-related disorders in a more efficient and persistent way promoting enhanced symptom remission and recovery.
Collapse
Affiliation(s)
- N Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria.
| | - C Schmuckermair
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - N Whittle
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - A Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - K J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
45
|
Endogenous cannabinoid release within prefrontal-limbic pathways affects memory consolidation of emotional training. Proc Natl Acad Sci U S A 2014; 111:18333-8. [PMID: 25489086 DOI: 10.1073/pnas.1420285111] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Previous studies have provided extensive evidence that administration of cannabinoid drugs after training modulates the consolidation of memory for an aversive experience. The present experiments investigated whether the memory consolidation is regulated by endogenously released cannabinoids. The experiments first examined whether the endocannabinoids anandamide (AEA) and 2-arachidonoyl glycerol (2-AG) are released by aversive training. Inhibitory avoidance training with higher footshock intensity produced increased levels of AEA in the amygdala, hippocampus, and medial prefrontal cortex (mPFC) shortly after training in comparison with levels assessed in rats trained with lower footshock intensity or unshocked controls exposed only to the training apparatus. In contrast, 2-AG levels were not significantly elevated. The additional finding that posttraining infusions of the fatty acid amide hydrolase (FAAH) inhibitor URB597, which selectively increases AEA levels at active synapses, administered into the basolateral complex of the amygdala (BLA), hippocampus, or mPFC enhanced memory strongly suggests that the endogenously released AEA modulates memory consolidation. Moreover, in support of the view that this emotional training-associated increase in endocannabinoid neurotransmission, and its effects on memory enhancement, depends on the integrity of functional interactions between these different brain regions, we found that disruption of BLA activity blocked the training-induced increases in AEA levels as well as the memory enhancement produced by URB597 administered into the hippocampus or mPFC. Thus, the findings provide evidence that emotionally arousing training increases AEA levels within prefrontal-limbic circuits and strongly suggest that this cannabinoid activation regulates emotional arousal effects on memory consolidation.
Collapse
|
46
|
Ren Y, Wang J, Xu PB, Xu YJ, Miao CH. Systemic or intra-amygdala infusion of an endocannabinoid CB1 receptor antagonist AM251 blocked propofol-induced anterograde amnesia. Neurosci Lett 2014; 584:287-91. [PMID: 25445359 DOI: 10.1016/j.neulet.2014.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/13/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
Abstract
Propofol is well-known for its anterograde amnesic actions. However, a recent experiment showed that propofol can also produce retrograde memory enhancement effects via an interaction with the endocannabinoid CB1 system. Therefore, the authors hypothesized that the regulating effect of propofol on the endocannabinoid CB1 system might also decrease the anterograde amnesic effect of propofol under some conditions, which might be a risk factor for intraoperative awareness. Since, the basolateral amygdala (BLA) has been confirmed to mediate propofol-induced anterograde amnesia and the BLA contains a high concentration of CB1 receptors, the authors investigated whether and how the endocannabinoid system, particularly the CB1 receptor within BLA, influences propofol-induced anterograde amnesia. Male Sprague-Dawley rats trained with inhibitory avoidance (IA) were systematically pre-trained using a memory-impairing dose of propofol (25 mg/kg). Before propofol administration, rats received an intraperitoneal injection of a CB1 receptor antagonist AM251 (1 mg/kg or 2 mg/kg) or a bilateral intra-BLA injection of AM251 (0.6 ng or 6 ng per 0.5 μl). Twenty-four hours after IA training, the IA retention latency was tested. It was found that systemic or intra-BLA injection of a non-regulating dose of AM251 (2 mg/kg or 6 ng per 0.5 μl, respectively) blocked the memory-impairing effect of propofol. These results indicate that the anterograde amnesic effect of propofol is mediated, in part, by activation of the CB1 cannabinoid receptors in the BLA.
Collapse
Affiliation(s)
- Y Ren
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - J Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - P B Xu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Y J Xu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - C H Miao
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| |
Collapse
|
47
|
Papini S, Sullivan GM, Hien DA, Shvil E, Neria Y. Toward a translational approach to targeting the endocannabinoid system in posttraumatic stress disorder: a critical review of preclinical research. Biol Psychol 2014; 104:8-18. [PMID: 25448242 DOI: 10.1016/j.biopsycho.2014.10.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 10/18/2014] [Accepted: 10/27/2014] [Indexed: 12/12/2022]
Abstract
Despite the lack of clinical research, marijuana and synthetic cannabinoids have been approved to treat posttraumatic stress disorder (PTSD) in several states in the United States. This review critically examines preclinical research on the endocannabinoid system (ECS) in order to evaluate three key questions that are relevant to PTSD: (1) Does ECS dysfunction impact fear extinction? (2) Can stress-related symptoms be prevented by ECS modulation? (3) Is the ECS a potential target for enhancing PTSD treatment? Disruption of the ECS impaired fear extinction in rodents, and ECS abnormalities have been observed in PTSD. Targeting fear memories via the ECS had mixed results in rodents, whereas augmented cannabinoid receptor activation typically facilitated extinction. However, the translational value of these findings is limited by the paucity and inconsistency of human research. Further investigation is necessary to determine whether incorporating cannabinoids in treatment would benefit individuals with PTSD, with cautious attention to risks.
Collapse
Affiliation(s)
- Santiago Papini
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA; City College of New York, 160 Convent Avenue, New York, NY, 10031, USA; Columbia University Medical Center, W. 168th Street, New York, NY, 10032, USA
| | - Gregory M Sullivan
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA; Columbia University Medical Center, W. 168th Street, New York, NY, 10032, USA
| | - Denise A Hien
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA; City College of New York, 160 Convent Avenue, New York, NY, 10031, USA; Columbia University Medical Center, W. 168th Street, New York, NY, 10032, USA
| | - Erel Shvil
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA; Columbia University Medical Center, W. 168th Street, New York, NY, 10032, USA
| | - Yuval Neria
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA; Columbia University Medical Center, W. 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
48
|
Zhang JJ, Han J, Sui N. Okadaic acid blocks the effects of 5-aza-2-deoxycytidine on consolidation, acquisition and retrieval of morphine-induced place preference in rats. Neuropharmacology 2014; 86:282-93. [DOI: 10.1016/j.neuropharm.2014.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 07/18/2014] [Accepted: 08/05/2014] [Indexed: 12/22/2022]
|
49
|
Bukalo O, Pinard CR, Holmes A. Mechanisms to medicines: elucidating neural and molecular substrates of fear extinction to identify novel treatments for anxiety disorders. Br J Pharmacol 2014; 171:4690-718. [PMID: 24835117 DOI: 10.1111/bph.12779] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/28/2014] [Accepted: 05/04/2014] [Indexed: 12/11/2022] Open
Abstract
The burden of anxiety disorders is growing, but the efficacy of available anxiolytic treatments remains inadequate. Cognitive behavioural therapy for anxiety disorders focuses on identifying and modifying maladaptive patterns of thinking and behaving, and has a testable analogue in rodents in the form of fear extinction. A large preclinical literature has amassed in recent years describing the neural and molecular basis of fear extinction in rodents. In this review, we discuss how this work is being harnessed to foster translational research on anxiety disorders and facilitate the search for new anxiolytic treatments. We begin by summarizing the anatomical and functional connectivity of a medial prefrontal cortex (mPFC)-amygdala circuit that subserves fear extinction, including new insights from optogenetics. We then cover some of the approaches that have been taken to model impaired fear extinction and associated impairments with mPFC-amygdala dysfunction. The principal goal of the review is to evaluate evidence that various neurotransmitter and neuromodulator systems mediate fear extinction by modulating the mPFC-amygdala circuitry. To that end, we describe studies that have tested how fear extinction is impaired or facilitated by pharmacological manipulations of dopamine, noradrenaline, 5-HT, GABA, glutamate, neuropeptides, endocannabinoids and various other systems, which either directly target the mPFC-amygdala circuit, or produce behavioural effects that are coincident with functional changes in the circuit. We conclude that there are good grounds to be optimistic that the progress in defining the molecular substrates of mPFC-amygdala circuit function can be effectively leveraged to identify plausible candidates for extinction-promoting therapies for anxiety disorders.
Collapse
Affiliation(s)
- Olena Bukalo
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| | | | | |
Collapse
|
50
|
McLaughlin RJ, Hill MN, Gorzalka BB. A critical role for prefrontocortical endocannabinoid signaling in the regulation of stress and emotional behavior. Neurosci Biobehav Rev 2014; 42:116-31. [PMID: 24582908 DOI: 10.1016/j.neubiorev.2014.02.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 01/31/2014] [Accepted: 02/18/2014] [Indexed: 12/21/2022]
Abstract
The prefrontal cortex (PFC) provides executive control of the brain in humans and rodents, coordinating cognitive, emotional, and behavioral responses to threatening stimuli and subsequent feedback inhibition of the hypothalamic-pituitary-adrenal (HPA) axis. The endocannabinoid system has emerged as a fundamental regulator of HPA axis feedback inhibition and an important modulator of emotional behavior. However, the precise role of endocannabinoid signaling within the PFC with respect to stress coping and emotionality has only recently been investigated. This review discusses the current state of knowledge regarding the localization and function of the endocannabinoid system in the PFC, its sensitivity to stress and its role in modulating the neuroendocrine and behavioral responses to aversive stimuli. We propose a model whereby steady-state endocannabinoid signaling in the medial PFC indirectly regulates the outflow of pyramidal neurons by fine-tuning GABAergic inhibition. Local activation of this population of CB1 receptors increases the downstream targets of medial PFC activation, which include inhibitory interneurons in the basolateral amygdala, inhibitory relay neurons in the bed nucleus of the stria terminalis and monoamine cell bodies such as the dorsal raphe nucleus. This ultimately produces beneficial effects on emotionality (active coping responses to stress and reduced anxiety) and assists in constraining activation of the HPA axis. Under conditions of chronic stress, or in individuals suffering from mood disorders, this system may be uniquely recruited to help maintain appropriate function in the face of adversity, while breakdown of the endocannabinoid system in the medial PFC may be, in and of itself, sufficient to produce neuropsychiatric illness. Thus, we suggest that endocannabinoid signaling in the medial PFC may represent an attractive target for the treatment of stress-related disorders.
Collapse
Affiliation(s)
| | - Matthew N Hill
- Department of Cell Biology & Anatomy and Department of Psychiatry, Calgary, AB, Canada; Department of Psychiatry, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Boris B Gorzalka
- Department of Psychology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|