1
|
Dragon J, Obuchowicz E. How depression and antidepressant drugs affect endocannabinoid system?-review of clinical and preclinical studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:4511-4536. [PMID: 38280009 DOI: 10.1007/s00210-023-02938-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 12/30/2023] [Indexed: 01/29/2024]
Abstract
As major depressive disorder is becoming a more and more common issue in modern society, it is crucial to discover new possible grip points for its diagnosis and antidepressive therapy. One of them is endocannabinoid system, which has been proposed as a manager of emotional homeostasis, and thus, endocannabinoid alterations have been found in animals undergoing various preclinical models of depression procedures as well as in humans suffering from depressive-like disorders. In this review article, studies regarding those alterations have been summed up and analyzed. Another important issue raised by the researchers is the impact of currently used antidepressive drugs on endocannabinoid system so that it would be possible to predict reversibility of endocannabinoid alterations following stress exposure and, in the future, to be able to design individually personalized therapies. Preclinical studies investigating this topic have been analyzed and described in this article. Unfortunately, too few clinical studies in this field exist, what indicates an urgent need for collecting such data, so that it would be possible to compare them with preclinical outcomes and draw reliable conclusions.
Collapse
Affiliation(s)
- Jonasz Dragon
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków Street 18, 40-752, Katowice, Poland.
| | - Ewa Obuchowicz
- Department of Pharmacology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medyków Street 18, 40-752, Katowice, Poland
| |
Collapse
|
2
|
Zhang H, Sun Y, Yau SY, Zhou Y, Song X, Zhang HT, Zhu B, Wu H, Chen G. Synergistic effects of two natural compounds of iridoids on rapid antidepressant action by up-regulating hippocampal PACAP signaling. Br J Pharmacol 2022; 179:4078-4091. [PMID: 35362097 DOI: 10.1111/bph.15847] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/30/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Current mainstream antidepressants have limited efficacy and a delayed onset of action. Yueju is a traditional herbal medicine conferring rapid antidepressant activity. Here we attempted to identify the effective compounds from Yueju and the underlying mechanisms. EXPERIMENTAL APPROACH A transcriptomic analysis was employed to discover key candidate molecules for rapid antidepressant response. The enriched compounds in Yueju were identified with HPLC. Antidepressant effects were evaluated periodically using various behavioral paradigms. The mechanistic signaling was assessed using site-directed pharmacological intervention or optogenetic manipulation. KEY RESULTS A transcriptomic analysis revealed that Yueju up-regulated pituitary adenylate cyclase activating polypeptide (PACAP) expression in the hippocampus. Two iridoids geniposide (GP) and shanzhiside methyl-ester (SM) were enriched in Yueju. Co-treatment of GP and SM each at an equivalent dose in Yueju synergistically increased PACAP expression and elicited rapid antidepressant effects, which were prevented by intra-hippocampal dentate gyrus (DG) infusions of a PACAP antagonist or optogenetic inactivation of PACAP-expressing neurons. GP-SM co-treatment rapidly reduced CaMKII phosphorylation and enhanced mTOR/4EBP1/P70S6k/BDNF signaling, while intra-DG infusions of a CaMKII activator blunted rapid antidepressant effects and BDNF expression up-regulation induced by GP-SM co-treatment. A single administration of GP-SM rapidly improved depression-like behaviors and up-regulated hippocampal PACAP signaling in the repeated corticosterone-induced depression model, further confirming its rapid antidepressant action and the involvement of PACAP. CONCLUSION AND IMPLICATIONS GP-SM co-treatment elicited a synergistic effect on rapid antidepressant effects via triggering hippocampal PACAP activity and associated CaMKII-BDNF signaling, shedding lights on the development of novel targeted antidepressants.
Collapse
Affiliation(s)
- Hailou Zhang
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders & School of Chinese Medicine, Jinan University, Guangzhou, China.,College of Traditional Chinese Medicine & College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Co-innovation Center of Neurogeneration, Nantong University, Nantong, Jiangsu, China
| | - Yan Sun
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders & School of Chinese Medicine, Jinan University, Guangzhou, China.,College of Traditional Chinese Medicine & College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hong Kong, China
| | - Yanmeng Zhou
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Xinxin Song
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai'an, China
| | - Han-Ting Zhang
- Departments of Neuroscience and Behavioral Medicine & Psychiatry, the Rockefeller Neuroscience Institute, West Virginia University Health Sciences Center, Morgantown, WV, USA
| | - Boran Zhu
- College of Traditional Chinese Medicine & College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Haoxin Wu
- College of Traditional Chinese Medicine & College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Gang Chen
- Interdisciplinary Institute for Personalized Medicine in Brain Disorders & School of Chinese Medicine, Jinan University, Guangzhou, China.,Departments of Psychiatry & Clinical and Translational Institute of Psychiatric Disorders, First Affiliated Hospital of Jinan University, Guangzhou, China.,Co-innovation Center of Neurogeneration, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
3
|
Zhao L, Guo R, Cao N, Lin Y, Yang W, Pei S, Ma X, Zhang Y, Li Y, Song Z, Du W, Xiao X, Liu C. An Integrative Pharmacology-Based Pattern to Uncover the Pharmacological Mechanism of Ginsenoside H Dripping Pills in the Treatment of Depression. Front Pharmacol 2021; 11:590457. [PMID: 33658934 PMCID: PMC7917282 DOI: 10.3389/fphar.2020.590457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 09/30/2020] [Indexed: 12/30/2022] Open
Abstract
Objectives: To evaluate the pharmacodynamical effects and pharmacological mechanism of Ginsenoside H dripping pills (GH) in chronic unpredictable mild stress (CUMS) model rats. Methods: First, the CUMS-induced rat model was established to assess the anti-depressant effects of GH (28, 56, and 112 mg/kg) by the changes of the behavioral indexes (sucrose preference, crossing score, rearing score) and biochemical indexes (serotonin, dopamine, norepinephrine) in Hippocampus. Then, the components of GH were identified by ultra-performance liquid chromatography-iron trap-time of flight-mass spectrometry (UPLC/IT-TOF MS). After network pharmacology analysis, the active ingredients of GH were further screened out based on OB and DL, and the PPI network of putative targets of active ingredients of GH and depression candidate targets was established based on STRING database. The PPI network was analyzed topologically to obtain key targets, so as to predict the potential pharmacological mechanism of GH acting on depression. Finally, some major target proteins involved in the predictive signaling pathway were validated experimentally. Results: The establishment of CUMS depression model was successful and GH has antidepressant effects, and the middle dose of GH (56 mg/kg) showed the best inhibitory effects on rats with depressant-like behavior induced by CUMS. Twenty-eight chemical components of GH were identified by UPLC/IT-TOF MS. Subsequently, 20(S)-ginsenoside Rh2 was selected as active ingredient and the PPI network of the 43 putative targets of 20(S)-ginsenoside Rh2 containing in GH and the 230 depression candidate targets, was established based on STRING database, and 47 major targets were extracted. Further network pharmacological analysis indicated that the cAMP signaling pathway may be potential pharmacological mechanism regulated by GH acting on depression. Among the cAMP signaling pathway, the major target proteins, namely, cAMP, PKA, CREB, p-CREB, BDNF, were used to verify in the CUMS model rats. The results showed that GH could activate the cAMP-PKA-CREB-BDNF signaling pathway to exert antidepressant effects. Conclusions: An integrative pharmacology-based pattern was used to uncover that GH could increase the contents of DA, NE and 5-HT, activate cAMP-PKA-CREB-BDNF signaling pathway exert antidepressant effects.
Collapse
Affiliation(s)
- Libin Zhao
- School of Graduate, Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Zhendong Research Institute, Shanxi Zhendong Pharmaceutical Co., Ltd, Beijing, China
| | - Rui Guo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Ningning Cao
- School of Graduate, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingxian Lin
- School of Graduate, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Wenjing Yang
- State Key Laboratory of Critical Technology in Innovative Chinese Medicine, TCM Research Center, Tianjin Tasly Pharmaceutical CO., LTD., Tianjin, China
| | - Shuai Pei
- School of Graduate, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaowei Ma
- Shandong Huayu University of Technology, Shandong, China
| | - Yu Zhang
- School of Graduate, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yingpeng Li
- School of Graduate, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Song
- State Key Laboratory of Critical Technology in Innovative Chinese Medicine, TCM Research Center, Tianjin Tasly Pharmaceutical CO., LTD., Tianjin, China
| | - Wuxun Du
- School of Graduate, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xuefeng Xiao
- School of Graduate, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Changxiao Liu
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin, China
| |
Collapse
|
4
|
Scarante FF, Ribeiro MA, Almeida-Santos AF, Guimarães FS, Campos AC. Glial Cells and Their Contribution to the Mechanisms of Action of Cannabidiol in Neuropsychiatric Disorders. Front Pharmacol 2021; 11:618065. [PMID: 33613284 PMCID: PMC7890128 DOI: 10.3389/fphar.2020.618065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022] Open
Abstract
Cannabidiol (CBD) is a phytocannabinoid with a broad-range of therapeutic potential in several conditions, including neurological (epilepsy, neurodegenerative diseases, traumatic and ischemic brain injuries) and psychiatric disorders (schizophrenia, addiction, major depressive disorder, and anxiety). The pharmacological mechanisms responsible for these effects are still unclear, and more than 60 potential molecular targets have been described. Regarding neuropsychiatric disorders, most studies investigating these mechanisms have focused on neuronal cells. However, glial cells (astrocytes, oligodendrocytes, microglia) also play a crucial role in keeping the homeostasis of the central nervous system. Changes in glial functions have been associated with neuropathological conditions, including those for which CBD is proposed to be useful. Mostly in vitro studies have indicated that CBD modulate the activation of proinflammatory pathways, energy metabolism, calcium homeostasis, and the proliferative rate of glial cells. Likewise, some of the molecular targets proposed for CBD actions are f expressed in glial cells, including pharmacological receptors such as CB1, CB2, PPAR-γ, and 5-HT1A. In the present review, we discuss the currently available evidence suggesting that part of the CBD effects are mediated by interference with glial cell function. We also propose additional studies that need to be performed to unveil the contribution of glial cells to CBD effects in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Franciele F. Scarante
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Melissa A. Ribeiro
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Ana F. Almeida-Santos
- Department of Physiology and Biophysics, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Francisco S. Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Alline C. Campos
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
5
|
Colangeli R, Teskey GC, Di Giovanni G. Endocannabinoid-serotonin systems interaction in health and disease. PROGRESS IN BRAIN RESEARCH 2021; 259:83-134. [PMID: 33541682 DOI: 10.1016/bs.pbr.2021.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Endocannabinoid (eCB) and serotonin (5-HT) neuromodulatory systems work both independently and together to finely orchestrate neuronal activity throughout the brain to strongly sculpt behavioral functions. Surprising parallelism between the behavioral effects of 5-HT and eCB activity has been widely reported, including the regulation of emotional states, stress homeostasis, cognitive functions, food intake and sleep. The distribution pattern of the 5-HT system and the eCB molecular elements in the brain display a strong overlap and several studies report a functional interplay and even a tight interdependence between eCB/5-HT signaling. In this review, we examine the available evidence of the interaction between the eCB and 5-HT systems. We first introduce the eCB system, then we describe the eCB/5-HT crosstalk at the neuronal and synaptic levels. Finally, we explore the potential eCB/5-HT interaction at the behavioral level with the implication for psychiatric and neurological disorders. The precise elucidation of how this neuromodulatory interaction dynamically regulates biological functions may lead to the development of more targeted therapeutic strategies for the treatment of depressive and anxiety disorders, psychosis and epilepsy.
Collapse
Affiliation(s)
- Roberto Colangeli
- Section of Neuroscience and Cell Biology, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy; Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.
| | - G Campbell Teskey
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Giuseppe Di Giovanni
- Laboratory of Neurophysiology, Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Msida, Malta; Neuroscience Division, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
6
|
Escartín Pérez RE, Mancilla Díaz JM, Cortés Salazar F, López Alonso VE, Florán Garduño B. CB1/5-HT/GABA interactions and food intake regulation. PROGRESS IN BRAIN RESEARCH 2021; 259:177-196. [PMID: 33541676 DOI: 10.1016/bs.pbr.2021.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Despite historically the serotonergic, GABAergic, and cannabinoid systems have been shown to play a crucial role in the central regulation of eating behavior, interest in the study of the interactions of these neurotransmission systems has only now been investigated. Current evidence suggests that serotonin may influence normal and pathological eating behavior in significantly more complex ways than was initially thought. This knowledge has opened the possibility of exploring the potential clinical utility of new therapeutic strategies more effective and safer than the current approaches to treat pathological eating behavior. Furthermore, the nature and complexity of the interactions between these neurotransmitter systems have provided a better understanding of the pathophysiological mechanisms not only of eating behavior and eating disorders but also of some of the comorbidities associated with modulation of cortical circuits, which are involved in high order cognitive processes. Accordingly, in the present chapter, the clinical and experimental findings of the interactions between serotonin, GABA, and cannabinoids are synthesized, emphasizing the pharmacological, neurophysiological, and neuroanatomical aspects that could potentially improve the current therapeutic approaches against pathological eating behavior.
Collapse
Affiliation(s)
- Rodrigo Erick Escartín Pérez
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México.
| | - Juan Manuel Mancilla Díaz
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México
| | - Felipe Cortés Salazar
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México
| | - Verónica Elsa López Alonso
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México
| | - Benjamín Florán Garduño
- Facultad de Estudios Superiores Iztacala, División de Investigación y Posgrado, Laboratorio de Neurobiología de la Alimentación, Universidad Nacional Autónoma de México, México, México
| |
Collapse
|
7
|
Rana T, Behl T, Sehgal A, Mehta V, Singh S, Kumar R, Bungau S. Integrating Endocannabinoid Signalling In Depression. J Mol Neurosci 2021; 71:2022-2034. [PMID: 33471311 DOI: 10.1007/s12031-020-01774-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 12/09/2020] [Indexed: 12/24/2022]
Abstract
Depression is a common mental disorder and is the leading cause of suicide globally. Because of the significant diversity in mental disorders, accurate diagnosis is difficult. Hence, the investigation of novel biomarkers is a key research perspective in psychotherapy to enable an individually tailored treatment approach. The prefrontal cortex (PFC) is a vital cortical region whose circuitry has been implicated in the development of depressive disorder. The endocannabinoid system (ECS) has garnered increasing attention because of its involvement in several diverse physiological brain processes including regulation of emotional, motivational and cognitive functions. The current review article explores the function of the key elements of the ECS as a biomarker in depressive disorder. The activity of endocannabinoids is thought to be moderated by the CB1 receptors in the central nervous system (CNS). Variations in the concentration of endocannabinoids and the binding affinity of CB1 receptors and their density have been identified in the PFC of persons with depression. Such discoveries support our theory that alteration in endocannabinoid function leads to the pathophysiological features of depressive disorders. Moreover, evidence from animal and human studies has revealed that dysfunction in endocannabinoid signalling can produce depression-like behaviours; therefore, improvement of endocannabinoid signalling may represent a new therapeutic approach for the management of depressive disorders.
Collapse
Affiliation(s)
- Tarapati Rana
- Government Pharmacy College, Seraj, Mandi, Himachal Pradesh, India.,Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Vineet Mehta
- Distt. Shimla, Government College of Pharmacy, Himachal Pradesh, Rohru, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Ravinder Kumar
- Cardiovascular Research Institute, Icahn School of Medicine, New York, USA
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
8
|
Neuroprotective effect of chronic administration of cannabidiol during the abstinence period on methamphetamine-induced impairment of recognition memory in the rats. Behav Pharmacol 2020; 31:385-396. [DOI: 10.1097/fbp.0000000000000544] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
9
|
Coccurello R. Anhedonia in depression symptomatology: Appetite dysregulation and defective brain reward processing. Behav Brain Res 2019; 372:112041. [DOI: 10.1016/j.bbr.2019.112041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/22/2022]
|
10
|
Batista LA, Moreira FA. Cannabinoid CB 1 receptors mediate the anxiolytic effects induced by systemic alprazolam and intra-periaqueductal gray 5-HT 1A receptor activation. Neurosci Lett 2019; 703:5-10. [PMID: 30858018 DOI: 10.1016/j.neulet.2019.03.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/01/2019] [Accepted: 03/07/2019] [Indexed: 01/04/2023]
Abstract
The endocannabinoid system has been implicated in the modulation of behaviors related to anxiety and panic disorders. Accordingly, facilitation of CB1 receptor signaling reduces the consequences of aversive stimuli in animal models. However, the role of the CB1 receptor in the effects of anxiolytic drugs has remained unclear. Here, we tested the hypothesis that the anxiolytic and panicolytic responses to systemic alprazolam injection and local 5-HT1A receptor activation in the dorsolateral periaqueductal gray (dlPAG) depend on CB1 receptor activation. Systemic injection of alprazolam (4 mg/kg) induced an anxiolytic-like effect in the elevated T maze (ETM) model of panic and anxiety, which was prevented by the CB1 antagonist AM251 (0.3 mg/kg). Likewise, intra-dlPAG injection of the 5-HT1A receptor agonist 8-OH-DPAT (3.2 nmol/0.2 u L) also reduced anxiety-like behavior, a response prevented by intra-dlPAG injection of AM251 (100 pmol/0.2 µL). 8-OH-DPAT (8 nmol/0.2 µL) also presented a panicolytic-like activity in the escape reaction induced by chemical stimulation of the dlPAG, which was not prevented by AM251 (100 pmol/0.2 µL). These results suggest that CB1 receptor signaling is involved in the effects of anxiolytic drugs, with potential implications for developing new treatments for anxiety disorders.
Collapse
Affiliation(s)
- Luara A Batista
- Graduate School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil.
| | - Fabricio A Moreira
- Graduate School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| |
Collapse
|
11
|
Abstract
Chronic pain is a frequent condition that affects an estimated 20% of people worldwide, accounting for 15%-20% of doctors' appointments (Treede et al., 2015). It lacks the acute warning function of physiologic nociception, and instead involves the activation of multiple neurophysiologic mechanisms in the somatosensory system, a complex neuronal network under the control of powerful autoregulatory loops and able to undergo rapid neuroplastic alteration (Verdu et al., 2008). There is a growing body of research suggesting that some such pathways are shared by major psychologic disorders such as depression and anxiety, opening new avenues in co-treatment strategies. In particular, besides anticonvulsants, which are today used as analgesics, other psychopharmaceuticals, such as the tricyclic antidepressants, are displaying efficacy in the treatment of neuropathic and nociceptive chronic pain. The state of the art regarding the mechanisms of nociception and the pharmacology of both the neurotransmitters involved and the wide range of psychoactive compounds that may be useful in the treatment of chronic pain are discussed.
Collapse
|
12
|
Mato S, Pilar-Cuéllar F, Valdizán EM, González-Maeso J, Rodríguez-Puertas R, Meana J, Sallés J, Crespo-Facorro B, Pazos Á. Selective up-regulation of cannabinoid CB 1 receptor coupling to Go-proteins in suicide victims with mood disorders. Biochem Pharmacol 2018; 157:258-265. [PMID: 30099006 PMCID: PMC6263149 DOI: 10.1016/j.bcp.2018.08.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/08/2018] [Indexed: 01/11/2023]
Abstract
Brain endocannabinoid system is proposed to play a role in the pathogenesis of affective disorders. In the present study, we analyzed the functionality of the cannabinoid receptor type 1 (CB1 receptor) at different transduction levels in prefrontal cortex (PFC) of depressed suicide victims. We examined stimulation of [35S]GTPγS binding, activation of Gα protein subunits and inhibition of adenylyl cyclase by the cannabinoid agonist WIN55,212-2, as well as [3H]CP55,940 binding, in PFC homogenates from suicide victims with major depression (MD) and matched control subjects. CB1 receptor-stimulated [35S]GTPγS binding was significantly greater in the PFC of MD compared with matched controls (23%, p < 0.05). This increase was most evident in the PFC from MD subgroup with negative blood test for antidepressants (AD) at the time of death (AD-free) (38%, p < 0.05), being absent when comparing the AD-treated MD cases with their controls. The density of CB1 receptors and their coupling to adenylyl cyclase were similar between MD and control cases, regardless of the existence of AD intake. Analysis of [35S]GTPγS-labelled Gα subunits allowed for the detection of upregulated CB1 receptor coupling to Gαo, but not to Gαi1, Gαi2, Gαi3, Gαz subunits, in the PFC from AD-free MD suicides. These results suggest that increased CB1 receptor functionality at the Gαi/o protein level in the PFC of MD subjects is due to enhanced coupling to Gαo proteins and might be modulated by AD intake. These data provide new insights into the role of endocannabinoid neurotransmission in the pathobiology of MD and suggest its regulation by ADs.
Collapse
Affiliation(s)
- Susana Mato
- Department of Physiology and Pharmacology, University of Cantabria, E-39011 Santander, Spain
| | - Fuencisla Pilar-Cuéllar
- Department of Physiology and Pharmacology, University of Cantabria, E-39011 Santander, Spain; Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), E-39011 Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), E-28029 Madrid, Spain
| | - Elsa M Valdizán
- Department of Physiology and Pharmacology, University of Cantabria, E-39011 Santander, Spain; Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), E-39011 Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), E-28029 Madrid, Spain
| | - Javier González-Maeso
- Department of Pharmacology, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain
| | | | - Javier Meana
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), E-28029 Madrid, Spain; Department of Pharmacology, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain
| | - Joan Sallés
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), E-28029 Madrid, Spain; Department of Pharmacology, University of the Basque Country UPV/EHU, E-48940 Leioa, Spain
| | - Benedicto Crespo-Facorro
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), E-28029 Madrid, Spain; Hospital Universitario Marqués de Valdecilla, University of Cantabria-IDIVAL, School of Medicine, Department of Psychiatry, Santander, Spain
| | - Ángel Pazos
- Department of Physiology and Pharmacology, University of Cantabria, E-39011 Santander, Spain; Institute of Biomedicine and Biotechnology of Cantabria (IBBTEC), E-39011 Santander, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), E-28029 Madrid, Spain.
| |
Collapse
|
13
|
Rajkumar R, Dawe GS. OBscure but not OBsolete: Perturbations of the frontal cortex in common between rodent olfactory bulbectomy model and major depression. J Chem Neuroanat 2018; 91:63-100. [DOI: 10.1016/j.jchemneu.2018.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/02/2018] [Accepted: 04/04/2018] [Indexed: 02/08/2023]
|
14
|
Regulation of noradrenergic and serotonergic systems by cannabinoids: relevance to cannabinoid-induced effects. Life Sci 2018; 192:115-127. [DOI: 10.1016/j.lfs.2017.11.029] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/29/2017] [Accepted: 11/16/2017] [Indexed: 12/11/2022]
|
15
|
Campos AC, Fogaça MV, Scarante FF, Joca SRL, Sales AJ, Gomes FV, Sonego AB, Rodrigues NS, Galve-Roperh I, Guimarães FS. Plastic and Neuroprotective Mechanisms Involved in the Therapeutic Effects of Cannabidiol in Psychiatric Disorders. Front Pharmacol 2017; 8:269. [PMID: 28588483 PMCID: PMC5441138 DOI: 10.3389/fphar.2017.00269] [Citation(s) in RCA: 112] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 05/01/2017] [Indexed: 12/25/2022] Open
Abstract
Beneficial effects of cannabidiol (CBD) have been described for a wide range of psychiatric disorders, including anxiety, psychosis, and depression. The mechanisms responsible for these effects, however, are still poorly understood. Similar to clinical antidepressant or atypical antipsychotic drugs, recent findings clearly indicate that CBD, either acutely or repeatedly administered, induces plastic changes. For example, CBD attenuates the decrease in hippocampal neurogenesis and dendrite spines density induced by chronic stress and prevents microglia activation and the decrease in the number of parvalbumin-positive GABA neurons in a pharmacological model of schizophrenia. More recently, it was found that CBD modulates cell fate regulatory pathways such as autophagy and others critical pathways for neuronal survival in neurodegenerative experimental models, suggesting the potential benefit of CBD treatment for psychiatric/cognitive symptoms associated with neurodegeneration. These changes and their possible association with CBD beneficial effects in psychiatric disorders are reviewed here.
Collapse
Affiliation(s)
- Alline C Campos
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Manoela V Fogaça
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Franciele F Scarante
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Sâmia R L Joca
- Department of Physical and Chemical, School of Pharmaceutical Science of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Amanda J Sales
- Department of Physical and Chemical, School of Pharmaceutical Science of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Felipe V Gomes
- Department of Neuroscience, University of PittsburghPittsburgh, PA, United States
| | - Andreza B Sonego
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Naielly S Rodrigues
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| | - Ismael Galve-Roperh
- Department of Biochemistry and Molecular Biology I, School of Biology, Complutense UniversityMadrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas, Instituto de Universitario de Investigación en Neuroquímica and Instituto Ramón y Cajal de Investigación SanitariaMadrid, Spain
| | - Francisco S Guimarães
- Department of Pharmacology, Centre for Interdisciplinary Research on Applied Neurosciences (NAPNA), School of Medicine of Ribeirão Preto, University of São PauloRibeirão Preto, Brazil
| |
Collapse
|
16
|
McLaughlin PJ, Jagielo-Miller JE, Plyler ES, Schutte KK, Vemuri VK, Makriyannis A. Differential effects of cannabinoid CB1 inverse agonists and antagonists on impulsivity in male Sprague Dawley rats: identification of a possibly clinically relevant vulnerability involving the serotonin 5HT 1A receptor. Psychopharmacology (Berl) 2017; 234:1029-1043. [PMID: 28144708 DOI: 10.1007/s00213-017-4548-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/21/2017] [Indexed: 12/18/2022]
Abstract
RATIONALE Cannabinoid CB1 inverse agonists hold therapeutic promise as appetite suppressants but have produced suicidal behaviors among a small subpopulation in clinical trials. Anatomical and pharmacological evidence implicate the 5HT1A serotonin receptor in suicide in humans and impulsivity in humans and animals. OBJECTIVE The objective of the study is to assess whether 5HT1A blockade is necessary for CB1 ligands to produce impulsivity. METHODS Sprague Dawley rats were administered the CB1 inverse agonist AM 251, the CB1 antagonist AM 6527, or the peripherally restricted antagonist AM 6545, with or without pretreatment with the 5HT1A antagonist WAY 100,635 (WAY) on the paced fixed consecutive number (FCN) task, which measures choice to terminate a chain of responses prematurely. As FCN is sensitive to changes in time perception, which have been demonstrated with CB1 blockade, a novel variable consecutive number task with discriminative stimulus (VCN-S D ) was also performed and proposed to be less sensitive to changes in timing. RESULTS Pretreatment with WAY enabled mild but significant reductions in FCN accuracy for AM 251 and AM 6527. No effects were found for AM 6545. On the VCN-S D task, substantial impairments were found for the combination of WAY and AM 251. CONCLUSIONS AM 251, but not the antagonists AM 6527 or AM 6545, produced impulsivity only following systemic 5HT1A blockade. Although preliminary, the results may indicate that disrupted serotonin signaling produces a vulnerability to undesirable effects of CB1 inverse agonists, which is not evident in the general population. Furthermore, neutral CB1 antagonists do not produce this effect and therefore may have greater safety.
Collapse
Affiliation(s)
- Peter J McLaughlin
- Department of Psychology, Edinboro University of Pennsylvania, 210 East Normal Street, Edinboro, PA, 16444, USA.
| | - Julia E Jagielo-Miller
- Department of Psychology, Edinboro University of Pennsylvania, 210 East Normal Street, Edinboro, PA, 16444, USA.,Department of Psychology, University of Kentucky, Lexington, KY, 40506, USA
| | - Emily S Plyler
- Department of Psychology, Edinboro University of Pennsylvania, 210 East Normal Street, Edinboro, PA, 16444, USA.,Department of Biomedical Sciences, Kent State University, Kent, OH, 44242, USA
| | - Kerry K Schutte
- Department of Psychology, Edinboro University of Pennsylvania, 210 East Normal Street, Edinboro, PA, 16444, USA.,Department of Counseling, Psychology, and Special Education, Duquesne University, 600 Forbes Avenue, G8B Canevin Hall, Pittsburgh, PA, 15282, USA
| | - V Kiran Vemuri
- Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| | - Alexandros Makriyannis
- Center for Drug Discovery, Northeastern University, 360 Huntington Avenue, Boston, MA, 02115, USA
| |
Collapse
|
17
|
Soares VP, Campos AC. Evidences for the Anti-panic Actions of Cannabidiol. Curr Neuropharmacol 2017; 15:291-299. [PMID: 27157263 PMCID: PMC5412699 DOI: 10.2174/1570159x14666160509123955] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 02/26/2016] [Accepted: 04/27/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Panic disorder (PD) is a disabling psychiatry condition that affects approximately 5% of the worldwide population. Currently, long-term selective serotonin reuptake inhibitors (SSRIs) are the first-line treatment for PD; however, the common side-effect profiles and drug interactions may provoke patients to abandon the treatment, leading to PD symptoms relapse. Cannabidiol (CBD) is the major non-psychotomimetic constituent of the Cannabis sativa plant with anti-anxiety properties that has been suggested as an alternative for treating anxiety disorders. The aim of the present review was to discuss the effects and mechanisms involved in the putative anti-panic effects of CBD. METHODS electronic database was used as source of the studies selected selected based on the studies found by crossing the following keywords: cannabidiol and panic disorder; canabidiol and anxiety, cannabidiol and 5-HT1A receptor). RESULTS In the present review, we included both experimental laboratory animal and human studies that have investigated the putative anti-panic properties of CBD. Taken together, the studies assessed clearly suggest an anxiolytic-like effect of CBD in both animal models and healthy volunteers. CONCLUSION CBD seems to be a promising drug for the treatment of PD. However, novel clinical trials involving patients with the PD diagnosis are clearly needed to clarify the specific mechanism of action of CBD and the safe and ideal therapeutic doses of this compound.
Collapse
Affiliation(s)
| | - Alline C Campos
- Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo, 3900 Bandeirantes avenue, Ribeirao Preto-SP, Brazil
| |
Collapse
|
18
|
Genetic variation in the tryptophan hydroxylase 2 gene moderates depressive symptom trajectories and remission over 8 weeks of escitalopram treatment. Int Clin Psychopharmacol 2016; 31:127-33. [PMID: 26745768 DOI: 10.1097/yic.0000000000000115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The serotonin system plays an important role in the pathogenesis of major depressive disorder (MDD) and genetic variations in serotonin-related genes affect the efficacy of antidepressants. The aim of this study was to investigate the relationship between genotypic variation in six candidate serotonergic genes (ADCY9, HTR1B, GNB3, HTR2A, TPH2, SLC6A4) and depressive and anxiety symptom severity trajectories as well as remission following escitalopram treatment. A total of 166 Chinese patients with MDD were treated with escitalopram (open-label) for 8 weeks. TPH2 rs4570625 GG carriers were more likely to achieve depressive and anxiety symptom remission compared with T-allele carriers. At the trend level (P(corrected)=0.05), depressive symptom severity trajectories were moderated by TPH2 rs4570625. Patients with the GT or the GG genotype showed more favorable depressive symptom severity trajectories compared with TT genotype carriers. Polymorphisms in ADCY9, HTR1B, and HTR2A were nominally associated with symptom remission, but did not withstand correction for multiple comparisons. The HTTLPR polymorphism was not included in our final analysis because of a high percentage of missing data. These results suggested that genotypic variation in TPH2 may moderate the therapeutic response to esciatlopram among Chinese patients with MDD.
Collapse
|
19
|
Gunduz-Cinar O, Flynn S, Brockway E, Kaugars K, Baldi R, Ramikie TS, Cinar R, Kunos G, Patel S, Holmes A. Fluoxetine Facilitates Fear Extinction Through Amygdala Endocannabinoids. Neuropsychopharmacology 2016; 41:1598-609. [PMID: 26514583 PMCID: PMC4832021 DOI: 10.1038/npp.2015.318] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 09/11/2015] [Accepted: 10/06/2015] [Indexed: 02/07/2023]
Abstract
Pharmacologically elevating brain endocannabinoids (eCBs) share anxiolytic and fear extinction-facilitating properties with classical therapeutics, including the selective serotonin reuptake inhibitor, fluoxetine. There are also known functional interactions between the eCB and serotonin systems and preliminary evidence that antidepressants cause alterations in brain eCBs. However, the potential role of eCBs in mediating the facilitatory effects of fluoxetine on fear extinction has not been established. Here, to test for a possible mechanistic contribution of eCBs to fluoxetine's proextinction effects, we integrated biochemical, electrophysiological, pharmacological, and behavioral techniques, using the extinction-impaired 129S1/Sv1mJ mouse strain. Chronic fluoxetine treatment produced a significant and selective increase in levels of anandamide in the BLA, and an associated decrease in activity of the anandamide-catabolizing enzyme, fatty acid amide hydrolase. Slice electrophysiological recordings showed that fluoxetine-induced increases in anandamide were associated with the amplification of eCB-mediated tonic constraint of inhibitory, but not excitatory, transmission in the BLA. Behaviorally, chronic fluoxetine facilitated extinction retrieval in a manner that was prevented by systemic or BLA-specific blockade of CB1 receptors. In contrast to fluoxetine, citalopram treatment did not increase BLA eCBs or facilitate extinction. Taken together, these findings reveal a novel, obligatory role for amygdala eCBs in the proextinction effects of a major pharmacotherapy for trauma- and stressor-related disorders and anxiety disorders.
Collapse
Affiliation(s)
- Ozge Gunduz-Cinar
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, USA,Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), 5625 Fishers Lane, Room 2N09, Rockville, MD 20852-9411, USA, Tel: +1 301 443 4052, Fax: +1 301 480 8035, E-mail: or
| | - Shaun Flynn
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Emma Brockway
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Katherine Kaugars
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Rita Baldi
- Department of Psychiatry and Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Teniel S Ramikie
- Department of Psychiatry and Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Resat Cinar
- Laboratory of Physiological Studies, NIAAA, NIH, Bethesda, MD, USA
| | - George Kunos
- Laboratory of Physiological Studies, NIAAA, NIH, Bethesda, MD, USA
| | - Sachin Patel
- Department of Psychiatry and Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, MD, USA,Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), 5625 Fishers Lane, Room 2N09, Rockville, MD 20852-9411, USA, Tel: +1 301 443 4052, Fax: +1 301 480 8035, E-mail: or
| |
Collapse
|
20
|
Jiménez-Sánchez L, Linge R, Campa L, Valdizán EM, Pazos Á, Díaz Á, Adell A. Behavioral, neurochemical and molecular changes after acute deep brain stimulation of the infralimbic prefrontal cortex. Neuropharmacology 2016; 108:91-102. [PMID: 27108934 DOI: 10.1016/j.neuropharm.2016.04.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 01/14/2023]
Abstract
Deep brain stimulation (DBS) is a treatment that has shown some efficacy in treatment-resistant depression. In particular, DBS of the subcallosal cingulate gyrus (Brodmann's area 25, Cg25) has been successfully applied to treat refractory depression. In the rat, we have demonstrated that DBS applied to infralimbic (IL) cortex elevates the levels of glutamate and monoamines in the prefrontal cortex, and requires the stimulation of cortical α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) glutamate receptors for its antidepressant-like effects. However, the molecular targets of IL DBS are not fully known. To gain insight into these pathways, we have investigated whether IL DBS is able to reverse the behavioral, biochemical and molecular changes exhibited by the olfactory bulbectomized (OBX) rat. Our results revealed that 1 h IL DBS diminished hyperlocomotion, hyperemotionality and anhedonia, and increased social interaction shown by the OBX rats. Further, IL DBS increased prefrontal efflux of glutamate and serotonin in both sham-operated and OBX rats. With regard to molecular targets, IL DBS increases the synthesis of brain-derived neurotrophic factor (BDNF) and the GluA1 AMPA receptor subunit, and stimulates the Akt/mammalian target of rapamycin (mTOR) as well as the AMPA receptor/c-AMP response element binding (CREB) pathways. Temsirolimus, a known in vivo mTOR blocker, suppressed the antidepressant-like effect of IL DBS in naïve rats in the forced swim test, thus demonstrating for the first time that mTOR signaling is required for the antidepressant-like effects of IL DBS, which is in line with the antidepressant response of other rapid-acting antidepressant drugs.
Collapse
Affiliation(s)
- Laura Jiménez-Sánchez
- Departamento de Neuroquímica y Neurofarmacología, Instituto de Investigaciones Biomédicas de Barcelona (CSIC, IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Raquel Linge
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC, Universidad de Cantabria), 39011 Santander, Spain
| | - Leticia Campa
- Departamento de Neuroquímica y Neurofarmacología, Instituto de Investigaciones Biomédicas de Barcelona (CSIC, IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Elsa M Valdizán
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC, Universidad de Cantabria), 39011 Santander, Spain
| | - Ángel Pazos
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC, Universidad de Cantabria), 39011 Santander, Spain
| | - Álvaro Díaz
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC, Universidad de Cantabria), 39011 Santander, Spain
| | - Albert Adell
- Departamento de Neuroquímica y Neurofarmacología, Instituto de Investigaciones Biomédicas de Barcelona (CSIC, IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Instituto de Biomedicina y Biotecnología de Cantabria, IBBTEC (CSIC, Universidad de Cantabria), 39011 Santander, Spain.
| |
Collapse
|
21
|
McRae-Clark AL, Baker NL, Gray KM, Killeen T, Hartwell KJ, Simonian SJ. Vilazodone for cannabis dependence: A randomized, controlled pilot trial. Am J Addict 2015; 25:69-75. [PMID: 26685701 DOI: 10.1111/ajad.12324] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/03/2015] [Accepted: 12/02/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The purpose of this study was to evaluate the efficacy of vilazodone, a selective serotonin receptor inhibitor and partial 5-HT1A agonist, for treatment of cannabis dependence. METHODS Seventy-six cannabis-dependent adults were randomized to receive either up to 40 mg/day of vilazodone (n = 41) or placebo (n = 35) for 8 weeks combined with a brief motivational enhancement therapy intervention and contingency management to encourage study retention. Cannabis use outcomes were assessed via weekly urine cannabinoid tests; secondary outcomes included cannabis use self-report and cannabis craving. RESULTS Participants in both groups reported reduced self-reported cannabis use over the course of the study; however, vilazodone provided no advantage over placebo in reducing cannabis use. Men had significantly lower creatinine-adjusted cannabinoid levels and a trend for increased negative urine cannabinoid tests than women. DISCUSSION AND CONCLUSIONS Vilazodone was not more efficacious than placebo in reducing cannabis use. Important gender differences were noted, with women having worse cannabis use outcomes than men. SCIENTIFIC SIGNIFICANCE Further medication development efforts for cannabis use disorders are needed, and gender should be considered as an important variable in future trials.
Collapse
Affiliation(s)
- Aimee L McRae-Clark
- Department of Psychiatry, Medical University of South Carolina, Charleston, South Carolina
| | - Nathaniel L Baker
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, South Carolina
| | - Kevin M Gray
- Department of Psychiatry, Medical University of South Carolina, Charleston, South Carolina
| | - Therese Killeen
- Department of Psychiatry, Medical University of South Carolina, Charleston, South Carolina
| | - Karen J Hartwell
- Department of Psychiatry, Medical University of South Carolina, Charleston, South Carolina.,Ralph H. Johnson VA Medical Center, Charleston, South Carolina
| | - Susan J Simonian
- Department of Psychology, College of Charleston, Charleston, South Carolina
| |
Collapse
|
22
|
Buspirone treatment of cannabis dependence: A randomized, placebo-controlled trial. Drug Alcohol Depend 2015; 156:29-37. [PMID: 26386827 PMCID: PMC4633378 DOI: 10.1016/j.drugalcdep.2015.08.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/04/2015] [Accepted: 08/17/2015] [Indexed: 12/28/2022]
Abstract
BACKGROUND The purpose of this study was to evaluate the efficacy of buspirone, a partial 5-HT1A agonist, for treatment of cannabis dependence. METHODS One hundred seventy-five cannabis-dependent adults were randomized to receive either up to 60mg/day of buspirone (n=88) or placebo (n=87) for 12 weeks combined with a brief motivational enhancement therapy intervention and contingency management to encourage study retention. Cannabis use outcomes were assessed via weekly urine cannabinoid tests. RESULTS Participants in both groups reported reduced cannabis craving over the course of the study; however, buspirone provided no advantage over placebo in reducing cannabis use. Significant gender by treatment interactions were observed, with women randomized to buspirone having fewer negative urine cannabinoid tests than women randomized to placebo (p=0.007), and men randomized to buspirone having significantly lower creatinine adjusted cannabinoid levels as compared to those randomized to placebo (p=0.023). An evaluation of serotonin allelic variations did not find an association with buspirone treatment response. CONCLUSIONS Buspirone was not more efficacious than placebo in reducing cannabis use. Important gender differences were noted, with women having worse cannabis use outcomes with buspirone treatment. Considerations for future medication trials in this challenging population are discussed.
Collapse
|
23
|
Farb DH, Ratner MH. Targeting the modulation of neural circuitry for the treatment of anxiety disorders. Pharmacol Rev 2015; 66:1002-32. [PMID: 25237115 DOI: 10.1124/pr.114.009126] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Anxiety disorders are a major public health concern. Here, we examine the familiar area of anxiolysis in the context of a systems-level understanding that will hopefully lead to revealing an underlying pharmacological connectome. The introduction of benzodiazepines nearly half a century ago markedly improved the treatment of anxiety disorders. These agents reduce anxiety rapidly by allosterically enhancing the postsynaptic actions of GABA at inhibitory type A GABA receptors but side effects limit their use in chronic anxiety disorders. Selective serotonin reuptake inhibitors and serotonin/norepinephrine reuptake inhibitors have emerged as an effective first-line alternative treatment of such anxiety disorders. However, many individuals are not responsive and side effects can be limiting. Research into a relatively new class of agents known as neurosteroids has revealed novel modulatory sites and mechanisms of action that are providing insights into the pathophysiology of certain anxiety disorders, potentially bridging the gap between the GABAergic and serotonergic circuits underlying anxiety. However, translating the pharmacological activity of compounds targeted to specific receptor subtypes in rodent models of anxiety to effective therapeutics in human anxiety has not been entirely successful. Since modulating any one of several broad classes of receptor targets can produce anxiolysis, we posit that a systems-level discovery platform combined with an individualized medicine approach based on noninvasive brain imaging would substantially advance the development of more effective therapeutics.
Collapse
Affiliation(s)
- David H Farb
- Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | - Marcia H Ratner
- Laboratory of Molecular Neurobiology, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
24
|
Fitzgibbon M, Finn DP, Roche M. High Times for Painful Blues: The Endocannabinoid System in Pain-Depression Comorbidity. Int J Neuropsychopharmacol 2015; 19:pyv095. [PMID: 26342110 PMCID: PMC4815466 DOI: 10.1093/ijnp/pyv095] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Accepted: 08/17/2015] [Indexed: 01/06/2023] Open
Abstract
Depression and pain are two of the most debilitating disorders worldwide and have an estimated cooccurrence of up to 80%. Comorbidity of these disorders is more difficult to treat, associated with significant disability and impaired health-related quality of life than either condition alone, resulting in enormous social and economic cost. Several neural substrates have been identified as potential mediators in the association between depression and pain, including neuroanatomical reorganization, monoamine and neurotrophin depletion, dysregulation of the hypothalamo-pituitary-adrenal axis, and neuroinflammation. However, the past decade has seen mounting evidence supporting a role for the endogenous cannabinoid (endocannabinoid) system in affective and nociceptive processing, and thus, alterations in this system may play a key role in reciprocal interactions between depression and pain. This review will provide an overview of the preclinical evidence supporting an interaction between depression and pain and the evidence supporting a role for the endocannabinoid system in this interaction.
Collapse
Affiliation(s)
| | | | - Michelle Roche
- Physiology (Ms Fitzgibbon and Dr Roche), and Pharmacology and Therapeutics (Dr Finn), School of Medicine, Galway Neuroscience Centre and Centre for Pain Research (Ms Fitzgibbon, Dr Finn, and Dr Roche), National Centre for Biomedical Engineering Science, National University of Ireland Galway, Ireland.
| |
Collapse
|
25
|
Erdozain AM, Rubio M, Valdizan EM, Pazos A, Meana JJ, Fernández-Ruiz J, Alexander SPH, Callado LF. The endocannabinoid system is altered in the post-mortem prefrontal cortex of alcoholic subjects. Addict Biol 2015; 20:773-83. [PMID: 25041461 DOI: 10.1111/adb.12160] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
There is strong biochemical, pharmacological and genetic evidence for the involvement of the endocannabinoid system (ECS) in alcohol dependence. However, the majority of studies have been performed in animal models. The aim of the present study was to assess the state of the CB1 receptor, the enzymes fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL), and the extracellular signal-regulated kinase (ERK) and cyclic-AMP response element-binding protein (CREB) in the post-mortem prefrontal cortex of alcoholic subjects. Experiments were performed in samples from 44 subjects classified in four experimental groups: (1) non-suicidal alcoholic subjects (n = 11); (2) suicidal alcoholic subjects (n = 11); (3) non-alcoholic suicide victims (n = 11); and (4) control subjects (n = 11). We did not observe statistically significant differences in CB1 mRNA relative expression among the four experimental groups. Conversely, our results showed an increase in CB1 receptor protein expression in the prefrontal cortex of the suicidal alcoholic group (127.2 ± 7.3%), with no changes in functionality with regard to either G protein activation or the inhibition of adenylyl cyclase. In parallel, alcoholic subjects presented lower levels of MAGL activity, regardless of the cause of death. A significant decrease in the active form of ERK and CREB levels was also observed in both alcoholic groups. Taken together, our data are consistent with a role for the ECS in the neurobiological mechanisms underlying alcoholism. Moreover, the alterations reported here should be of great interest for the therapeutic treatment of this chronic psychiatric disease.
Collapse
Affiliation(s)
- Amaia M. Erdozain
- Department of Pharmacology; University of the Basque Country UPV/EHU; Bizkaia Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM); Madrid Spain
| | - Marina Rubio
- Department of Biochemistry and Molecular Biology; Faculty of Medicine; Complutense University; Madrid Spain
| | - Elsa M. Valdizan
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM); Madrid Spain
- Department of Physiology and Pharmacology; Institute of Biomedicine and Biotechnology IBBTEC (Universidad de Cantabria-CSIC-IDICAN) Santander; Cantabria Spain
| | - Angel Pazos
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM); Madrid Spain
- Department of Physiology and Pharmacology; Institute of Biomedicine and Biotechnology IBBTEC (Universidad de Cantabria-CSIC-IDICAN) Santander; Cantabria Spain
| | - J Javier Meana
- Department of Pharmacology; University of the Basque Country UPV/EHU; Bizkaia Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM); Madrid Spain
- BioCruces Health Research Institute; Bizkaia Spain
| | - Javier Fernández-Ruiz
- Department of Biochemistry and Molecular Biology; Faculty of Medicine; Complutense University; Madrid Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
- Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS); Madrid Spain
| | | | - Luis F. Callado
- Department of Pharmacology; University of the Basque Country UPV/EHU; Bizkaia Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM); Madrid Spain
- BioCruces Health Research Institute; Bizkaia Spain
| |
Collapse
|
26
|
Di Marzo V, Centonze D. Placebo effects in a multiple sclerosis spasticity enriched clinical trial with the oromucosal cannabinoid spray (THC/CBD): dimension and possible causes. CNS Neurosci Ther 2015; 21:215-21. [PMID: 25475413 PMCID: PMC6495119 DOI: 10.1111/cns.12358] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 11/26/2022] Open
Abstract
Regulatory authorities admit clinical studies with an initial enrichment phase to select patients that respond to treatment before randomization (Enriched Design Studies; EDSs). The trial period aims to prevent long-term drug exposure risks in patients with limited chances of improvement while optimizing costs. In EDSs for symptom control therapies providing early improvements and without a wash-out period, it is difficult to show further improvements and thus large therapeutic gains versus placebo. Moreover, in trials with cannabinoids, the therapeutic gains can be further biased in the postenrichment randomized phase because of carryover and other effects. The aims of the present review article are to examine the placebo effects in the enrichment and postenrichment phases of an EDS with Δ(9) -tetrahydrocannabinol and cannabidiol (THC/CBD) oromucosal spray in patients with multiple sclerosis (MS) spasticity and to discuss the possible causes of maintained efficacy after randomization in the placebo-allocated patients. The overall mean therapeutic gain of THC/CBD spray over placebo in resistant MS spasticity after 16 weeks can be estimated as a ~1.27-point improvement on the spasticity 0-10 Numerical Rating Scale (NRS; ~-20.1% of the baseline NRS score). We conclude that careful interpretation of the results of EDSs is required, especially when cannabinoid-based medications are being investigated.
Collapse
Affiliation(s)
- Vincenzo Di Marzo
- Endocannabinoid Research GroupInstitute of Biomolecular ChemistryConsiglio Nazionale delle RicerchePozzuoliNaplesItaly
| | - Diego Centonze
- Department of Systems MedicineMultiple Sclerosis Clinical and Research CenterTor Vergata UniversityRomeItaly
- IRCCS Santa Lucia FoundationRomeItaly
| |
Collapse
|
27
|
El Mansari M, Manta S, Oosterhof C, El Iskandrani KS, Chenu F, Shim S, Blier P. Restoration of serotonin neuronal firing following long-term administration of bupropion but not paroxetine in olfactory bulbectomized rats. Int J Neuropsychopharmacol 2015; 18:pyu050. [PMID: 25522394 PMCID: PMC4360219 DOI: 10.1093/ijnp/pyu050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Olfactory bulbectomized rats generally manifest many of the neurochemical, physiological, and behavioral features of major depressive disorder in humans. Another interesting feature of this model is that it responds to chronic but not acute antidepressant treatments, including selective serotonin reuptake inhibitors. The purpose of the present study was first to characterize the firing activity of dorsal raphe serotonin neurons in olfactory bulbectomized rats and then examine the effects of 2 antidepressants, bupropion and paroxetine. METHODS Olfactory bulbectomy was performed by aspirating olfactory bulbs in anesthetized rats. Vehicle and drugs were delivered for 2 and 14 days via subcutaneously implanted minipumps. In vivo electrophysiological recordings were carried out in male anesthetized Sprague-Dawley rats. RESULTS Following ablation of olfactory bulbs, the firing rate of serotonin neurons was decreased by 36%, leaving those of norepinephrine and dopamine neurons unchanged. In olfactory bulbectomized rats, bupropion (30 mg/kg/d) restored the firing rate of serotonin neurons to the control level following 2- and 14-day administration and also induced an increase in the tonic activation of serotonin(1A) receptors; paroxetine (10 mg/kg/d) did not result in a return to normal of the attenuated firing of serotonin neurons in olfactory bulbectomized rats. In the hippocampus, although at a higher dose of WAY 100635 than that required in bupropion-treated animals, paroxetine administration also resulted in an increase in the tonic activation of serotonin(1A) receptors. CONCLUSIONS The present results indicate that unlike paroxetine, bupropion administration normalized serotonin neuronal activity and increased tonic activation of the serotonin(1A) receptors in hippocampus.
Collapse
Affiliation(s)
- Mostafa El Mansari
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada.
| | - Stella Manta
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | - Chris Oosterhof
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | | | - Franck Chenu
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | - Stacey Shim
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| | - Pierre Blier
- University of Ottawa Institute of Mental Health Research, Ottawa, ON, Canada
| |
Collapse
|
28
|
Effects of desipramine treatment on stress-induced up-regulation of norepinephrine transporter expression in rat brains. Psychopharmacology (Berl) 2015; 232:379-90. [PMID: 25038868 PMCID: PMC9338776 DOI: 10.1007/s00213-014-3674-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 06/25/2014] [Indexed: 10/25/2022]
Abstract
RATIONALE Many studies demonstrate down-regulation of the norepinephrine transporter (NET) by desipramine (DMI) in vitro and in stress-naive rats. Little is known regarding regulation of the NET in stressed animals. OBJECTIVE The present study was designed to investigate effects of DMI on the expression of NET and protein kinases in the stress rat. METHODS Adult Fischer 344 rats were subjected to chronic social defeat (CSD) for 4 weeks. DMI (10 mg/kg, intraperitoneal (i.p.)) was administered concurrently with CSD or 1 or 2 weeks after cessation of CSD. Sucrose consumption, NET expression, and protein kinases were measured. RESULTS CSD significantly increased messenger RNA (mRNA) and protein levels of NET in the locus coeruleus, as well as NET protein levels in the hippocampus, frontal cortex, and amygdala. These effects were nearly abolished when DMI was administered concurrently with CSD. CSD-induced up-regulation of NET expression in the locus coeruleus, hippocampus, and amygdala lasted at least 2 weeks after cessation of CSD, an effect that was significantly attenuated by 1 or 2 weeks of DMI treatment starting from cessation of the CSD. Concurrent administration of DMI with CSD did not markedly interfere with CSD-induced decreases in protein levels of protein kinases A and C in these brain regions, but it did reverse the CSD-induced reduction in phosphorylated cAMP response element-binding (pCREB) protein levels in most brain regions. CONCLUSION These findings suggest that NET regulation by DMI occurs in both stressed and behaviorally naive rats, and DMI-induced changes in pCREB may be involved.
Collapse
|
29
|
Smaga I, Bystrowska B, Gawliński D, Przegaliński E, Filip M. The endocannabinoid/endovanilloid system and depression. Curr Neuropharmacol 2014; 12:462-74. [PMID: 25426013 PMCID: PMC4243035 DOI: 10.2174/1570159x12666140923205412] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/15/2014] [Accepted: 09/12/2014] [Indexed: 12/20/2022] Open
Abstract
Depression is one of the most frequent causes of disability in the 21st century. Despite the many preclinical and clinical studies that have addressed this brain disorder, the pathophysiology of depression is not well understood and the available antidepressant drugs are therapeutically inadequate in many patients. In recent years, the potential role of lipid-derived molecules, particularly endocannabinoids (eCBs) and endovanilloids, has been highlighted in the pathogenesis of depression and in the action of antidepressants. There are many indications that the eCB/endovanilloid system is involved in the pathogenesis of depression, including the localization of receptors, modulation of monoaminergic transmission, inhibition of the stress axis and promotion of neuroplasticity in the brain. Preclinical pharmacological and genetic studies of eCBs in depression also suggest that facilitating the eCB system exerts antidepressant-like behavioral responses in rodents. In this article, we review the current knowledge of the role of the eCB/endovanilloid system in depression, as well as the effects of its ligands, models of depression and antidepressant drugs in preclinical and clinical settings.
Collapse
Affiliation(s)
- Irena Smaga
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University, College of Medicum, Medyczna 9, PL 30-688 Kraków, Poland
| | - Beata Bystrowska
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University, College of Medicum, Medyczna 9, PL 30-688 Kraków, Poland
| | - Dawid Gawliński
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University, College of Medicum, Medyczna 9, PL 30-688 Kraków, Poland
| | - Edmund Przegaliński
- Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343 Kraków, Poland
| | - Małgorzata Filip
- Department of Toxicology, Faculty of Pharmacy, Jagiellonian University, College of Medicum, Medyczna 9, PL 30-688 Kraków, Poland ; Laboratory of Drug Addiction Pharmacology, Department of Pharmacology, Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, PL 31-343 Kraków, Poland
| |
Collapse
|
30
|
Smaga I, Bystrowska B, Gawliński D, Pomierny B, Stankowicz P, Filip M. Antidepressants and changes in concentration of endocannabinoids and N-acylethanolamines in rat brain structures. Neurotox Res 2014; 26:190-206. [PMID: 24652522 PMCID: PMC4067538 DOI: 10.1007/s12640-014-9465-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/26/2014] [Accepted: 03/06/2014] [Indexed: 12/26/2022]
Abstract
The endocannabinoid (eCB) system has recently been implicated in both the pathogenesis of depression and the action of antidepressants. Here, we investigated the effect of acutely or chronically administering antidepressants [imipramine (IMI) (15 mg/kg), escitalopram (ESC) (10 mg/kg), and tianeptine (10 mg/kg)] on the levels of both eCBs [anandamide (AEA) and 2-arachidonoylglycerol (2-AG)] and N-acylethanolamines (NAEs) [palmitoylethanolamide (PEA) and oleoylethanolamide (OEA)] in various rat brain regions. We also examined the ability of the acute and chronic administration of N-acetylcysteine (NAC) (a mucolytic drug; 100 mg/kg) or URB597 (a fatty acid amide hydrolase inhibitor; 0.3 mg/kg), which have both elicited antidepressant activity in preclinical studies, to affect eCB and NAE levels. Next, we determined whether the observed effects are stable 10 days after the chronic administration of these drugs was halted. We report that the chronic administration of all investigated drugs increased AEA levels in the hippocampus and also increased both AEA and 2-AG levels in the dorsal striatum. NAE levels in limbic regions also increased after treatment with IMI (PEA/OEA), ESC (PEA), and NAC (PEA/OEA). Removing chronic ESC treatment for 10 days affected eCB and NAE levels in the frontal cortex, hippocampus, dorsal striatum, and cerebellum, while a similar tianeptine-free period enhanced accumbal NAE levels. All other drugs maintained their effects after the 10-day washout period. Therefore, the eCB system appears to play a significant role in the mechanism of action of clinically effective and potential antidepressants and may serve as a target for drug design and discovery.
Collapse
Affiliation(s)
- Irena Smaga
- Department of Toxicology, Faculty of Pharmacy, College of Medicum, Jagiellonian University, 9, Medyczna Street, 30-688, Kraków, Poland,
| | | | | | | | | | | |
Collapse
|
31
|
Moreno E, Andradas C, Medrano M, Caffarel MM, Pérez-Gómez E, Blasco-Benito S, Gómez-Cañas M, Pazos MR, Irving AJ, Lluís C, Canela EI, Fernández-Ruiz J, Guzmán M, McCormick PJ, Sánchez C. Targeting CB2-GPR55 receptor heteromers modulates cancer cell signaling. J Biol Chem 2014; 289:21960-72. [PMID: 24942731 DOI: 10.1074/jbc.m114.561761] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The G protein-coupled receptors CB2 (CB2R) and GPR55 are overexpressed in cancer cells and human tumors. Because a modulation of GPR55 activity by cannabinoids has been suggested, we analyzed whether this receptor participates in cannabinoid effects on cancer cells. Here we show that CB2R and GPR55 form heteromers in cancer cells, that these structures possess unique signaling properties, and that modulation of these heteromers can modify the antitumoral activity of cannabinoids in vivo. These findings unveil the existence of previously unknown signaling platforms that help explain the complex behavior of cannabinoids and may constitute new targets for therapeutic intervention in oncology.
Collapse
Affiliation(s)
- Estefanía Moreno
- From the Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, the Institute of Biomedicine of the University of Barcelona, 08028 Barcelona, Spain
| | - Clara Andradas
- the Department of Biochemistry and Molecular Biology I, School of Biology and the Instituto de Investigación Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Mireia Medrano
- From the Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, the Institute of Biomedicine of the University of Barcelona, 08028 Barcelona, Spain
| | - María M Caffarel
- the Department of Biochemistry and Molecular Biology I, School of Biology and
| | - Eduardo Pérez-Gómez
- the Department of Biochemistry and Molecular Biology I, School of Biology and the Instituto de Investigación Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Sandra Blasco-Benito
- the Department of Biochemistry and Molecular Biology I, School of Biology and the Instituto de Investigación Hospital 12 de Octubre, 28041 Madrid, Spain
| | - María Gómez-Cañas
- the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, Department of Biochemistry and Molecular Biology III/Instituto Universitario de Investigación en Neuroquímica, School of Medicine, Complutense University, 28040 Madrid, Spain, the Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - M Ruth Pazos
- the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, Department of Biochemistry and Molecular Biology III/Instituto Universitario de Investigación en Neuroquímica, School of Medicine, Complutense University, 28040 Madrid, Spain
| | - Andrew J Irving
- the Division of Neuroscience, Ninewells Hospital, University of Dundee, Dundee DD1 9SY, United Kingdom, and
| | - Carme Lluís
- From the Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, the Institute of Biomedicine of the University of Barcelona, 08028 Barcelona, Spain
| | - Enric I Canela
- From the Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, the Institute of Biomedicine of the University of Barcelona, 08028 Barcelona, Spain
| | - Javier Fernández-Ruiz
- the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, Department of Biochemistry and Molecular Biology III/Instituto Universitario de Investigación en Neuroquímica, School of Medicine, Complutense University, 28040 Madrid, Spain, the Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Manuel Guzmán
- the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, the Department of Biochemistry and Molecular Biology I, School of Biology and the Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Peter J McCormick
- From the Department of Biochemistry and Molecular Biology, University of Barcelona, 08028 Barcelona, Spain, the Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain, the Institute of Biomedicine of the University of Barcelona, 08028 Barcelona, Spain, the School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, United Kingdom
| | - Cristina Sánchez
- the Department of Biochemistry and Molecular Biology I, School of Biology and the Instituto de Investigación Hospital 12 de Octubre, 28041 Madrid, Spain,
| |
Collapse
|
32
|
McPartland JM, Guy GW, Di Marzo V. Care and feeding of the endocannabinoid system: a systematic review of potential clinical interventions that upregulate the endocannabinoid system. PLoS One 2014; 9:e89566. [PMID: 24622769 PMCID: PMC3951193 DOI: 10.1371/journal.pone.0089566] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 01/21/2014] [Indexed: 12/31/2022] Open
Abstract
Background The “classic” endocannabinoid (eCB) system includes the cannabinoid receptors CB1 and CB2, the eCB ligands anandamide (AEA) and 2-arachidonoylglycerol (2-AG), and their metabolic enzymes. An emerging literature documents the “eCB deficiency syndrome” as an etiology in migraine, fibromyalgia, irritable bowel syndrome, psychological disorders, and other conditions. We performed a systematic review of clinical interventions that enhance the eCB system—ways to upregulate cannabinoid receptors, increase ligand synthesis, or inhibit ligand degradation. Methodology/Principal Findings We searched PubMed for clinical trials, observational studies, and preclinical research. Data synthesis was qualitative. Exclusion criteria limited the results to 184 in vitro studies, 102 in vivo animal studies, and 36 human studies. Evidence indicates that several classes of pharmaceuticals upregulate the eCB system, including analgesics (acetaminophen, non-steroidal anti-inflammatory drugs, opioids, glucocorticoids), antidepressants, antipsychotics, anxiolytics, and anticonvulsants. Clinical interventions characterized as “complementary and alternative medicine” also upregulate the eCB system: massage and manipulation, acupuncture, dietary supplements, and herbal medicines. Lifestyle modification (diet, weight control, exercise, and the use of psychoactive substances—alcohol, tobacco, coffee, cannabis) also modulate the eCB system. Conclusions/Significance Few clinical trials have assessed interventions that upregulate the eCB system. Many preclinical studies point to other potential approaches; human trials are needed to explore these promising interventions.
Collapse
Affiliation(s)
- John M. McPartland
- GW Pharmaceuticals, Porton Down Science Park, Salisbury, Wiltshire, United Kingdom
- Department of Family Medicine, University of Vermont, Burlington, Vermont, United States of America
- * E-mail:
| | - Geoffrey W. Guy
- GW Pharmaceuticals, Porton Down Science Park, Salisbury, Wiltshire, United Kingdom
| | - Vincenzo Di Marzo
- Endocannabinoid Research Group, Istituto di Chimica Biomoleculare, CNR, Via Campi Flegrei, Pozzuoli, Napoli, Italy
| |
Collapse
|
33
|
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: 9.2] [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
|
34
|
Hillard CJ, Liu QS. Endocannabinoid signaling in the etiology and treatment of major depressive illness. Curr Pharm Des 2014; 20:3795-811. [PMID: 24180398 PMCID: PMC4002665 DOI: 10.2174/13816128113196660735] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/23/2013] [Indexed: 12/28/2022]
Abstract
The purpose of this review is to examine human and preclinical data that are relevant to the following hypotheses. The first hypothesis is that deficient CB1R-mediated signaling results in symptoms that mimic those seen in depression. The second hypothesis is that activation of CB1R-mediated signaling results in behavioral, endocrine and other effects that are similar to those produced by currently used antidepressants. The third hypothesis is that conventional antidepressant therapies act through enhanced CB1R mediated signaling. Together the available data indicate that activators of CB1R signaling, particularly inhibitors of fatty acid amide hydrolase, should be considered for clinical trials for the treatment of depression.
Collapse
MESH Headings
- Animals
- Antidepressive Agents/pharmacology
- Antidepressive Agents/therapeutic use
- Brain/drug effects
- Brain/enzymology
- Brain/metabolism
- Brain/pathology
- Cannabis
- Depressive Disorder, Major/drug therapy
- Depressive Disorder, Major/etiology
- Depressive Disorder, Major/genetics
- Depressive Disorder, Major/pathology
- Disease Models, Animal
- Endocannabinoids/genetics
- Endocannabinoids/metabolism
- Humans
- Magnetic Resonance Imaging
- Neurogenesis/drug effects
- Polymorphism, Genetic
- Receptor, Cannabinoid, CB1/genetics
- Receptor, Cannabinoid, CB1/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
| | - Qing-song Liu
- Neuroscience Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226.
| |
Collapse
|
35
|
Fogaça MV, Galve-Roperh I, Guimarães FS, Campos AC. Cannabinoids, Neurogenesis and Antidepressant Drugs: Is there a Link? Curr Neuropharmacol 2013; 11:263-75. [PMID: 24179463 PMCID: PMC3648779 DOI: 10.2174/1570159x11311030003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/14/2012] [Accepted: 01/04/2013] [Indexed: 11/22/2022] Open
Abstract
Similar to clinically used antidepressants, cannabinoids can also regulate anxiety and depressive symptoms. Although the mechanisms of these effects are not completely understood, recent evidence suggests that changes in endocannabinoid system could be involved in some actions of antidepressants. Chronic antidepressant treatment modifies the expression of CB1 receptors and endocannabinoid (EC) content in brain regions related to mood and anxiety control. Moreover, both antidepressant and cannabinoids activate mitogen-activated protein (MAP) kinase and phosphoinositide 3-kinase(PI3-K)/Akt or PKB signaling, intracellular pathways that regulate cell proliferation and neural cell survival. Facilitation of hippocampal neurogenesis is proposed as a common effect of chronic antidepressant treatment. Genetic or pharmacological manipulations of cannabinoid receptors (CB1 and CB2) or enzymes responsible for endocannabinoid-metabolism have also been shown to control proliferation and neurogenesis in the hippocampus. In the present paper we reviewed the studies that have investigated the potential contribution of cannabinoids and neurogenesisto antidepressant effects. Considering the widespread brain distribution of the EC system, a better understanding of this possible interaction could contribute to the development of therapeutic alternatives to mood and anxiety disorders.
Collapse
Affiliation(s)
- Manoela Viar Fogaça
- Department of Pharmacology; School of Medicine of RibeirãoPreto- University of São Paulo, Brazil ; Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), University of São Paulo, Brazil
| | | | | | | |
Collapse
|
36
|
Neural plasticity and proliferation in the generation of antidepressant effects: hippocampal implication. Neural Plast 2013; 2013:537265. [PMID: 23862076 PMCID: PMC3703717 DOI: 10.1155/2013/537265] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/01/2013] [Accepted: 05/08/2013] [Indexed: 12/15/2022] Open
Abstract
It is widely accepted that changes underlying depression and antidepressant-like effects involve not only alterations in the levels of neurotransmitters as monoamines and their receptors in the brain, but also structural and functional changes far beyond. During the last two decades, emerging theories are providing new explanations about the neurobiology of depression and the mechanism of action of antidepressant strategies based on cellular changes at the CNS level. The neurotrophic/plasticity hypothesis of depression, proposed more than a decade ago, is now supported by multiple basic and clinical studies focused on the role of intracellular-signalling cascades that govern neural proliferation and plasticity. Herein, we review the state-of-the-art of the changes in these signalling pathways which appear to underlie both depressive disorders and antidepressant actions. We will especially focus on the hippocampal cellularity and plasticity modulation by serotonin, trophic factors as brain-derived neurotrophic factor (BDNF), and vascular endothelial growth factor (VEGF) through intracellular signalling pathways—cAMP, Wnt/β-catenin, and mTOR. Connecting the classic monoaminergic hypothesis with proliferation/neuroplasticity-related evidence is an appealing and comprehensive attempt for improving our knowledge about the neurobiological events leading to depression and associated to antidepressant therapies.
Collapse
|
37
|
Continuous GSK-3β overexpression in the hippocampal dentate gyrus induces prodepressant-like effects and increases sensitivity to chronic mild stress in mice. J Affect Disord 2013; 146:45-52. [PMID: 23021826 DOI: 10.1016/j.jad.2012.08.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 08/21/2012] [Accepted: 08/21/2012] [Indexed: 01/07/2023]
Abstract
BACKGROUND Glycogen synthase kinase-3 (GSK-3) has been linked to prodepressant-like effects in rodents. However, the roles of GSK-3β and the hippocampal dentate gyrus in regulating these behavioral effects remain unclear. METHODS A lentiviral vector was utilized to site-specifically express GSK-3β constitutively in the hippocampal DG in a mouse model of CMS. We examine the forced swim, tail suspension and the sucrose intake test. Acute and chronic administrations were conducted by dissolving fluoxetine hydrochloride (10ml/kg). We examine behavior tests as before, cellular apoptosis, proliferation and differentiation in the hippocampus. RESULTS GSK-3β expression levels persistently and significantly increased in the hippocampus following lenti-GSK-3β injections. In mice previously exposed to CMS, pre-injection of lentivirus-expressing GSK-3β into the hippocampal dentate gyrus significantly decreased sucrose preferences in the sucrose intake test and increased immobility times in both forced swim and tail suspension tests. In addition, fluoxetine resulted in similar antidepressant-like effects following chronic, but not acute, administrations under the same experimental conditions. Cellular apoptosis was observed in the hippocampal DG using TUNEL, revealing many TUNEL-positive cells in the lenti-GSK-3β mice. There were no significant changes in proliferation and differentiation. LIMITATIONS We did not measure more biomarkers which were regulated by GSK-3β. CONCLUSIONS Results from this study demonstrated that site-specific injection of a lentivirus induced continuous GSK-3β expression in the hippocampal dentate gyrus of mice, resulting in prodepressant-like effects and increased sensitivity to chronic mild stress. Furthermore, chronic fluoxetine administration reversed these prodepressant-like effects and decreased neuronal apoptosis in the hippocampal DG in GSK-3β-overexpressing mice.
Collapse
|
38
|
Campos AC, de Paula Soares V, Carvalho MC, Ferreira FR, Vicente MA, Brandão ML, Zuardi AW, Zangrossi H, Guimarães FS. Involvement of serotonin-mediated neurotransmission in the dorsal periaqueductal gray matter on cannabidiol chronic effects in panic-like responses in rats. Psychopharmacology (Berl) 2013; 226:13-24. [PMID: 23007604 DOI: 10.1007/s00213-012-2878-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 09/06/2012] [Indexed: 11/30/2022]
Abstract
RATIONALE Cannabidiol (CBD) is a non-psychotomimetic constituent of Cannabis sativa plant that promotes antianxiety and anti-panic effects in animal models after acute systemic or intra-dorsal periaqueductal gray (DPAG) administration. However, the effects of CBD repeated administration, and the possible mechanisms involved, in animal models of anxiety- and panic-related responses remain poorly understood. OBJECTIVE The present study evaluates the role of the serotonergic neurotransmission within the DPAG in the modulation of escape responses of rats chronically treated with CBD. METHODS Male Wistar rats received acute or repeated (5 mg/Kg/daily/21 days) administration of CBD and were submitted to the elevated T-maze (ETM). We also investigated if CBD effects on the ETM depend on facilitation of 5-HT1A-mediated neurotransmission in the DPAG. To this latter aim, we verified if these effects would be prevented by intra-DPAG injection of the 5-HT1A receptor antagonist WAY100635 (0.37 nmol/0.2 μL). Also, we verified, by in vivo microdialysis, if CBD chronic treatment increases serotonin (5-HT) release and, by quantitative polymerase chain reaction, if there are changes in 5HT-1A or 5HT-2C mRNA expression in DPAG. RESULTS The results showed that repeated but not acute peripheral administration of CBD decreases escape responses in the ETM, suggesting a panicolytic effect. This treatment did not change 5HT-1A or 5-HT-2C receptor mRNA expression nor modify serotonin extracellular concentrations in the DPAG. CBD effects were prevented by DPAG injection of the 5-HT1A receptor antagonist. CONCLUSIONS Together, these findings suggest that repeated treatment with CBD induces anti-panic effects by acting on 5-HT1A receptors in DPAG.
Collapse
Affiliation(s)
- Alline Cristina Campos
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, 3900 Bandeirantes av., Monte Alegre, Ribeirão Preto, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Effects of fluoxetine on CRF and CRF1 expression in rats exposed to the learned helplessness paradigm. Psychopharmacology (Berl) 2013; 225:647-59. [PMID: 22960774 DOI: 10.1007/s00213-012-2859-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 08/16/2012] [Indexed: 12/25/2022]
Abstract
RATIONALE Stress is a common antecedent reported by people suffering major depression. In these patients, extrahypothalamic brain areas, like the hippocampus and basolateral amygdala (BLA), have been found to be affected. The BLA synthesizes CRF, a mediator of the stress response, and projects to hippocampus. The main hippocampal target for this peptide is the CRF subtype 1 receptor (CRF1). Evidence points to a relationship between dysregulation of CRF/CRF1 extrahypothalamic signaling and depression. OBJECTIVE Because selective serotonin reuptake inhibitors (SSRIs) are the first-line pharmacological treatment for depression, we investigated the effect of chronic treatment with the SSRI fluoxetine on long-term changes in CRF/CRF1 signaling in animals showing a depressive-like behavior. METHODS Male Wistar rats were exposed to the learned helplessness paradigm (LH). After evaluation of behavioral impairment, the animals were treated with fluoxetine (10 mg/kg i.p.) or saline for 21 days. We measured BLA CRF expression with RT-PCR and CRF1 expression in CA3 and the dentate gyrus of the hippocampus with in situ hybridization. We also studied the activation of one of CRF1's major intracellular signaling targets, the extracellular signal-related kinases 1 and 2 (ERK1/2) in CA3. RESULTS In saline-treated LH animals, CRF expression in the BLA increased, while hippocampal CRF1 expression and ERK1/2 activation decreased. Treatment with fluoxetine reversed the changes in CRF and CRF1 expressions, but not in ERK1/2 activation. CONCLUSION In animals exposed to the learned helplessness paradigm, there are long-term changes in CRF and CRF1 expression that are restored with a behaviorally effective antidepressant treatment.
Collapse
|
40
|
The Effects of Reboxetine Treatment on Depression-like Behavior, Brain Neurotrophins, and ERK Expression in Rats Exposed to Chronic Mild Stress. J Mol Neurosci 2012; 50:88-97. [DOI: 10.1007/s12031-012-9872-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 08/01/2012] [Indexed: 10/27/2022]
|
41
|
Early intervention with fluoxetine reverses abnormalities in the serotonergic system and behavior of rats exposed prenatally to dexamethasone. Neuropharmacology 2012; 63:292-300. [DOI: 10.1016/j.neuropharm.2012.03.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 03/29/2012] [Accepted: 03/31/2012] [Indexed: 11/19/2022]
|
42
|
Esteban S, García-Sevilla JA. Effects induced by cannabinoids on monoaminergic systems in the brain and their implications for psychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2012; 38:78-87. [PMID: 22133541 DOI: 10.1016/j.pnpbp.2011.11.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Revised: 11/14/2011] [Accepted: 11/14/2011] [Indexed: 12/19/2022]
Abstract
The endocannabinoid system and CB(1) receptors participate in the control of emotional behavior and mood through a functional coupling with the classic monoaminergic systems. In general, the acute stimulation of CB(1) receptors increases the activity (spontaneous firing rate) of noradrenergic (NE), serotonergic (5-HT) and dopaminergic (DA) neurons as well as the synthesis and/or release of the corresponding neurotransmitter in specific brain regions. Notably, the antagonist/inverse agonist rimonabant (SR141617A) can decrease the basal activity of NE and 5-HT neurons, suggesting a tonic/constitutive regulation of these neuronal systems by endocannabinoids acting at CB(1) receptors. Monoaminergic systems are modulated via CB(1) receptors by direct or indirect effects depending on the localization of this inhibitory receptor, which can be present on monoaminergic neurons themselves and/or inhibitory (GABAergic) and/or excitatory (glutamatergic) regulatory neurons. The repeated stimulation of CB(1) receptors is not associated with the induction of tolerance (receptor desensitization) on the activity of NE, 5-HT and DA neurons, in contrast to chronic agonist effects on neurotransmitter synthesis and/or release in some brain regions. CB(1) receptor desensitization may alter the direct and/or indirect effects of cannabinoid drugs modulating the functionality of monoaminergic systems. The sustained activation of monoaminergic neurons by cannabinoid drugs can also be related to changes in the function of presynaptic inhibitory α(2)-adrenoceptors or 5-HT(1A) receptors (autoreceptors and heteroreceptors), whose sensitivity is downregulated or upregulated upon chronic CB(1) agonist exposure. The functional interactions between endocannabinoids and monoaminergic systems in the brain indicate a potential role for CB(1) receptor signaling in the neurobiology of various psychiatric disorders, including major depression and schizophrenia as the major syndromes.
Collapse
Affiliation(s)
- Susana Esteban
- Laboratorio de Neurofarmacología, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears (UIB), Palma de Mallorca, Spain.
| | | |
Collapse
|
43
|
Choi MR, Oh DH, Kim SH, Jung KH, Das ND, Chai YG. Fluoxetine increases the expression of NCAM140 and pCREB in rat C6 glioma cells. Psychiatry Investig 2012; 9:180-6. [PMID: 22707970 PMCID: PMC3372567 DOI: 10.4306/pi.2012.9.2.180] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 06/11/2011] [Accepted: 07/27/2011] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Dysfunction of neural plasticity in the brain is known to alter neural networks, resulting in depression. To understand how fluoxetine regulates molecules involved in neural plasticity, the expression levels of NCAM, NCAM140, CREB and pCREB, in rat C6 glioma cells after fluoxetine treatment were examined. METHODS C6 cells were cultured after 20 min or after 6, 24 or 72 h treatments with 10 µM fluoxetine. Immunocytochemistry was used to determine the effect of fluoxetine on the expression of NCAM. Western blot analysis was used to measure the expression levels of NCAM140 and CREB and the induction of pCREB after fluoxetine treatment. RESULTS NCAM expression following 72-h fluoxetine treatment was significantly increased around cell membranes compared to control cells. Cells treated with fluoxetine for 6 and 72 h showed a significant increase in NCAM140 expression compared to cells treated for 20 min. The level of pCREB in the cells treated with fluoxetine for 72 h not only increased more than 60%, but was also significantly different when compared with the other treatment times. The 72-h fluoxetine treatment led to the increase of NCAM140 and the phosphorylation of CREB in C6 cells. CONCLUSION Our findings indicate that fluoxetine treatment regulates neuronal plasticity and neurite outgrowth by phosphorylating and activating CREB via the NCAM140 homophilic interaction-induced activation of the Ras-MAPK pathway.
Collapse
Affiliation(s)
- Mi Ran Choi
- Division of Molecular and Life Science, Hanyang University, Ansan, Korea
| | - Dong Hoon Oh
- Department of Neuropsychiatry, College of Medicine and Institute of Mental Health, Hanyang University, Seoul, Korea
| | - Seok Hyeon Kim
- Department of Neuropsychiatry, College of Medicine and Institute of Mental Health, Hanyang University, Seoul, Korea
| | - Kyoung Hwa Jung
- Division of Molecular and Life Science, Hanyang University, Ansan, Korea
| | - Nando Dulal Das
- Division of Molecular and Life Science, Hanyang University, Ansan, Korea
| | - Young Gyu Chai
- Division of Molecular and Life Science, Hanyang University, Ansan, Korea
| |
Collapse
|
44
|
Li HF, Yu X, Yang K, He CY, Kou SJ, Cao SX, Xie GR. The relationship between single nucleotide polymorphisms in 5-HT2A signal transduction-related genes and the response efficacy to selective serotonin reuptake inhibitor treatments in Chinese patients with major depressive disorder. Genet Test Mol Biomarkers 2012; 16:667-71. [PMID: 22480177 DOI: 10.1089/gtmb.2011.0232] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE To explore the possible relationship between six single nucleotide polymorphisms (SNPs) (rs6311 and rs6305 of 5-HT2A, rs5443 of Gβ3, rs2230739 of ACDY9, rs1549870 of PDE1A and rs255163 of CREB1, which are all related with 5-HT2A the signal transduction pathway) and the response efficacy to selective serotonin reuptake inhibitor (SSRI) treatments in major depressive disorder (MDD) Chinese. METHODS This study included 194 depressed patients to investigate the influence of 6 polymorphisms in 5-HT2A signal transduction-related genes on the efficacy of SSRIs assessed over 1 year. The efficacies of SSRIs on 194 MDD patients were evaluated in an 8-week open-trial study. Over 1 year, a follow-up study was completed for 174 of them to observe the long-term efficacy of SSRIs. The optimal-scaling regression analysis was used for testing the relationship between the different genotypes of five SNPs and the efficacy in MDD. RESULTS It showed that the patients with rs5443TT and rs2230739GG have a relatively good efficacy in response to short-term SSRIs. We also found that good efficacy appeared in depressed patients with rs2230739GG in response to long-term SSRIs. CONCLUSIONS It suggested that different genotypes of rs5443 and rs2230739 might influence the signal transduction pathways of second message and affect therapeutic efficacy.
Collapse
Affiliation(s)
- Heng-Fen Li
- Department of Psychiatry, the First Affiliated Hospital, Zhengzhou University, Zhengzhou, P.R. China
| | | | | | | | | | | | | |
Collapse
|
45
|
Valdizán EM, Díaz A, Pilar-Cuéllar F, Lantero A, Mostany R, Villar AV, Laorden ML, Hurlé MA. Chronic treatment with the opioid antagonist naltrexone favours the coupling of spinal cord μ-opioid receptors to Gαz protein subunits. Neuropharmacology 2011; 62:757-64. [PMID: 21903117 DOI: 10.1016/j.neuropharm.2011.08.029] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2011] [Revised: 08/03/2011] [Accepted: 08/19/2011] [Indexed: 11/30/2022]
Abstract
Sustained administration of opioid antagonists to rodents results in an enhanced antinociceptive response to agonists. We investigated the changes in spinal μ-opioid receptor signalling underlying this phenomenon. Rats received naltrexone (120 μg/h; 7 days) via osmotic minipumps. The antinociceptive response to the μ-agonist sufentanil was tested 24 h after naltrexone withdrawal. In spinal cord samples, we determined the interaction of μ-receptors with Gα proteins (agonist-stimulated [(35)S]GTPγS binding and immunoprecipitation of [(35)S]GTPγS-labelled Gα subunits) as well as μ-opioid receptor-dependent inhibition of the adenylyl cyclase (AC) activity. Chronic naltrexone treatment augmented DAMGO-stimulated [(35)S]GTPγS binding, potentiated the inhibitory effect of DAMGO on the AC/cAMP pathway, and increased the inverse agonist effect of naltrexone on cAMP accumulation. In control rats, the inhibitory effect of DAMGO on cAMP production was antagonized by pertussis toxin (PTX) whereas, after chronic naltrexone, the effect became resistant to the toxin, suggesting a coupling of μ-receptors to PTX-insensitive Gα(z) subunits. Immunoprecipitation assays confirmed the transduction switch from Gα(i/o) to Gα(z) proteins. The consequence was an enhancement of the antinociceptive response to sufentanil that, in consonance with the neurochemical data, was prevented by Gα(z)-antisense oligodeoxyribonucleotides but not by PTX. Such changes in opioid receptor signalling can be a double-edged sword. On the one hand, they may have potential applicability to the optimisation of the analgesic effects of opioid drugs for the control of pain. On the other hand, they represent an important homeostatic dysregulation of the endogenous opioid system that might account for undesirable effects in patients chronically treated with opioid antagonists. This article is part of a Special Issue entitled 'Post-Traumatic Stress Disorder'.
Collapse
Affiliation(s)
- Elsa M Valdizán
- Departamento de Fisiología y Farmacología, Facultad de Medicina, Universidad de Cantabria, 39011 Santander, Cantabria, Spain
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Putative role of endocannabinoid signaling in the etiology of depression and actions of antidepressants. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:1575-85. [PMID: 21111017 DOI: 10.1016/j.pnpbp.2010.11.021] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2010] [Revised: 11/15/2010] [Accepted: 11/15/2010] [Indexed: 11/23/2022]
Abstract
In the last few years, there have been several advances in the determination of the role of the endocannabinoid system in the etiology of depression and the functional actions of antidepressant drugs. Specifically, a deficiency in endocannabinoid signaling is sufficient to produce a "depressive-like" phenotype at the preclinical level (including changes in rewarding, emotional and cognitive behavior and biological changes such as increased HPA axis activity, impaired stress adaptation, reduced neurogenesis and altered serotonin negative feedback), and capable of inducing symptoms of depression in humans at a clinical level. In line with these findings, clinical populations diagnosed with depression are found to have reduced levels of circulating endocannabinoids and preclinical models of depression reveal a deficit in central endocannabinoid signaling. Moreover, facilitation of endocannabinoid signaling is sufficient to produce all of the behavioral and biochemical effects of conventional antidepressant treatments. Further, many forms of antidepressant treatments significantly alter endocannabinoid signaling, and in some of these cases this recruitment of endocannabinoid signaling is involved in the neuroadaptive effects of these treatments. Ultimately, these data present a compelling picture of the putative role of the endocannabinoid system in the processes subserving both the development and treatment of depression.
Collapse
|
47
|
Nakajima J, Takahashi M, Nonaka R, Seto T, Suzuki J, Yoshida M, Kanai C, Hamano T. Identification and quantitation of a benzoylindole (2-methoxyphenyl)(1-pentyl-1H-indol-3-yl)methanone and a naphthoylindole 1-(5-fluoropentyl-1H-indol-3-yl)-(naphthalene-1-yl)methanone (AM-2201) found in illegal products obtained via the Internet and their cannabimimetic effects evaluated by in vitro [35S]GTPγS binding assays. Forensic Toxicol 2011. [DOI: 10.1007/s11419-011-0114-5] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
48
|
The Effects of Fluoxetine Treatment in a Chronic Mild Stress Rat Model on Depression-Related Behavior, Brain Neurotrophins and ERK Expression. J Mol Neurosci 2011; 45:246-55. [DOI: 10.1007/s12031-011-9515-5] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 03/08/2011] [Indexed: 12/25/2022]
|
49
|
Rodrigues LCDM, Conti CL, Nakamura-Palacios EM. Clozapine and SCH 23390 prevent the spatial working memory disruption induced by Δ9-THC administration into the medial prefrontal cortex. Brain Res 2011; 1382:230-7. [PMID: 21281616 DOI: 10.1016/j.brainres.2011.01.069] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 01/18/2011] [Accepted: 01/20/2011] [Indexed: 01/07/2023]
Abstract
Marijuana (Cannabis sativa) is one of the most widely used illicit drugs in the world. Its use is associated with impairments in cognitive function. We previously reported that Δ(9)-tetrahydrocannabinol (Δ(9)-THC), the primary psychoactive component of marijuana, impaired spatial working memory in the radial maze task when injected intracortically (IC) into the medial prefrontal cortex (mPFC) of rats. Here, we used this paradigm to evaluate the involvement of prefrontal dopamine receptors in working memory disruption induced by Δ(9)-THC. Intracortical pre-treatment of animals with either the D(1)- or D(2)-like dopamine receptor antagonists SCH 23390 or clozapine, respectively, significantly reduced the number of errors rats made in the radial maze following treatment with Δ(9)-THC also administered intracortically. These results were obtained in the absence of locomotor impairment, as evidenced by the time spent in each arm a rat visited. Our findings suggest that prefrontal dopamine receptors are involved in Δ(9)-THC-induced disruption of spatial working memory. This interaction between the cannabinoid system and dopamine release in the PFC contributes to new directions in research and to treatments for cognitive dysfunctions associated with drug abuse and dependence.
Collapse
|
50
|
Longone P, di Michele F, D’Agati E, Romeo E, Pasini A, Rupprecht R. Neurosteroids as neuromodulators in the treatment of anxiety disorders. Front Endocrinol (Lausanne) 2011; 2:55. [PMID: 22654814 PMCID: PMC3356011 DOI: 10.3389/fendo.2011.00055] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 10/03/2011] [Indexed: 11/22/2022] Open
Abstract
Anxiety disorders are the most common psychiatric disorders. They are frequently treated with benzodiazepines, which are fast acting highly effective anxiolytic agents. However, their long-term use is impaired by tolerance development and abuse liability. In contrast, antidepressants such as selective serotonin reuptake inhibitors (SSRIs) are considered as first-line treatment but have a slow onset of action. Neurosteroids are powerful allosteric modulators of GABA(A) and glutamate receptors. However, they also modulate sigma receptors and they are modulated themselves by SSRIs. Both pre-clinical and clinical studies have shown that neurosteroid homeostasis is altered in depression and anxiety disorders and antidepressants may act in part through restoring neurosteroid disbalance. Moreover, novel drugs interfering with neurosteroidogenesis such as ligands of the translocator protein (18 kDa) may represent an attractive pharmacological option for novel anxiolytics which lack the unwarranted side effects of benzodiazepines. Thus, neurosteroids are important endogenous neuromodulators for the physiology and pathophysiology of anxiety and they may constitute a novel therapeutic approach in the treatment of these disorders.
Collapse
Affiliation(s)
- Patrizia Longone
- Molecular Neurobiology Unit, Experimental NeurologyFondazione Santa Lucia, Rome, Italy
- *Correspondence: Patrizia Longone, Molecular Neurobiology Unit, Room 201, Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy. e-mail: ; Rainer Rupprecht, Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053 Regensburg, Germany. e-mail:
| | | | - Elisa D’Agati
- Unit of Child Neurology and Psychiatry, Department of Neuroscience, University of Rome “Tor Vergata,”Rome, Italy
| | - Elena Romeo
- Department of Neuroscience, University of Rome “Tor Vergata,”Rome, Italy
| | - Augusto Pasini
- Unit of Child Neurology and Psychiatry, Department of Neuroscience, University of Rome “Tor Vergata,”Rome, Italy
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy, University RegensburgRegensburg, Germany
- *Correspondence: Patrizia Longone, Molecular Neurobiology Unit, Room 201, Fondazione Santa Lucia, Via del Fosso di Fiorano 64, 00143 Rome, Italy. e-mail: ; Rainer Rupprecht, Department of Psychiatry and Psychotherapy, University Regensburg, Universitätsstrasse 84, 93053 Regensburg, Germany. e-mail:
| |
Collapse
|