1
|
Sharp T, Ippolito A. Neuropsychopharmacology of hallucinogenic and non-hallucinogenic 5-HT 2A receptor agonists. Br J Pharmacol 2025. [PMID: 40405723 DOI: 10.1111/bph.70050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/13/2025] [Accepted: 02/02/2025] [Indexed: 05/24/2025] Open
Abstract
Psychedelic drugs such as LSD and psilocin were once relegated to the fringes of medical research because of their association with counterculture movements and a perceived concern about harm through recreational use, and their consequent legal prohibition in the early 1970s. However, these drugs are now experiencing a renaissance in the field of psychiatry based on increasing evidence that they can produce long-lasting improvements in health across a wide variety of mental illnesses, including major depression, addictions and anxiety disorders. These drugs interact with many different 5-HT receptor subtypes but the powerful psychedelic experience, which (depending on set and setting) includes profound alterations in perception, mood and cognition, accompanied by vivid hallucinations, is now widely considered mediated by an agonist action at 5-HT2A receptors. However, the link between the psychedelic experience, 5-HT2A receptor agonism and therapeutic effects is currently uncertain. Indeed, recent research has revealed a new class of 5-HT2A receptor agonists which appear to retain the therapeutic potential of psychedelics drugs without inducing disorienting hallucinatory experiences. Biased signalling, partial agonism and non-selectivity at the 5-HT2A receptor are amongst the possible explanations for the differential properties of these drugs, whereas increased neuroplasticity offers a likely account of their common therapeutic effects. This article explores the neuropsychopharmacological properties of hallucinogenic and non-hallucinogenic 5-HT2A receptor agonists in the context of their promise as novel drug treatments in psychiatry.
Collapse
Affiliation(s)
- Trevor Sharp
- University Department of Pharmacology, Oxford, UK
| | | |
Collapse
|
2
|
Mottarlini F, Caffino L, Fumagalli F, Calabrese F, Brivio P. NeuropsychopharmARCology: Shaping Neuroplasticity through Arc/ Arg3.1 Modulation. Curr Neuropharmacol 2025; 23:650-670. [PMID: 39473108 PMCID: PMC12163496 DOI: 10.2174/011570159x338335240903075655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 11/05/2024] Open
Abstract
Activity-regulated cytoskeleton-associated protein (aka activity-regulated gene Arg3.1) belongs to the effector gene family of the immediate early genes. This family encodes effector proteins, which act directly on cellular homeostasis and function. Arc/Arg3.1 is localized at dendritic processes, allowing the protein local synthesis on demand, and it is considered a reliable index of activity- dependent synaptic changes. Evidence also exists showing the critical role of Arc/Arg3.1 in memory processes. The high sensitivity to changes in neuronal activity, its specific localization as well as its involvement in long-term synaptic plasticity indeed make this effector gene a potential, critical target of the action of psychotropic drugs. In this review, we focus on antipsychotic and antidepressant drugs as well as on psychostimulants, which belong to the category of drugs of abuse but can also be used as drugs for specific disorders of the central nervous system (i.e., Attention Deficit Hyperactivity Disorder). It is demonstrated that psychotropic drugs with different mechanisms of action converge on Arc/Arg3.1, providing a means whereby Arc/Arg3.1 synaptic modulation may contribute to their therapeutic activity. The potential translational implications for different neuropsychiatric conditions are also discussed, recognizing that the treatment of these disorders is indeed complex and involves the simultaneous regulation of several dysfunctional mechanisms.
Collapse
Affiliation(s)
- Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Francesca Calabrese
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| | - Paola Brivio
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’, Università degli Studi di Milano, Via Balzaretti 9, 20133 Milano, Italy
| |
Collapse
|
3
|
Kolasa M, Nikiforuk A, Korlatowicz A, Solich J, Potasiewicz A, Dziedzicka-Wasylewska M, Bugno R, Hogendorf A, Bojarski A, Faron-Górecka A. Unraveling psilocybin's therapeutic potential: behavioral and neuroplasticity insights in Wistar-Kyoto and Wistar male rat models of treatment-resistant depression. Psychopharmacology (Berl) 2024:10.1007/s00213-024-06644-3. [PMID: 38963553 DOI: 10.1007/s00213-024-06644-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
RATIONALE Our study aimed to unravel the unknown mechanisms behind the exceptional efficacy of Psilocybin (PSI) in treating treatment-resistant depression (TRD). Focusing on Wistar-Kyoto (WKY) rats with a TRD phenotype and Wistar (WIS) rats as a normative comparison, we investigated behavioral and neuroplasticity-related responses to PSI, striving to shed light on the distinctive features of its antidepressant effects. OBJECTIVES We set out to assess the behavioral impact of acute and prolonged PSI administration on WKY and WIS rats, employing Novel Object Recognition (NORT), Social Interaction (SI), and Forced Swimming Test (FST). Our secondary objectives involved exploring strain-specific alterations in neuroplasticity-related parameters, including brain-derived neurotrophic factor (BDNF) and activity-regulated cytoskeleton-associated protein (Arc). METHODS Conducting post-acute and extended assessments after a single PSI administration, we applied behavioral tests and biochemical analyses to measure serum BDNF levels and neuroplasticity-related parameters in the prefrontal cortex. Statistical analyses were deployed to discern significant differences between the rat strains and assess the impact of PSI on behavioral and biochemical outcomes. RESULTS Our findings uncovered significant behavioral disparities between WKY and WIS rats, indicating passive behavior and social withdrawal in the former. PSI demonstrated pronounced pro-social and antidepressant effects in both strains, each with its distinctive temporal trajectory. Notably, we identified strain-specific variations in BDNF-related signaling and observed the modulation of Arc expression in WKY rats. CONCLUSIONS Our study delineated mood-related behavioral nuances between WKY and WIS rat strains, underscoring the antidepressant and pro-social properties of PSI in both groups. The distinct temporal patterns of observed changes and the identified strain-specific neuroplasticity alterations provide valuable insights into the TRD phenotype and the mechanisms underpinning the efficacy of PSI.
Collapse
Affiliation(s)
- Magdalena Kolasa
- Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Agnieszka Nikiforuk
- Department of Behavioral Neuroscience & Drug Development, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Agata Korlatowicz
- Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Joanna Solich
- Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Agnieszka Potasiewicz
- Department of Behavioral Neuroscience & Drug Development, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | | | - Ryszard Bugno
- Department of Medicinal Chemistry, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Adam Hogendorf
- Department of Medicinal Chemistry, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Andrzej Bojarski
- Department of Medicinal Chemistry, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Agata Faron-Górecka
- Department of Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland.
| |
Collapse
|
4
|
Hatzipantelis CJ, Olson DE. The Effects of Psychedelics on Neuronal Physiology. Annu Rev Physiol 2024; 86:27-47. [PMID: 37931171 PMCID: PMC10922499 DOI: 10.1146/annurev-physiol-042022-020923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Psychedelics are quite unique among drugs that impact the central nervous system, as a single administration of a psychedelic can both rapidly alter subjective experience in profound ways and produce sustained effects on circuits relevant to mood, fear, reward, and cognitive flexibility. These remarkable properties are a direct result of psychedelics interacting with several key neuroreceptors distributed across the brain. Stimulation of these receptors activates a variety of signaling cascades that ultimately culminate in changes in neuronal structure and function. Here, we describe the effects of psychedelics on neuronal physiology, highlighting their acute effects on serotonergic and glutamatergic neurotransmission as well as their long-lasting effects on structural and functional neuroplasticity in the cortex. We propose that the neurobiological changes leading to the acute and sustained effects of psychedelics might be distinct, which could provide opportunities for engineering compounds with optimized safety and efficacy profiles.
Collapse
Affiliation(s)
- Cassandra J Hatzipantelis
- Institute for Psychedelics and Neurotherapeutics, University of California, Davis, Davis, California, USA;
- Department of Chemistry, University of California, Davis, Davis, California, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
| | - David E Olson
- Institute for Psychedelics and Neurotherapeutics, University of California, Davis, Davis, California, USA;
- Department of Chemistry, University of California, Davis, Davis, California, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA
- Center for Neuroscience, University of California, Davis, Davis, California, USA
| |
Collapse
|
5
|
Heifets BD, Olson DE. Therapeutic mechanisms of psychedelics and entactogens. Neuropsychopharmacology 2024; 49:104-118. [PMID: 37488282 PMCID: PMC10700553 DOI: 10.1038/s41386-023-01666-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/26/2023]
Abstract
Recent clinical and preclinical evidence suggests that psychedelics and entactogens may produce both rapid and sustained therapeutic effects across several indications. Currently, there is a disconnect between how these compounds are used in the clinic and how they are studied in preclinical species, which has led to a gap in our mechanistic understanding of how these compounds might positively impact mental health. Human studies have emphasized extra-pharmacological factors that could modulate psychedelic-induced therapeutic responses including set, setting, and integration-factors that are poorly modelled in current animal experiments. In contrast, animal studies have focused on changes in neuronal activation and structural plasticity-outcomes that are challenging to measure in humans. Here, we describe several hypotheses that might explain how psychedelics rescue neuropsychiatric disease symptoms, and we propose ways to bridge the gap between human and rodent studies. Given the diverse pharmacological profiles of psychedelics and entactogens, we suggest that their rapid and sustained therapeutic mechanisms of action might best be described by the collection of circuits that they modulate rather than their actions at any single molecular target. Thus, approaches focusing on selective circuit modulation of behavioral phenotypes might prove more fruitful than target-based methods for identifying novel compounds with rapid and sustained therapeutic effects similar to psychedelics and entactogens.
Collapse
Affiliation(s)
- Boris D Heifets
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, 94305, USA.
| | - David E Olson
- Institute for Psychedelics and Neurotherapeutics, University of California, Davis, Davis, CA, 95616, USA.
- Department of Chemistry, University of California, Davis, Davis, CA, 95616, USA.
- Center for Neuroscience, University of California, Davis, Davis, CA, 95618, USA.
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, 95817, USA.
| |
Collapse
|
6
|
Sharp T, Collins H. Mechanisms of SSRI Therapy and Discontinuation. Curr Top Behav Neurosci 2024; 66:21-47. [PMID: 37955823 DOI: 10.1007/7854_2023_452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
SSRIs are one of the most widely used drug therapies in primary care and psychiatry, and central to the management of the most common mental health problems in today's society. Despite this, SSRIs suffer from a slow onset of therapeutic effect and relatively poor efficacy as well as adverse effects, with recent concerns being focused on a disabling SSRI discontinuation syndrome. The mechanism underpinning their therapeutic effect has long shifted away from thinking that SSRIs act simply by increasing 5-HT in the synapse. Rather, a current popular view is that increased 5-HT is just the beginning of a series of complex downstream signalling events, which trigger changes in neural plasticity at the functional and structural level. These changes in plasticity are then thought to interact with neuropsychological processes to enhance re-learning of emotional experiences that ultimately brings about changes in mood. This compelling view of SSRI action is underpinning attempts to understand fast-acting antidepressants, such as ketamine and psychedelic drugs, and aid the development of future therapies. An important gap in the theory is evidence that changes in plasticity are causally linked to relevant behavioural effects. Also, predictions that the SSRI-induced neural plasticity might have applicability in other areas of medicine have not yet been borne out. In contrast to the sophisticated view of the antidepressant action of SSRIs, the mechanism underpinning SSRI discontinuation is little explored. Nevertheless, evidence of rebound increases in 5-HT neuron excitability immediately on cessation of SSRI treatment provide a starting point for future investigation. Indeed, this evidence allows formulation of a mechanistic explanation of SSRI discontinuation which draws on parallels with the withdrawal states of other psychotropic drugs.
Collapse
Affiliation(s)
- Trevor Sharp
- Department of Pharmacology, University of Oxford, Oxford, UK.
| | - Helen Collins
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
7
|
Maćkowiak M. Psychedelics action and schizophrenia. Pharmacol Rep 2023; 75:1350-1361. [PMID: 37899392 PMCID: PMC10661800 DOI: 10.1007/s43440-023-00546-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/09/2023] [Accepted: 10/16/2023] [Indexed: 10/31/2023]
Abstract
Psychedelics are compounds acting by serotonin 5-hydroxytryptamine (5-HT)2A receptor activation and induce several behavioral responses. They are of special interest because of their positive effects on neuropsychiatric disorders (depression and posttraumatic stress disorder). However, several findings revealed that some psychedelic actions are similar to symptoms observed in schizophrenia (psychosis, sensorimotor gating impairments, attention, and working memory deficits) which might limit their clinical applications. Psychedelics activate some neurotransmitters, i.e., serotonergic, and glutamatergic, that are also impaired in schizophrenia. Therefore, the neurobiological background of psychedelics and schizophrenia is partially similar. Another important aspect to discuss is the perspective of using psychedelics in schizophrenia therapy. Postmortem studies showed a loss of synapses in schizophrenia, and the positive effects of psychedelics on neuroplasticity (synaptogenesis, neurogenesis, and neuritogenesis) might be essential in the context of schizophrenia therapy. However, because of psychedelics' psychotic action, the recommended doses of psychedelics in schizophrenia treatment are not established, and subpsychedelic dosing or microdosing are considered. Exploratory studies are needed to determine the tolerability of treatment and appropriate dosing regimen. Another therapeutic option is using non-hallucinogenic psychedelic analogs that also induce neuroplastic outcomes but do not have psychotogenic effects. Further preclinical and clinical studies are needed to recognize the potential effectiveness of 5-HT2A agonists in schizophrenia therapy.
Collapse
Affiliation(s)
- Marzena Maćkowiak
- Laboratory of Pharmacology and Brain Biostructure, Pharmacology Department, Maj Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland.
| |
Collapse
|
8
|
Chen T, Xu YP, Chen Y, Sun S, Yan ZZ, Wang YH. Arc regulates brain damage and neuroinflammation via Sirt1 signaling following subarachnoid hemorrhage. Brain Res Bull 2023; 203:110780. [PMID: 37820952 DOI: 10.1016/j.brainresbull.2023.110780] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 10/04/2023] [Accepted: 10/09/2023] [Indexed: 10/13/2023]
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) accounts for only 5 % of all stroke cases, but carries a heavy burden of morbidity and mortality. Activity regulated cytoskeleton associated protein (Arc) is an immediate early gene (IEG)-coded postsynaptic protein that is involved in synaptic plasticity. Increasing evidence and our previous studies have shown that Arc might be involved in the pathological mechanism of various neurological diseases, such as traumatic brain injury (TBI). In this study, we investigated the level of Arc in cerebrospinal fluids (CSF) of aSAH patients and its potential role in brain damage following experimental SAH model. We found that the levels of Arc in aSAH patients' CSF positively correlated with Hunt-Hess (H&H) grades. Knockdown of endogenous Arc expression by small interfere RNA (siRNA) significantly increased brain edema and oxidative stress following SAH. The results of immunostaining in brain sections showed that knockdown of Arc enhanced activation of microglia and astrocytes. In congruent, generation of inflammatory cytokines following SAH was increased by Si-Arc transfection. The results of western blot analysis showed that knockdown of Arc inhibited the expression of Sirt1 and Nrf2, which was accompanied by decreased enzymatic activities of superoxide dismutase (SOD) and glutathione peroxidase (GSH-px). In addition, activation of sirtuin 1 (Sirt1) via agonist SRT2104 markedly decreased the brain damage and neuroinflammation induced by Arc knockdown. In conclusion, knockdown of endogenous Arc could aggravate brain damage and neuroinflammation following experimental SAH, and Arc levels in aSAH patients' CSF might be a potential indicator of brain damage and prognosis.
Collapse
Affiliation(s)
- Tao Chen
- Department of Neurosurgery, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Ye-Ping Xu
- Department of Neurosurgery, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Yang Chen
- Department of Neurology, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Shu Sun
- Department of Pharmacy, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Zhi-Zhong Yan
- Department of Neurosurgery, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China
| | - Yu-Hai Wang
- Department of Neurosurgery, Wuxi Taihu Hospital, Wuxi Clinical College of Anhui Medical University, Wuxi, Jiangsu 214044, China.
| |
Collapse
|
9
|
Morici JF, Cicuttin G, Silva A, Gallo FT, Miranda M, Beluscio M, Zold C, Bekinschtein P, Weisstaub NV. Serotonin Type 2a Receptor in the Prefrontal Cortex Controls Perirhinal Cortex Excitability During Object Recognition Memory Recall. Neuroscience 2022; 497:196-205. [PMID: 35597334 DOI: 10.1016/j.neuroscience.2022.05.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/06/2022] [Accepted: 05/12/2022] [Indexed: 10/18/2022]
Abstract
Previous experiences can drive adaptive behavior based on different characteristics, including contextual ones. Indeed, contextual information can be used as a criterion to guide the recall of the most relevant memory trace and the inhibition of others. The medial Prefontal Cortex (mPFC) has been proposed as an area that plays a pivotal role in regulating the retrieval of memory traces in downstream regions. Also, we have shown that mPFC Serotonin 2a Receptors (5-HT2aR) modulates the retrieval of a contextually guided recognition memory task and modulates the retrieval and reconsolidation of memories in the Perirhinal Cortex (PRH). However, how the mPFC output mediated by the 5-HT2aR activity is modulating memory retrieval in the PRH is a question that remains unclear. To tackle this question, we analyzed neuronal activity in the PRH and mPFC, by measuring expression of the immediate early gene c-Fos. We combined behavioral, pharmacological and immunohistochemical techniques to examine how mPFC 5-HT2aR controls mPFC and the PRH activity. We found that blockade of mPFC 5-HT2aR increase the level of c-Fos expression in the PHR and that this increase correlates with animals' performance in the task. We also found an increase in c-Fos expression in the mPFC after mPFC 5-HT2aR blockade that does not correlate with the animals' behavioral response. However, these changes showed a significant correlation with those observed in the PRH. These results suggest that mPFC 5-HT2aR signaling may modulate the behavioral response during memory recall by controlling the neuronal activation in the PRH.
Collapse
Affiliation(s)
- J F Morici
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - G Cicuttin
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - A Silva
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
| | - F T Gallo
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - M Miranda
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - M Beluscio
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
| | - C Zold
- Instituto de Fisiología y Biofísica "Bernardo Houssay" (IFIBIO-Houssay), Grupo de Neurociencia de Sistemas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) Buenos Aires, Argentina
| | - P Bekinschtein
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina
| | - N V Weisstaub
- Instituto de Neurociencias Cognitiva y Traslacional, Concejo Nacional de Investigaciones Científicas y tecnológicas (CONICET), Universidad Favaloro, Instituto de Neurología Cognitiva (INECO), Pacheco de Melo 1860, C1126AAB Buenos Aires, Argentina.
| |
Collapse
|
10
|
Abstract
In addition to producing profound subjective effects following acute administration, psychedelic compounds can induce beneficial behavioral changes relevant to the treatment of neuropsychiatric disorders that last long after the compounds have been cleared from the body. One hypothesis with the potential to explain the remarkable enduring effects of psychedelics is related to their abilities to promote structural and functional neuroplasticity in the prefrontal cortex (PFC). A hallmark of many stress-related neuropsychiatric diseases, including depression, post-traumatic stress disorder (PTSD), and addiction, is the atrophy of neurons in the PFC. Psychedelics appear to be particularly effective catalysts for the growth of these key neurons, ultimately leading to restoration of synaptic connectivity in this critical brain region. Furthermore, evidence suggests that the hallucinogenic effects of psychedelics are not directly linked to their ability to promote structural and functional neuroplasticity. If we are to develop improved alternatives to psychedelics for treating neuropsychiatric diseases, we must fully characterize the molecular mechanisms that give rise to psychedelic-induced neuroplasticity. Here, I review our current understanding of the biochemical signaling pathways activated by psychedelics and related neuroplasticity-promoting molecules, with an emphasis on key unanswered questions.
Collapse
Affiliation(s)
- David E. Olson
- Department of Chemistry, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA,Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Davis, 2700 Stockton Blvd, Suite 2102, Sacramento, CA 95817, USA,Center for Neuroscience, University of California, Davis, 1544 Newton Ct, Davis, CA 95618, USA,Corresponding Author: David E. Olson,
| |
Collapse
|
11
|
Saeger HN, Olson DE. Psychedelic-inspired approaches for treating neurodegenerative disorders. J Neurochem 2021; 162:109-127. [PMID: 34816433 DOI: 10.1111/jnc.15544] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/19/2021] [Accepted: 11/21/2021] [Indexed: 12/21/2022]
Abstract
Psychedelics are increasingly being recognized for their potential to treat a wide range of brain disorders including depression, post-traumatic stress disorder (PTSD), and substance use disorder. Their broad therapeutic potential might result from an ability to rescue cortical atrophy common to many neuropsychiatric and neurodegenerative diseases by impacting neurotrophic factor gene expression, activating neuronal growth and survival mechanisms, and modulating the immune system. While the therapeutic potential of psychedelics has not yet been extended to neurodegenerative disorders, we provide evidence suggesting that approaches based on psychedelic science might prove useful for treating these diseases. The primary target of psychedelics, the 5-HT2A receptor, plays key roles in cortical neuron health and is dysregulated in Alzheimer's disease. Moreover, evidence suggests that psychedelics and related compounds could prove useful for treating the behavioral and psychological symptoms of dementia (BPSD). While more research is needed to probe the effects of psychedelics in models of neurodegenerative diseases, the robust effects of these compounds on structural and functional neuroplasticity and inflammation clearly warrant further investigation.
Collapse
Affiliation(s)
- Hannah N Saeger
- Pharmacology and Toxicology Graduate Group, University of California, Davis, Davis, California, USA
| | - David E Olson
- Department of Chemistry, University of California, Davis, Davis, California, USA.,Department of Biochemistry & Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, California, USA.,Center for Neuroscience, University of California, Davis, Davis, California, USA
| |
Collapse
|
12
|
Banks MI, Zahid Z, Jones NT, Sultan ZW, Wenthur CJ. Catalysts for change: the cellular neurobiology of psychedelics. Mol Biol Cell 2021; 32:1135-1144. [PMID: 34043427 PMCID: PMC8351556 DOI: 10.1091/mbc.e20-05-0340] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/23/2021] [Accepted: 03/31/2021] [Indexed: 01/18/2023] Open
Abstract
The resurgence of interest in the therapeutic potential of psychedelics for treating psychiatric disorders has rekindled efforts to elucidate their mechanism of action. In this Perspective, we focus on the ability of psychedelics to promote neural plasticity, postulated to be central to their therapeutic activity. We begin with a brief overview of the history and behavioral effects of the classical psychedelics. We then summarize our current understanding of the cellular and subcellular mechanisms underlying these drugs' behavioral effects, their effects on neural plasticity, and the roles of stress and inflammation in the acute and long-term effects of psychedelics. The signaling pathways activated by psychedelics couple to numerous potential mechanisms for producing long-term structural changes in the brain, a complexity that has barely begun to be disentangled. This complexity is mirrored by that of the neural mechanisms underlying psychiatric disorders and the transformations of consciousness, mood, and behavior that psychedelics promote in health and disease. Thus, beyond changes in the brain, psychedelics catalyze changes in our understanding of the neural basis of psychiatric disorders, as well as consciousness and human behavior.
Collapse
Affiliation(s)
- Matthew I. Banks
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706
- Neuroscience Training Program, University of Wisconsin–Madison, Madison, WI 53706
| | - Zarmeen Zahid
- Neuroscience Training Program, University of Wisconsin–Madison, Madison, WI 53706
| | - Nathan T. Jones
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin–Madison, Madison, WI 53706
| | - Ziyad W. Sultan
- Department of Anesthesiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53706
| | - Cody J. Wenthur
- Neuroscience Training Program, University of Wisconsin–Madison, Madison, WI 53706
- Molecular and Cellular Pharmacology Training Program, University of Wisconsin–Madison, Madison, WI 53706
- School of Pharmacy, University of Wisconsin–Madison, Madison, WI 53705
| |
Collapse
|
13
|
Cortical influences of serotonin and glutamate on layer V pyramidal neurons. PROGRESS IN BRAIN RESEARCH 2021; 261:341-378. [PMID: 33785135 DOI: 10.1016/bs.pbr.2020.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Layer V pyramidal neurons constitute principle output neurons of the medial prefrontal cortex (mPFC)/neocortex to subcortical regions including the intralaminar/midline thalamic nuclei, amygdala, basal ganglia, brainstem nuclei and the spinal cord. The effects of 5-hydroxytryptamine (5-HT) on layer V pyramidal cells primarily reflect a range of excitatory influences through 5-HT2A receptors and inhibitory influences through non-5-HT2A receptors, including 5-HT1A receptors. While the 5-HT2A receptor is primarily a postsynaptic receptor on throughout the apical dendritic field of 5-HT2A receptors, activation of a minority of 5-HT2A receptors also appears to increase spontaneous excitatory postsynaptic currents/potentials (EPSCs/EPSPs) via a presynaptic effect on thalamocortical terminals arising from the midline and intralaminar thalamic nuclei. Activation of 5-HT2A receptors by the phenethylamine hallucinogen also appears to increase asynchronous release of glutamate upon the layer V pyramidal dendritic field, an effect that is suppressed by 5-HT itself through non-5-HT2A receptors. Serotonergic hallucinogens acting on 5-HT2A receptors also appears to increase gene expression of immediate early genes (iEG) and other receptors appearing to induce an iEG-like response like BDNF. Psychedelic hallucinogens acting on 5-HT2A receptors also induce head twitches in rodents that appear related to induction of glutamate release. These electrophysiological, biochemical and behavioral effects of serotonergic hallucinogens appear to be related to modulating glutamatergic thalamocortical neurotransmission and/or shifting the balance toward 5-HT2A receptor activation and away from non-5-HT2A receptor activation. These 5-HT2A receptor induced responses are modulated by feedback homeostatic mechanisms through mGlu2, mGlu4, and mGlu8 presynaptic receptors on thalamocortical terminals. These 5-HT2A receptor and glutamatergic interactions also appear to play a role on higher cortical functions of the mPFC such as motoric impulsivity and antidepressant-like behavioral responses on the differential-reinforcement-of low rate 72-s (DRL 72-s schedule). These mutually opposing effects between 5-HT2A receptor and mGlu autoreceptor activation (e.g., blocking 5-HT2A receptors and enhancing activity at mGlu2 receptors) may play a clinical role with respect to currently prescribed or novel antidepressant drugs. Thus, there is an important balance between 5-HT2A receptor activation and activation of mGlu autoreceptors on prefrontal cortical layer V pyramidal cells with respect to the electrophysiological, biochemical and behavioral effects serotonergic hallucinogenic drugs.
Collapse
|
14
|
Chronic treatment with a metabotropic mGlu2/3 receptor agonist diminishes behavioral response to a phenethylamine hallucinogen. Psychopharmacology (Berl) 2019; 236:821-830. [PMID: 30448990 PMCID: PMC6778591 DOI: 10.1007/s00213-018-5118-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 11/08/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND There is evidence that mGlu2/3 receptors regulate 5-HT2A signaling, interactions that have been theorized to play a role in the antipsychotic-like effects of mGlu2/3 agonists as well as the hallucinogenic effects of 5-HT2A agonists. One approach to unraveling this interaction is through the chronic administration of agonists at the two receptors, which should influence the functional properties of the targeted receptor due to receptor downregulation or desensitization and thereby alter crosstalk between the two receptors. In this study, we investigated whether chronic treatment with the mGlu2/3 agonist LY379268 would alter the behavioral response to a phenethylamine hallucinogen, 25CN-NBOH, which acts as a selective 5-HT2A agonist. METHODS We first conducted a dose response of 25CN-NBOH (0.1, 0.3, 1, 3, or 10 mg/kg) to confirm the effects on head-twitch response (HTR) and then blockade studies with either the M100907 (0.1 mg/kg) or SB242084 (0.1, 0.3, or 1 mg/kg) to determine the contribution of 5-HT2A and 5-HT2C to 25CN-NBOH-induced HTR, respectively. To determine whether an mGlu2/3 agonist could block 25CN-NBOH-induced HTR, mice were pretreated with vehicle or LY379268 (0.1, 1, or 10 mg/kg) prior to 25CN-NBOH, and HTR was assessed. The effects of chronic LY379268 on 5-HT2A agonist-induced HTR were evaluated by treating mice with either vehicle or LY379268 (10 mg/kg) for 21 days and measuring 25CN-NBOH-induced HTR 48 h after the final LY379268 treatment. The following day (72 h after the final LY379268 treatment), the ability of acute LY379268 to block PCP-induced locomotor activity was assessed. RESULTS 25CN-NBOH dose-dependently increased the HTR, a 5-HT2A-mediated behavior, in mice. The selective 5-HT2A antagonist M100907 completely blocked the HTR induced by 25CN-NBOH, whereas the selective 5-HT2C antagonist SB242084 had no effect on the HTR. Administration of LY379268 (10 mg/kg SC) attenuated the HTR induced by 1 mg/kg 25CN-NBOH by ~ 50%. Chronic treatment (21 days) with LY379268 also attenuated the HTR response to 25CN-NBOH when tested 48 h after the last dose of LY379268. In locomotor tests, acute LY379268 significantly attenuated PCP-induced locomotor activity in the chronic vehicle treatment group; by contrast, there was only a trend for an overall interaction in the chronic LY379268 group, with LY379268 blocking the locomotor-stimulating effects of PCP only during the last 20 min. CONCLUSIONS These data are consistent with a functional interaction between mGlu2/3 and 5-HT2A receptors, although the specific mechanism for the interaction is not known. These data support the hypothesis that mGlu2/3 receptors play a prominent role in modulating the behavioral response to 5-HT2A receptor activation.
Collapse
|
15
|
Antoniadou I, Kouskou M, Arsiwala T, Singh N, Vasudevan SR, Fowler T, Cadirci E, Churchill GC, Sharp T. Ebselen has lithium-like effects on central 5-HT 2A receptor function. Br J Pharmacol 2018; 175:2599-2610. [PMID: 29488218 DOI: 10.1111/bph.14179] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 01/23/2018] [Accepted: 01/30/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Lithium's antidepressant action may be mediated by inhibition of inositol monophosphatase (IMPase), a key enzyme in Gq -protein coupled receptor signalling. Recently, the antioxidant agent ebselen was identified as an IMPase inhibitor. Here, we investigated both ebselen and lithium in models of the 5-HT2A receptor, a Gq -protein coupled receptor involved in lithium's actions. EXPERIMENTAL APPROACH 5-HT2A receptor function was assessed in mice by measuring the behavioural (head-twitches, ear scratches) and molecular (cortical immediate early gene [IEG] mRNA; Arc, c-fos, Egr2) responses to 5-HT2A receptor agonists. Ebselen and lithium were administered either acutely or repeatedly prior to assessment of 5-HT2A receptor function. Because lithium and 5-HT2A receptor antagonists augment the action of selective serotonin reuptake inhibitors (SSRIs), ebselen was tested for this activity by co-administration with the SSRI citalopram in microdialysis (extracellular 5-HT) experiments. KEY RESULTS Acute and repeated administration of ebselen inhibited behavioural and IEG responses to the 5-HT2A receptor agonist DOI. Repeated lithium also inhibited DOI-evoked behavioural and IEG responses. In comparison, a selective IMPase inhibitor (L-690330) attenuated the behavioural response to DOI whereas glycogen synthase kinase inhibitor (AR-A014418) did not. Finally, ebselen enhanced the increase in extracellular 5-HT induced by citalopram, and also increased regional brain 5-HT synthesis. CONCLUSIONS AND IMPLICATIONS Our data demonstrated lithium-mimetic effects of ebselen in different experimental models of 5-HT2A receptor function, probably mediated by IMPase inhibition. This evidence of lithium-like neuropharmacological effects of ebselen adds further support for the clinical testing of ebselen in mood disorders, including as an antidepressant augmenting agent.
Collapse
Affiliation(s)
- I Antoniadou
- Department of Pharmacology, University of Oxford, Oxford, UK.,Department of Pharmacy, European University of Cyprus, Nicosia, Cyprus
| | - M Kouskou
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - T Arsiwala
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - N Singh
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - S R Vasudevan
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - T Fowler
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - E Cadirci
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - G C Churchill
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - T Sharp
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
de Bartolomeis A, Buonaguro EF, Latte G, Rossi R, Marmo F, Iasevoli F, Tomasetti C. Immediate-Early Genes Modulation by Antipsychotics: Translational Implications for a Putative Gateway to Drug-Induced Long-Term Brain Changes. Front Behav Neurosci 2017; 11:240. [PMID: 29321734 PMCID: PMC5732183 DOI: 10.3389/fnbeh.2017.00240] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/22/2017] [Indexed: 12/12/2022] Open
Abstract
An increasing amount of research aims at recognizing the molecular mechanisms involved in long-lasting brain architectural changes induced by antipsychotic treatments. Although both structural and functional modifications have been identified following acute antipsychotic administration in humans, currently there is scarce knowledge on the enduring consequences of these acute changes. New insights in immediate-early genes (IEGs) modulation following acute or chronic antipsychotic administration may help to fill the gap between primary molecular response and putative long-term changes. Moreover, a critical appraisal of the spatial and temporal patterns of IEGs expression may shed light on the functional "signature" of antipsychotics, such as the propensity to induce motor side effects, the potential neurobiological mechanisms underlying the differences between antipsychotics beyond D2 dopamine receptor affinity, as well as the relevant effects of brain region-specificity in their mechanisms of action. The interest for brain IEGs modulation after antipsychotic treatments has been revitalized by breakthrough findings such as the role of early genes in schizophrenia pathophysiology, the involvement of IEGs in epigenetic mechanisms relevant for cognition, and in neuronal mapping by means of IEGs expression profiling. Here we critically review the evidence on the differential modulation of IEGs by antipsychotics, highlighting the association between IEGs expression and neuroplasticity changes in brain regions impacted by antipsychotics, trying to elucidate the molecular mechanisms underpinning the effects of this class of drugs on psychotic, cognitive and behavioral symptoms.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Elisabetta F Buonaguro
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Gianmarco Latte
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Rodolfo Rossi
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Federica Marmo
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Felice Iasevoli
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| | - Carmine Tomasetti
- Laboratory of Molecular and Translational Psychiatry and Unit of Treatment Resistant Psychosis, Section of Psychiatry, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University School of Medicine "Federico II", Naples, Italy
| |
Collapse
|
17
|
Sood A, Pati S, Bhattacharya A, Chaudhari K, Vaidya VA. Early emergence of altered 5‐HT
2A
receptor‐evoked behavior, neural activation and gene expression following maternal separation. Int J Dev Neurosci 2017; 65:21-28. [DOI: 10.1016/j.ijdevneu.2017.10.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 09/20/2017] [Accepted: 10/12/2017] [Indexed: 01/15/2023] Open
Affiliation(s)
- Ankit Sood
- Department of Biological SciencesTata Institute of Fundamental ResearchMumbaiMaharashtraIndia
| | - Sthitapranjya Pati
- Department of Biological SciencesTata Institute of Fundamental ResearchMumbaiMaharashtraIndia
| | - Amrita Bhattacharya
- Department of Biological SciencesTata Institute of Fundamental ResearchMumbaiMaharashtraIndia
| | - Karina Chaudhari
- Department of Biological SciencesTata Institute of Fundamental ResearchMumbaiMaharashtraIndia
| | - Vidita A. Vaidya
- Department of Biological SciencesTata Institute of Fundamental ResearchMumbaiMaharashtraIndia
| |
Collapse
|
18
|
Jaggar M, Weisstaub N, Gingrich JA, Vaidya VA. 5-HT 2A receptor deficiency alters the metabolic and transcriptional, but not the behavioral, consequences of chronic unpredictable stress. Neurobiol Stress 2017. [PMID: 28626787 PMCID: PMC5470573 DOI: 10.1016/j.ynstr.2017.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Chronic stress enhances risk for psychiatric disorders, and in animal models is known to evoke depression-like behavior accompanied by perturbed neurohormonal, metabolic, neuroarchitectural and transcriptional changes. Serotonergic neurotransmission, including serotonin2A (5-HT2A) receptors, have been implicated in mediating specific aspects of stress-induced responses. Here we investigated the influence of chronic unpredictable stress (CUS) on depression-like behavior, serum metabolic measures, and gene expression in stress-associated neurocircuitry of the prefrontal cortex (PFC) and hippocampus in 5-HT2A receptor knockout (5-HT2A−/−) and wild-type mice of both sexes. While 5-HT2A−/− male and female mice exhibited a baseline reduced anxiety-like state, this did not alter the onset or severity of behavioral despair during and at the cessation of CUS, indicating that these mice can develop stress-evoked depressive behavior. Analysis of metabolic parameters in serum revealed a CUS-evoked dyslipidemia, which was abrogated in 5-HT2A−/− female mice with a hyperlipidemic baseline phenotype. 5-HT2A−/− male mice in contrast did not exhibit such a baseline shift in their serum lipid profile. Specific stress-responsive genes (Crh, Crhr1, Nr3c1, and Nr3c2), trophic factors (Bdnf, Igf1) and immediate early genes (IEGs) (Arc, Fos, Fosb, Egr1-4) in the PFC and hippocampus were altered in 5-HT2A−/− mice both under baseline and CUS conditions. Our results support a role for the 5-HT2A receptor in specific metabolic and transcriptional, but not behavioral, consequences of CUS, and highlight that the contribution of the 5-HT2A receptor to stress-evoked changes is sexually dimorphic.
Collapse
Affiliation(s)
- Minal Jaggar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Noelia Weisstaub
- Department of Physiology, Faculty of Medicine, University of Buenos Aires, Argentina
| | - Jay A Gingrich
- Department of Psychiatry, Columbia University, New York, United States
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
19
|
Abstract
The classic serotonergic hallucinogens, or psychedelics, have the ability to profoundly alter perception and behavior. These can include visual distortions, hallucinations, detachment from reality, and mystical experiences. Some psychedelics, like LSD, are able to produce these effects with remarkably low doses of drug. Others, like psilocybin, have recently been demonstrated to have significant clinical efficacy in the treatment of depression, anxiety, and addiction that persist for at least several months after only a single therapeutic session. How does this occur? Much work has recently been published from imaging studies showing that psychedelics alter brain network connectivity. They facilitate a disintegration of the default mode network, producing a hyperconnectivity between brain regions that allow centers that do not normally communicate with each other to do so. The immediate and acute effects on both behaviors and network connectivity are likely mediated by effector pathways downstream of serotonin 5-HT2A receptor activation. These acute molecular processes also influence gene expression changes, which likely influence synaptic plasticity and facilitate more long-term changes in brain neurochemistry ultimately underlying the therapeutic efficacy of a single administration to achieve long-lasting effects. In this review, we summarize what is currently known about the molecular genetic responses to psychedelics within the brain and discuss how gene expression changes may contribute to altered cellular physiology and behaviors.
Collapse
|
20
|
Martin DA, Nichols CD. Psychedelics Recruit Multiple Cellular Types and Produce Complex Transcriptional Responses Within the Brain. EBioMedicine 2016; 11:262-277. [PMID: 27649637 PMCID: PMC5050000 DOI: 10.1016/j.ebiom.2016.08.049] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 08/24/2016] [Accepted: 08/31/2016] [Indexed: 11/22/2022] Open
Abstract
There has recently been a resurgence of interest in psychedelics, substances that profoundly alter perception and cognition and have recently demonstrated therapeutic efficacy to treat anxiety, depression, and addiction in the clinic. The receptor mechanisms that drive their molecular and behavioral effects involve activation of cortical serotonin 5-HT2A receptors, but the responses of specific cellular populations remain unknown. Here, we provide evidence that a small subset of 5-HT2A-expressing excitatory neurons is directly activated by psychedelics and subsequently recruits other select cell types including subpopulations of inhibitory somatostatin and parvalbumin GABAergic interneurons, as well as astrocytes, to produce distinct and regional responses. To gather data regarding the response of specific neuronal populations, we developed methodology for fluorescence-activated cell sorting (FACS) to segregate and enrich specific cellular subtypes in the brain. These methods allow for robust neuronal sorting based on cytoplasmic epitopes followed by downstream nucleic acid analysis, expanding the utility of FACS in neuroscience research. Psychedelics activate distinct transcription across cell types, including excitatory neurons, inhibitory neurons, and astrocytes Psychedelics induce internalization of 5-HT2A receptors throughout the cortex and claustrum FACS can separate neuronal subpopulations that require non-nuclear markers
Psychedelic drugs are known to act through the 5-HT2A receptor to produce many of their effects, however, the precise cellular populations in the brain which respond to this class of drugs remain unknown. We use flow cytometric analyses, immunohistochemistry, and gene expression analyses to identify small populations of specific cells in the brain that are activated by the psychedelic drug, (R)-DOI. The methodology used in these studies will be useful to determine the molecular effects of any manipulation or disease on particular brain cells.
Collapse
Affiliation(s)
- David A Martin
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Charles D Nichols
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA.
| |
Collapse
|
21
|
Pereira M, Martynhak BJ, Andreatini R, Svenningsson P. 5-HT6 receptor agonism facilitates emotional learning. Front Pharmacol 2015; 6:200. [PMID: 26441657 PMCID: PMC4584947 DOI: 10.3389/fphar.2015.00200] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 08/31/2015] [Indexed: 01/18/2023] Open
Abstract
Serotonin (5-HT) and its receptors play crucial roles in various aspects of mood and cognitive functions. However, the role of specific 5-HT receptors in these processes remains to be better understood. Here, we examined the effects of the selective and potent 5-HT6 agonist (WAY208466) on mood, anxiety and emotional learning in mice. Male C57Bl/6J mice were therefore tested in the forced swim test (FST), elevated plus-maze (EPM), and passive avoidance tests (PA), respectively. In a dose-response experiment, mice were treated intraperitoneally with WAY208466 at 3, 9, or 27 mg/kg and examined in an open field arena open field test (OFT) followed by the FST. 9 mg/kg of WAY208466 reduced immobility in the FST, without impairing the locomotion. Thus, the dose of 9 mg/kg was subsequently used for tests of anxiety and emotional learning. There was no significant effect of WAY208466 in the EPM. In the PA, mice were trained 30 min before the treatment with saline or WAY208466. Two separate sets of animals were used for short term memory (tested 1 h post-training) or long term memory (tested 24 h post-training). WAY208466 improved both short and long term memories, evaluated by the latency to enter the dark compartment, in the PA. The WAY208466-treated animals also showed more grooming and rearing in the light compartment. To better understand the molecular mechanisms and brain regions involved in the facilitation of emotional learning by WAY208466, we studied its effects on signal transduction and immediate early gene expression. WAY208466 increased the levels of phospho-Ser845-GluA1 and phospho-Ser217/221-MEK in the caudate-putamen. Levels of phospho-Thr202/204-Erk1/2 and the ratio mature BDNF/proBDNF were increased in the hippocampus. Moreover, WAY208466 increased c-fos in the hippocampus and Arc expression in both hippocampus and prefrontal cortex (PFC). The results indicate antidepressant efficacy and facilitation of emotional learning by 5-HT6 receptor agonism via mechanisms that promote neuronal plasticity in caudate putamen, hippocampus, and PFC.
Collapse
Affiliation(s)
- Marcela Pereira
- Section of Translational Neuropharmacology, Department of Clinical Neuroscience, Center of Molecular Medicine, Karolinska Institute Stockholm, Sweden
| | - Bruno J Martynhak
- Section of Translational Neuropharmacology, Department of Clinical Neuroscience, Center of Molecular Medicine, Karolinska Institute Stockholm, Sweden
| | - Roberto Andreatini
- Department of Pharmacology, Federal University of Paraná Curitiba, Brazil
| | - Per Svenningsson
- Section of Translational Neuropharmacology, Department of Clinical Neuroscience, Center of Molecular Medicine, Karolinska Institute Stockholm, Sweden
| |
Collapse
|
22
|
Li Y, Pehrson AL, Waller JA, Dale E, Sanchez C, Gulinello M. A critical evaluation of the activity-regulated cytoskeleton-associated protein (Arc/Arg3.1)'s putative role in regulating dendritic plasticity, cognitive processes, and mood in animal models of depression. Front Neurosci 2015; 9:279. [PMID: 26321903 PMCID: PMC4530346 DOI: 10.3389/fnins.2015.00279] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 07/23/2015] [Indexed: 11/13/2022] Open
Abstract
Major depressive disorder (MDD) is primarily conceptualized as a mood disorder but cognitive dysfunction is also prevalent, and may limit the daily function of MDD patients. Current theories on MDD highlight disturbances in dendritic plasticity in its pathophysiology, which could conceivably play a role in the production of both MDD-related mood and cognitive symptoms. This paper attempts to review the accumulated knowledge on the basic biology of the activity-regulated cytoskeleton-associated protein (Arc or Arg3.1), its effects on neural plasticity, and how these may be related to mood or cognitive dysfunction in animal models of MDD. On a cellular level, Arc plays an important role in modulating dendritic spine density and remodeling. Arc also has a close, bidirectional relationship with postsynaptic glutamate neurotransmission, since it is stimulated by multiple glutamatergic receptor mechanisms but also modulates α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor internalization. The effects on AMPA receptor trafficking are likely related to Arc's ability to modulate phenomena such as long-term potentiation, long-term depression, and synaptic scaling, each of which are important for maintaining proper cognitive function. Chronic stress models of MDD in animals show suppressed Arc expression in the frontal cortex but elevation in the amygdala. Interestingly, cognitive tasks depending on the frontal cortex are generally impaired by chronic stress, while those depending on the amygdala are enhanced, and antidepressant treatments stimulate cortical Arc expression with a timeline that is reminiscent of the treatment efficacy lag observed in the clinic or in preclinical models. However, pharmacological treatments that stimulate regional Arc expression do not universally improve relevant cognitive functions, and this highlights a need to further refine our understanding of Arc on a subcellular and network level.
Collapse
Affiliation(s)
- Yan Li
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Alan L Pehrson
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Jessica A Waller
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Elena Dale
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Connie Sanchez
- External Sourcing and Scientific Excellence, Lundbeck Research USA, Inc. Paramus, NJ, USA
| | - Maria Gulinello
- Behavioral Core Facility, Department of Neuroscience, Albert Einstein College of Medicine Bronx, NY, USA
| |
Collapse
|
23
|
Halberstadt AL. Recent advances in the neuropsychopharmacology of serotonergic hallucinogens. Behav Brain Res 2015; 277:99-120. [PMID: 25036425 PMCID: PMC4642895 DOI: 10.1016/j.bbr.2014.07.016] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 07/07/2014] [Accepted: 07/08/2014] [Indexed: 12/12/2022]
Abstract
Serotonergic hallucinogens, such as (+)-lysergic acid diethylamide, psilocybin, and mescaline, are somewhat enigmatic substances. Although these drugs are derived from multiple chemical families, they all produce remarkably similar effects in animals and humans, and they show cross-tolerance. This article reviews the evidence demonstrating the serotonin 5-HT2A receptor is the primary site of hallucinogen action. The 5-HT2A receptor is responsible for mediating the effects of hallucinogens in human subjects, as well as in animal behavioral paradigms such as drug discrimination, head twitch response, prepulse inhibition of startle, exploratory behavior, and interval timing. Many recent clinical trials have yielded important new findings regarding the psychopharmacology of these substances. Furthermore, the use of modern imaging and electrophysiological techniques is beginning to help unravel how hallucinogens work in the brain. Evidence is also emerging that hallucinogens may possess therapeutic efficacy.
Collapse
Affiliation(s)
- Adam L Halberstadt
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
24
|
Waters S, Ponten H, Edling M, Svanberg B, Klamer D, Waters N. The dopaminergic stabilizers pridopidine and ordopidine enhance cortico-striatal Arc gene expression. J Neural Transm (Vienna) 2014; 121:1337-47. [PMID: 24817271 DOI: 10.1007/s00702-014-1231-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 04/24/2014] [Indexed: 12/29/2022]
Abstract
The dopaminergic stabilizers pridopidine [4-(3-(methylsulfonyl)phenyl)-1-propylpiperidine] and ordopidine [1-ethyl-4-(2-fluoro-3-(methylsulfonyl)phenyl)piperidine] inhibit psychostimulant-induced hyperactivity, and stimulate behaviour in states of hypoactivity. While both compounds act as dopamine D2 receptor antagonists in vitro, albeit with low affinity, their specific state-dependent behavioural effect profile is not shared by D2 receptor antagonists in general. To further understand the neuropharmacological effects of pridopidine and ordopidine, and how they differ from other dopaminergic compounds in vivo, we assessed the expression of activity-regulated cytoskeleton-associated protein/activity-regulated gene 3.1 (Arc), an immediate early gene marker associated with synaptic activation, in the frontal cortex and striatum. Furthermore, monoamine neurochemistry and locomotor activity were assessed. The effects of pridopidine and ordopidine were compared to reference dopamine D1 and D2 receptor agonists and antagonists, as well as the partial dopamine D2 agonist aripiprazole. Pridopidine and ordopidine induced significant increases in cortical Arc expression, reaching 2.2- and 1.7-fold levels relative to control, respectively. In contrast, none of the reference dopamine D1 and D2 compounds tested increased cortical Arc expression. In the striatum, significant increases in Arc expression were seen with both pridopidine and ordopidine as well as the dopamine D2 receptor antagonists, remoxipride and haloperidol. Interestingly, striatal Arc expression correlated strongly and positively with striatal 3,4-dihydroxyphenylacetic acid, suggesting that antagonism of dopamine D2 receptors increases Arc expression in the striatum. In conclusion, the concurrent increase in cortical and striatal Arc expression induced by pridopidine and ordopidine appears unique for the dopaminergic stabilizers, as it was not shared by the reference compounds tested. The increase in cortical Arc expression is hypothesized to reflect enhanced N-methyl-D-aspartic acid receptor-mediated signalling in the frontal cortex, which could contribute to the state-dependent locomotor effects of pridopidine and ordopidine.
Collapse
Affiliation(s)
- Susanna Waters
- Department of Pharmacology, Institute of Neuroscience and Physiology, University of Gothenburg, Box 431, 405 30, Gōteborg, Sweden,
| | | | | | | | | | | |
Collapse
|
25
|
Santini MA, Balu DT, Puhl MD, Hill-Smith TE, Berg AR, Lucki I, Mikkelsen JD, Coyle JT. D-serine deficiency attenuates the behavioral and cellular effects induced by the hallucinogenic 5-HT(2A) receptor agonist DOI. Behav Brain Res 2013; 259:242-6. [PMID: 24269270 DOI: 10.1016/j.bbr.2013.11.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 11/07/2013] [Accepted: 11/12/2013] [Indexed: 01/15/2023]
Abstract
Both the serotonin and glutamate systems have been implicated in the pathophysiology of schizophrenia, as well as in the mechanism of action of antipsychotic drugs. Psychedelic drugs act through the serotonin 2A receptor (5-HT2AR), and elicit a head-twitch response (HTR) in mice, which directly correlates to 5-HT2AR activation and is absent in 5-HT2AR knockout mice. The precise mechanism of this response remains unclear, but both an intrinsic cortico-cortical pathway and a thalamo-cortical pathway involving glutamate release have been proposed. Here, we used a genetic model of NMDAR hypofunction, the serine racemase knockout (SRKO) mouse, to explore the role of glutamatergic transmission in regulating 5-HT2AR-mediated cellular and behavioral responses. SRKO mice treated with the 5-HT2AR agonist (±)-2,5-dimethoxy-4-iodoamphetamine (DOI) showed a clearly diminished HTR and lower induction of c-fos mRNA. These altered functional responses in SRKO mice were not associated with changes in cortical or hippocampal 5-HT levels or in 5-HT2AR and metabotropic glutamate-2 receptor (mGluR2) mRNA and protein expression. Together, these findings suggest that D-serine-dependent NMDAR activity is involved in mediating the cellular and behavioral effects of 5-HT2AR activation.
Collapse
Affiliation(s)
- Martin A Santini
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Darrick T Balu
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Matthew D Puhl
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | | | - Alexandra R Berg
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, MA, USA
| | - Irwin Lucki
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA; Department of Pharmacology, University of Pennsylvania, Philadelphia, PA, USA
| | - Jens D Mikkelsen
- Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark; Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, MA, USA
| | - Joseph T Coyle
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, MA, USA; Department of Psychiatry, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Induction of the plasticity-associated immediate early gene Arc by stress and hallucinogens: role of brain-derived neurotrophic factor. Int J Neuropsychopharmacol 2013; 16:405-15. [PMID: 22404904 DOI: 10.1017/s1461145712000168] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Exposure to stress and hallucinogens in adulthood evokes persistent alterations in neurocircuitry and emotional behaviour. The structural and functional changes induced by stress and hallucinogen exposure are thought to involve transcriptional alterations in specific effector immediate early genes. The immediate early gene, activity regulated cytoskeletal-associated protein (Arc), is important for both activity and experience dependent plasticity. We sought to examine whether trophic factor signalling through brain-derived neurotrophic factor (BDNF) contributes to the neocortical regulation of Arc mRNA in response to distinct stimuli such as immobilization stress and the hallucinogen 2,5-dimethoxy-4-iodoamphetamine (DOI). Acute exposure to either immobilization stress or DOI induced Arc mRNA levels within the neocortex. BDNF infusion into the neocortex led to a robust up-regulation of local Arc transcript expression. Further, baseline Arc mRNA expression in the neocortex was significantly decreased in inducible BDNF knockout mice with an adult-onset, forebrain specific BDNF loss. The induction of Arc mRNA levels in response to both acute immobilization stress or a single administration of DOI was significantly attenuated in the inducible BDNF knockout mice. Taken together, our results implicate trophic factor signalling through BDNF in the regulation of cortical Arc mRNA expression, both under baseline conditions and following stress and hallucinogen exposure. These findings suggest the possibility that the regulation of Arc expression via BDNF provides a molecular substrate for the structural and synaptic plasticity observed following stimuli such as stress and hallucinogens.
Collapse
|
27
|
Santini MA, Ratner C, Aznar S, Klein AB, Knudsen GM, Mikkelsen JD. Enhanced prefrontal serotonin 2A receptor signaling in the subchronic phencyclidine mouse model of schizophrenia. J Neurosci Res 2013; 91:634-41. [DOI: 10.1002/jnr.23198] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 11/19/2012] [Accepted: 12/10/2012] [Indexed: 12/31/2022]
|
28
|
Serres F, Rodriguez M, Rivet JM, Galizzi JP, Lockhart B, Sharp T, Millan MJ. Blockade of α2-adrenoceptors induces Arc gene expression in rat brain in a glutamate receptor-dependent manner: a combined qPCR, in situ hybridisation and immunocytochemistry study. Neuropharmacology 2012; 63:992-1001. [PMID: 22828637 DOI: 10.1016/j.neuropharm.2012.06.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 06/27/2012] [Accepted: 06/30/2012] [Indexed: 10/28/2022]
Abstract
Studies of 5-HT-glutamate interactions suggest that activation of brain 5-HT(2A) receptors leads to an AMPA receptor-mediated induction of the immediate early (activity-dependent) gene, Arc (Arg3.1). In this respect, noradrenaline-glutamate interactions are poorly characterised. Here we investigated the influence on regional brain Arc gene expression of selective blockade of α(2)-adrenoceptors in rats. Several complementary techniques were used: qPCR (mRNA, discrete tissue punches), in situ hybridisation (mRNA, sections) and immunocytochemistry. The α(2)-adrenoceptor antagonist, RX 821002, dose-dependently and time-dependently (maximal effect 2 h) increased Arc mRNA levels as demonstrated both by qPCR and in situ hybridisation. The α(2)-adrenoceptor antagonist, atipamezole, also increased Arc mRNA in in situ hybridisation studies. Changes in Arc mRNA after RX 821002 were of similar magnitude in punches and intact tissue sections and region-specific, with effects being most pronounced in parietal cortex and caudate putamen, less robust in frontal cortex, and not detectable in hippocampal sub-regions. Both qPCR and in situ hybridisation studies demonstrated that RX 821002-induced Arc mRNA was blocked by the AMPA antagonist, GYKI 52466. Pretreatment with the NMDA antagonist MK 801 also prevented RX 821002-induced Arc mRNA, as did the mGluR5 antagonist MPEP, whilst the mGluR2/3 antagonist, LY341495, had no effect. Finally, immunocytochemical studies showed that RX 821002 increased Arc-immunoreactivity in cells in close apposition to α(2)-adrenoceptor-positive processes. Thus, employing three complementary techniques, these observations demonstrate that blockade of α(2)-adrenoceptors triggers brain expression of the immediate early gene, Arc, and that this effect involves the recruitment of AMPA, NMDA and mGluR5 but not mGluR2/3 glutamatergic receptors.
Collapse
Affiliation(s)
- Florence Serres
- University Department of Pharmacology, Mansfield Road, Oxford OX1 3QT, UK
| | | | | | | | | | | | | |
Collapse
|
29
|
Molecular adaptation to chronic antidepressant treatment: evidence for a more rapid response to the novel α₂-adrenoceptor antagonist/5-HT-noradrenaline reuptake inhibitor (SNRI), S35966, compared to the SNRI, venlafaxine. Int J Neuropsychopharmacol 2012; 15:617-29. [PMID: 21733241 DOI: 10.1017/s1461145711000733] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Evidence of early changes in neural plasticity may aid the prediction of rapid-onset antidepressant drugs. Here we compared the dual α₂-adrenoceptor antagonist/5-HT-noradrenaline reuptake inhibitor (SNRI), S35966, to the SNRI, venlafaxine, with regards to their effect on rat brain expression of a panel of neural plasticity-related genes: Arc, BDNF, and VGLUT1, as well as Homer1a and Shank1B (not studied previously). Abundance of mRNA was determined by in-situ hybridization in cortical and hippocampal regions 2 h and 16 h following drug administration for 14, 7 and 1 d. After 14 d, both S35966 and venlafaxine increased mRNA of all genes, including Homer1a and Shank1B, and effects were similarly time- and region-dependent. After 7 d, S35966 elevated Arc, Shank1B and BDNF mRNA, whereas venlafaxine increased Shank1B mRNA only. Finally, after 1 d (acute administration), S35966 increased Arc and Homer1a mRNA whereas venlafaxine had no effect on any gene examined. In summary, a 14-d course of treatment with S35966 or venlafaxine induced similar region- and time-dependent increases in expression of neural plasticity-related genes including Shank1B and Homer1a. Some genes responded earlier to S35966, suggesting that drugs with combined α₂-adrenoceptor antagonist/SNRI properties may elicit more rapid changes in markers of neural plasticity than a SNRI alone.
Collapse
|
30
|
Dyrvig M, Hansen HH, Christiansen SH, Woldbye DPD, Mikkelsen JD, Lichota J. Epigenetic regulation of Arc and c-Fos in the hippocampus after acute electroconvulsive stimulation in the rat. Brain Res Bull 2012; 88:507-13. [PMID: 22613772 DOI: 10.1016/j.brainresbull.2012.05.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/04/2012] [Accepted: 05/04/2012] [Indexed: 01/17/2023]
Abstract
Electroconvulsive stimulation (ECS) remains one of the most effective treatments of major depression. However, the underlying molecular changes still remain to be elucidated. Since ECS causes rapid and significant changes in gene expression we have looked at epigenetic regulation of two important immediate early genes that are both induced after ECS: c-Fos and Arc. We examined Arc and c-Fos protein expression and found Arc present over 4 h, in contrast to c-Fos presence lasting only 1 h. Both genes had returned to baseline expression at 24 h post-ECS. Histone H4 acetylation (H4Ac) is one of the important epigenetic marks associated with gene activation. We show increased H4Ac at the c-Fos promoter at 1 h post-ECS. Surprisingly, we also observed a significant increase in DNA methylation of the Arc gene promoter at 24 h post-ECS. DNA methylation, which is responsible for gene silencing, is a rather stable covalent modification. This suggests that Arc expression has been repressed and may consequently remain inhibited for a prolonged period post-ECS. Arc plays a critical role in the maintenance phase of long-term potentiation (LTP) and consolidation of memory in the rat brain. Thus, this study is one of the first to demonstrate DNA methylation as a regulator of ECS-induced gene expression and it provides a molecular link to the memory deficits observed after ECS.
Collapse
Affiliation(s)
- Mads Dyrvig
- Laboratory of Neurobiology, Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | | | | | | | | | | |
Collapse
|
31
|
Marek GJ. Activation of adenosine₁ receptors induces antidepressant-like, anti-impulsive effects on differential reinforcement of low-rate 72-s behavior in rats. J Pharmacol Exp Ther 2012; 341:564-70. [PMID: 22323824 PMCID: PMC3336810 DOI: 10.1124/jpet.112.191718] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 02/07/2012] [Indexed: 11/22/2022] Open
Abstract
Stress and psychiatric illness have been associated with a dysregulation of glutamatergic neurotransmission. Recently, positive allosteric modulators (PAMs) of the metabotropic glutamate 2 (mGlu₂) receptor have been found to exert antidepressant-like activity in rats performing under a differential reinforcement of low rate (DRL) 72-s schedule. An autoreceptor role at glutamatergic synapses is the most salient physiological role played by the mGlu₂ receptor. Adenosine A₁ receptors play a heteroreceptor role at many of the same forebrain synapses where mGlu₂ autoreceptors are found. Agonists and/or PAMs of mGlu₂ receptors act similarly to adenosine A₁ receptor agonists with respect to a wide range of electrophysiological, biochemical, and behavioral responses mediated by limbic circuitry thought to play a role in the pathophysiology of neuropsychiatric disease and to mediate therapeutic drug effects. Therefore, the role of adenosine A₁ receptor activation on rat DRL 72-s behavior was explored to provide preclinical evidence consistent or inconsistent with potential antidepressant effects. The adenosine A₁ receptor agonist N⁶-cyclohexyladenosine (CHA) increased the reinforcement rate, decreased the response rate, and induced a rightward shift in inter-response time distributions in a dose-dependent fashion similar to most known antidepressant drugs. The adenosine A₁ receptor antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) blocked these antidepressant-like effects. These novel observations with CHA and DPCPX suggest that activation of adenosine A₁ receptors could contribute to antidepressant effects, in addition to previous preclinical reports of anxiolytic and antipsychotic effects. By implication, targeting a dysregulated glutamatergic system may be an important principle in discovering novel antidepressant agents that may also possess anti-impulsive activity.
Collapse
Affiliation(s)
- Gerard J Marek
- Abbott Laboratories, Neuroscience Development, GPRD, R48B, Bldg. AP04-1, 100 Abbott Park Road, Abbott Park, IL 60064-6075, USA.
| |
Collapse
|
32
|
Santini M, Klein A, El-Sayed M, Ratner C, Knudsen G, Mikkelsen J, Aznar S. Novelty-induced activity-regulated cytoskeletal-associated protein (Arc) expression in frontal cortex requires serotonin 2A receptor activation. Neuroscience 2011; 190:251-7. [DOI: 10.1016/j.neuroscience.2011.05.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2011] [Revised: 05/12/2011] [Accepted: 05/19/2011] [Indexed: 01/12/2023]
|
33
|
Bozzi Y, Dunleavy M, Henshall DC. Cell signaling underlying epileptic behavior. Front Behav Neurosci 2011; 5:45. [PMID: 21852968 PMCID: PMC3151612 DOI: 10.3389/fnbeh.2011.00045] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 07/13/2011] [Indexed: 12/12/2022] Open
Abstract
Epilepsy is a complex disease, characterized by the repeated occurrence of bursts of electrical activity (seizures) in specific brain areas. The behavioral outcome of seizure events strongly depends on the brain regions that are affected by overactivity. Here we review the intracellular signaling pathways involved in the generation of seizures in epileptogenic areas. Pathways activated by modulatory neurotransmitters (dopamine, norepinephrine, and serotonin), involving the activation of extracellular-regulated kinases and the induction of immediate early genes (IEGs) will be first discussed in relation to the occurrence of acute seizure events. Activation of IEGs has been proposed to lead to long-term molecular and behavioral responses induced by acute seizures. We also review deleterious consequences of seizure activity, focusing on the contribution of apoptosis-associated signaling pathways to the progression of the disease. A deep understanding of signaling pathways involved in both acute- and long-term responses to seizures continues to be crucial to unravel the origins of epileptic behaviors and ultimately identify novel therapeutic targets for the cure of epilepsy.
Collapse
Affiliation(s)
- Yuri Bozzi
- Laboratory of Molecular Neuropathology, Centre for Integrative Biology, University of Trento Trento, Italy
| | | | | |
Collapse
|
34
|
Benneyworth MA, Smith RL, Sanders-Bush E. Chronic phenethylamine hallucinogen treatment alters behavioral sensitivity to a metabotropic glutamate 2/3 receptor agonist. Neuropsychopharmacology 2008; 33:2206-16. [PMID: 17957214 DOI: 10.1038/sj.npp.1301600] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent clinical studies in schizophrenic patients show that a selective agonist of group II metabotropic glutamate (mGlu) receptors has robust efficacy in treating positive and negative symptoms. Group II mGlu receptor agonists also modulate the in vivo activity of psychotomimetic drugs, reducing the ability of psychotomimetic hallucinogens to increase glutamatergic transmission. The use of mouse models provides an opportunity to investigate the dynamic action that mGlu2/3 receptors play in regulating the behavioral effects of hallucinogen-induced glutamatergic neurotransmission using genetic as well as pharmacological strategies. The current study sought to characterize the use of the two-lever drug discrimination paradigm in ICR (CD-1) mice, using the hallucinogenic 5-HT2A/2C receptor agonist (-)-2,5-dimethoxy-4-bromoamphetamine [(-)-DOB)] as a stimulus-producing drug. The (-)-DOB discriminative stimulus was dose-dependent, generalized to the hallucinogen lysergic acid diethylamide, and was potently blocked by the 5-HT2A receptor antagonist M100907. However, contrary to our prediction, the hallucinogen-induced discriminative stimulus was not regulated by mGlu2/3 receptors. In a series of follow-up studies using hallucinogen-induced head twitch response and phencyclidine-induced hyperlocomotion, it was additionally discovered that the repeated dosing regimen required for discrimination training attenuated the behavioral effects of the mGlu2/3 receptor agonist LY379268. Furthermore chronic studies, using a 14 day (-)-DOB treatment, confirmed that repeated hallucinogen treatment causes a loss of behavioral activity of mGlu2/3 receptors, likely resulting from persistent activation of mGlu2/3 receptors by a hallucinogen-induced hyperglutamatergic state.
Collapse
Affiliation(s)
- Michael A Benneyworth
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | |
Collapse
|
35
|
Zhang C, Marek GJ. AMPA receptor involvement in 5-hydroxytryptamine2A receptor-mediated pre-frontal cortical excitatory synaptic currents and DOI-induced head shakes. Prog Neuropsychopharmacol Biol Psychiatry 2008; 32:62-71. [PMID: 17728034 DOI: 10.1016/j.pnpbp.2007.07.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2007] [Revised: 07/03/2007] [Accepted: 07/04/2007] [Indexed: 11/23/2022]
Abstract
Glutamate plays an important role in the psychotomimetic effects of both channel blocking N-methyl D-aspartate (NMDA) receptor antagonists and hallucinogenic drugs which activate 5-hydroxytryptamine2A (5-HT2A) receptors. Previous work suggested that activation of non-NMDA ionotropic glutamate receptors mediates the effects of 5-HT-induced excitatory post-synaptic potentials/currents (EPSPs/EPSCs) when recording from layer V pyramidal cells in the rat medial pre-frontal cortex (mPFC). However, those effects are mediated by either alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate (AMPA) or kainate receptors of the iGluk5 subtype. To test whether activation of AMPA receptors is sufficient to mediate 5-HT-induced EPSCs, a 2,3-benzodiazepine that selectively blocks AMPA receptors was assessed. This selective AMPA receptor antagonist potently suppressed 5-HT-induced EPSCs. Since phenethylamine hallucinogens induce head shakes by activating 5-HT2A receptors in the mPFC and this action is modulated by glutamate, we also examined whether selective blockade of AMPA receptors would suppress DOI-induced head shakes. As predicted, we found that selective blockade of AMPA receptors suppressed DOI-induced head shakes. Given evidence that activation of AMPA receptors is an important downstream effect for both channel blocking NMDA receptor antagonists and phenethylamine hallucinogens, we also tested multiple doses of DOI with a sub-anesthetic dose of MK-801. Synergistic action between these two classes of psychotomimetic drugs was demonstrated by MK-801 enhancing DOI-induced head shakes and locomotor activity. These findings expand the dependence of both channel blocking NMDA receptor antagonists and phenethylamine hallucinogens on enhancing extracellular glutamate.
Collapse
Affiliation(s)
- Ce Zhang
- Department of Psychiatry, Yale University School of Medicine, Ribicoff Research Facilities of the Connecticut Mental Health Center, New Haven, Connecticut, USA
| | | |
Collapse
|
36
|
Jennings KA, Sheward WJ, Harmar AJ, Sharp T. Evidence that genetic variation in 5-HT transporter expression is linked to changes in 5-HT2A receptor function. Neuropharmacology 2007; 54:776-83. [PMID: 18241894 DOI: 10.1016/j.neuropharm.2007.12.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Revised: 12/06/2007] [Accepted: 12/06/2007] [Indexed: 01/05/2023]
Abstract
Variability in expression of the 5-HT transporter (5-HTT) gene in the human population has been associated with a range of behavioural phenotypes. The underlying mechanisms are unclear but may involve changes in 5-HT receptor levels and/or signalling. The present study used a novel 5-HTT overexpressing transgenic mouse to test the hypothesis that variability in 5-HTT expression may alter 5-HT(2A) receptor function. In wildtype mice, the 5-HT(2) receptor agonist DOI increased regional brain mRNA expression of two immediate early genes (c-fos and Arc), and induced head twitches, and both effects were abolished by pre-treatment with the 5-HT(2A) receptor antagonist MDL 100907. In 5-HTT overexpressing mice, DOI induced a greater increase in both c-fos and Arc mRNA expression in cortical brain regions, and more head twitches, compared to wildtype mice. Autoradiographic and in situ hybridisation experiments showed that 5-HT(2A) receptor binding sites and 5-HT(2A) receptor mRNA did not differ between transgenic and wildtype mice. Finally, the transgenic mice had lower regional brain 5-HT levels compared to wildtype mice. This depletion of 5-HT may underpin the increase in 5-HT(2A) receptor function because in wildtype mice 5-HT depletion using the 5-HT synthesis inhibitor, p-chlorophenylalanine, enhanced the head twitch response to DOI. These data demonstrate that elevated 5-HTT expression is accompanied by increased 5-HT(2A) receptor function, an effect possibly mediated by decreased availability of synaptic 5-HT. Variation in levels of 5-HTT expression may therefore be a source of variability in 5-HT(2A) receptor function, which may be an important modifier of 5-HTT-linked phenotypes.
Collapse
Affiliation(s)
- K A Jennings
- University Department of Pharmacology, Mansfield Road, Oxford, OX1 3QT, UK.
| | | | | | | |
Collapse
|
37
|
Thakker-Varia S, Krol JJ, Nettleton J, Bilimoria PM, Bangasser DA, Shors TJ, Black IB, Alder J. The neuropeptide VGF produces antidepressant-like behavioral effects and enhances proliferation in the hippocampus. J Neurosci 2007; 27:12156-67. [PMID: 17989282 PMCID: PMC3363962 DOI: 10.1523/jneurosci.1898-07.2007] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Revised: 09/14/2007] [Accepted: 09/17/2007] [Indexed: 01/16/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is upregulated in the hippocampus by antidepressant treatments, and BDNF produces antidepressant-like effects in behavioral models of depression. In our previous work, we identified genes induced by BDNF and defined their specific roles in hippocampal neuronal development and plasticity. To identify genes downstream of BDNF that may play roles in psychiatric disorders, we examined a subset of BDNF-induced genes also regulated by 5-HT (serotonin), which includes the neuropeptide VGF (nonacronymic). To explore the function of VGF in depression, we first investigated the expression of the neuropeptide in animal models of depression. VGF was downregulated in the hippocampus after both the learned helplessness and forced swim test (FST) paradigms. Conversely, VGF infusion in the hippocampus of mice subjected to FST reduced the time spent immobile for up to 6 d, thus demonstrating a novel role for VGF as an antidepressant-like agent. Recent evidence indicates that chronic treatment of rodents with antidepressants increases neurogenesis in the adult dentate gyrus and that neurogenesis is required for the behavioral effects of antidepressants. Our studies using [(3)H]thymidine and bromodeoxyuridine as markers of DNA synthesis indicate that chronic VGF treatment enhances proliferation of hippocampal progenitor cells both in vitro and in vivo with survival up to 21 d. By double immunocytochemical analysis of hippocampal neurons, we demonstrate that VGF increases the number of dividing cells that express neuronal markers in vitro. Thus, VGF may act downstream of BDNF and exert its effects as an antidepressant-like agent by enhancing neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Smita Thakker-Varia
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, and
| | - Jennifer Jernstedt Krol
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, and
| | - Jacob Nettleton
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, and
| | - Parizad M. Bilimoria
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, and
| | - Debra A. Bangasser
- Department of Psychology and Center for Collaborative Neuroscience, Rutgers University, Piscataway, New Jersey 08854
| | - Tracey J. Shors
- Department of Psychology and Center for Collaborative Neuroscience, Rutgers University, Piscataway, New Jersey 08854
| | - Ira B. Black
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, and
| | - Janet Alder
- Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, and
| |
Collapse
|
38
|
de Foubert G, O'Neill MJ, Zetterström TSC. Acute onset by 5-HT6-receptor activation on rat brain brain-derived neurotrophic factor and activity-regulated cytoskeletal-associated protein mRNA expression. Neuroscience 2007; 147:778-85. [PMID: 17560041 DOI: 10.1016/j.neuroscience.2007.04.045] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 04/25/2007] [Accepted: 04/29/2007] [Indexed: 11/21/2022]
Abstract
A number of previous studies have shown that chronic but not acute treatment with antidepressant drugs targeting the central 5-HT system, enhances mRNA expression for a number of genes including, brain-derived neurotrophic factor (BDNF) and the effector immediate early gene (IEG), activity-regulated, cytoskeletal-associated protein (Arc). The present study investigated the effects of 5-HT(6)-receptor activation on hippocampal and cortical levels of mRNA expression of BDNF and Arc in the rat. The selective 5-HT(6)-receptor agonist LY-586713 was administered acutely (0.1-10 mg/kg, s.c.) and mRNA levels of BDNF and Arc were measured 18 h later. Administration of LY-586713 caused a bell-shaped dose response on hippocampal BDNF mRNA expression, having no effect at 0.1 mg/kg, a significant up-regulation at 1 mg/kg and no effect at 10 mg/kg. The up-regulation in BDNF expression observed at 1 mg/kg was completely blocked by pre-treatment with the selective 5-HT(6)-receptor antagonist SB-271046 (10 mg/kg, s.c.). The effective dose (1 mg/kg) of LY-586713 on the induction of BDNF expression was also tested on Arc expression. Acute administration of LY-586713 at this dose caused marked increases of the Arc mRNA levels in cortical and hippocampal regions. These increases were also attenuated by SB-271046 (10 mg/kg) in all regions of the hippocampus, as well as the parietal cortex. However, in frontal cortical regions there was no attenuation by the antagonist. Moreover, SB-271046 alone increased Arc expression in these regions. The results presented here provide the first evidence for the involvement of the 5-HT(6) receptor in regulating BDNF and Arc mRNA expression, suggesting that LY-586713 has potential effects on neuronal plasticity. Overall, these findings suggest that, as opposed to more general 5-HT receptor activation by, for example, antidepressants, direct 5-HT(6)-receptor activation results in a more rapid rise in BDNF and Arc mRNA expression which does not require repeated administration.
Collapse
Affiliation(s)
- G de Foubert
- Leicester School of Pharmacy, Faculty of Health and Life Sciences, De Montfort University, The Gateway, Leicester, UK
| | | | | |
Collapse
|
39
|
Levine JB, Youngs RM, MacDonald ML, Chu M, Leeder AD, Berthiaume F, Konradi C. Isolation rearing and hyperlocomotion are associated with reduced immediate early gene expression levels in the medial prefrontal cortex. Neuroscience 2007; 145:42-55. [PMID: 17239545 DOI: 10.1016/j.neuroscience.2006.11.063] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2006] [Revised: 11/17/2006] [Accepted: 11/20/2006] [Indexed: 11/30/2022]
Abstract
Environmental deprivation contributes in important ways to the development of a wide range of psychiatric disorders. Isolation rearing of rodents, a model for environmental deprivation in humans, consistently produces hyperlocomotion, which provides a measurable parameter to study the underlying mechanisms of early adverse psychosocial stressors. Male Sprague-Dawley rat pups were separated from dams at postnatal (PN) day 20 and reared either in groups of three or in isolation. On PN 38, locomotion was assessed in the open field. On PN 46, rats were killed and gene expression patterns examined in the medial prefrontal cortex (mPFC). Isolation-reared rats displayed increased locomotor activity and decreased resting time in the open field. Specific gene expression patterns in the mPFC were associated with both isolation rearing and hyperlocomotive behavior in the open field. Genes involved in these expression patterns included immediate early genes (IEGs) and genes that regulate cell differentiation and apoptosis. The study of these genes could provide important insights into how abnormal early psychosocial events affect brain function and behavior.
Collapse
Affiliation(s)
- J B Levine
- Laboratory of Neuroplasticity, McLean Hospital, Belmont, MA 02478, USA.
| | | | | | | | | | | | | |
Collapse
|
40
|
Schiltz CA, Kelley AE, Landry CF. Acute stress and nicotine cues interact to unveil locomotor arousal and activity-dependent gene expression in the prefrontal cortex. Biol Psychiatry 2007; 61:127-35. [PMID: 16631128 PMCID: PMC1698504 DOI: 10.1016/j.biopsych.2006.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2005] [Revised: 01/30/2006] [Accepted: 03/06/2006] [Indexed: 11/16/2022]
Abstract
BACKGROUND This study examines the interactive effects of acute stress and nicotine-associated contextual cues on locomotor activity and activity-dependent gene expression in subregions of the prefrontal cortex. METHODS Locomotor activity of rats was measured in a context associated with either low-dose nicotine or saline administration with or without 5 minutes of pre-exposure to ferrets, a nonphysical stressor. After 45 minutes in the test environment, plasma corticosterone levels and mRNA levels of the immediate-early genes Arc, NGFI-B, and c-Fos in prefrontal and primary motor cortical subregions were measured. RESULTS Stress alone increased plasma corticosterone and prefrontal cortex gene expression. Low-dose nicotine cues had no effect on corticosterone levels nor did they elicit conditioned motor activation, and they caused minor elevations in gene expression. Stress and low-dose nicotine cues, however, interacted to elicit conditioned motor activation and further increases in early response gene expression in prefrontal but not in the primary motor cortical subregions. CONCLUSIONS Stress interacts with nicotine-associated cues to uncover locomotor arousal, a state associated with prefrontal neuronal activation and immediate early gene expression. Thus, in nicotine-experienced individuals, stress may be an important determinant of subjective reactivity and prefrontal cortex activation that occurs in response to nicotine-associated cues.
Collapse
MESH Headings
- AIDS-Related Complex/genetics
- AIDS-Related Complex/metabolism
- Analysis of Variance
- Animals
- Behavior, Animal/drug effects
- Corticosterone/blood
- Cues
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Dose-Response Relationship, Drug
- Gene Expression/drug effects
- Gene Expression/physiology
- In Situ Hybridization/methods
- Male
- Motor Activity/drug effects
- Motor Activity/physiology
- Nicotine/administration & dosage
- Nicotinic Agonists/administration & dosage
- Nuclear Receptor Subfamily 4, Group A, Member 1
- Prefrontal Cortex/drug effects
- Prefrontal Cortex/metabolism
- Prefrontal Cortex/physiopathology
- Proto-Oncogene Proteins c-fos/genetics
- Proto-Oncogene Proteins c-fos/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Steroid/genetics
- Receptors, Steroid/metabolism
- Stress, Physiological/metabolism
- Stress, Physiological/physiopathology
- Time Factors
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- Craig A Schiltz
- Neuroscience Training Program, University of Wisconsin-Madison Medical School, Madison, Wisconsin 53719, USA
| | | | | |
Collapse
|
41
|
Millan MJ. Multi-target strategies for the improved treatment of depressive states: Conceptual foundations and neuronal substrates, drug discovery and therapeutic application. Pharmacol Ther 2006; 110:135-370. [PMID: 16522330 DOI: 10.1016/j.pharmthera.2005.11.006] [Citation(s) in RCA: 397] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2005] [Accepted: 11/28/2005] [Indexed: 12/20/2022]
Abstract
Major depression is a debilitating and recurrent disorder with a substantial lifetime risk and a high social cost. Depressed patients generally display co-morbid symptoms, and depression frequently accompanies other serious disorders. Currently available drugs display limited efficacy and a pronounced delay to onset of action, and all provoke distressing side effects. Cloning of the human genome has fuelled expectations that symptomatic treatment may soon become more rapid and effective, and that depressive states may ultimately be "prevented" or "cured". In pursuing these objectives, in particular for genome-derived, non-monoaminergic targets, "specificity" of drug actions is often emphasized. That is, priority is afforded to agents that interact exclusively with a single site hypothesized as critically involved in the pathogenesis and/or control of depression. Certain highly selective drugs may prove effective, and they remain indispensable in the experimental (and clinical) evaluation of the significance of novel mechanisms. However, by analogy to other multifactorial disorders, "multi-target" agents may be better adapted to the improved treatment of depressive states. Support for this contention is garnered from a broad palette of observations, ranging from mechanisms of action of adjunctive drug combinations and electroconvulsive therapy to "network theory" analysis of the etiology and management of depressive states. The review also outlines opportunities to be exploited, and challenges to be addressed, in the discovery and characterization of drugs recognizing multiple targets. Finally, a diversity of multi-target strategies is proposed for the more efficacious and rapid control of core and co-morbid symptoms of depression, together with improved tolerance relative to currently available agents.
Collapse
Affiliation(s)
- Mark J Millan
- Institut de Recherches Servier, Centre de Recherches de Croissy, Psychopharmacology Department, 125, Chemin de Ronde, 78290-Croissy/Seine, France.
| |
Collapse
|
42
|
Winter JC, Eckler JR, Rice KC, Rabin RA. Serotonergic/glutamatergic interactions: potentiation of phencyclidine-induced stimulus control by citalopram. Pharmacol Biochem Behav 2005; 81:694-700. [PMID: 15970314 PMCID: PMC1224745 DOI: 10.1016/j.pbb.2005.03.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2004] [Accepted: 03/14/2005] [Indexed: 10/25/2022]
Abstract
Previous investigations in our laboratory have found that the stimulus effects of the hallucinogenic serotonergic agonists DOM and LSD are potentiated by phencyclidine [PCP], a non-competitive NMDA antagonist. Also suggestive of behaviorally significant serotonergic/glutamatergic interactions is our finding that stimulus control by both PCP and LSD is partially antagonized by the mGlu2/3 agonist, LY 379268. These observations coupled with the fact that the stimulus effects of LSD and DOM are potentiated by selective serotonin reuptake inhibitors [SSRIs] led us in the present investigation to test the hypothesis that stimulus control by PCP is potentiated by the SSRI, citalopram. Stimulus control was established with PCP [3.0 mg/kg; 30 min pretreatment time] in a group of 12 rats. A two-lever, fixed ratio 10, positively reinforced task with saline controls was employed. Potentiation by citalopram of an intermediate dose of PCP was observed. In an attempt to establish the mechanism by which citalopram might interact with PCP, subsequent experiments examined the effects on that interaction of antagonists at serotonergic receptors. It was found that the selective 5-HT2C-selective antagonists, SDZ SER 082 and SB 242084, significantly, albeit only partially, blocked the effects of citalopram on PCP. In agreement with our previous conclusions regarding the interaction of citalopram with DOM, the present data suggest that potentiation of the stimulus effects of PCP by citalopram are mediated in part by agonist activity at 5-HT2C receptors.
Collapse
Affiliation(s)
- J C Winter
- Department of Pharmacology and Toxicology, School of Medicine and Biomedical Sciences, 102 Farber Hall, State University of New York at Buffalo, Buffalo, NY 14214-3000, USA.
| | | | | | | |
Collapse
|
43
|
Matsumoto K, Morishige R, Murakami Y, Tohda M, Takayama H, Sakakibara I, Watanabe H. Suppressive effects of isorhynchophylline on 5-HT2A receptor function in the brain: Behavioural and electrophysiological studies. Eur J Pharmacol 2005; 517:191-9. [PMID: 15963493 DOI: 10.1016/j.ejphar.2005.05.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2005] [Accepted: 05/20/2005] [Indexed: 11/26/2022]
Abstract
Isorhynchophylline is a major oxindole alkaloid found in Uncaria species which have long been used in traditional Chinese medicine. Here, we investigated the effects of isorhynchophylline and isorhynchophylline-related alkaloids on 5-hydroxytryptamine (5-HT) receptor-mediated behavioural responses in mice and 5-HT-evoked current responses in Xenopus oocytes expressing 5-HT2A or 5-HT2C receptors. Isorhynchophylline dose-dependently inhibited 5-HT2A receptor-mediated head-twitch but not 5-HT1A receptor-mediated head-weaving responses evoked by 5-methoxy-N,N-dimethyltryptamine. Pretreatment with reserpine, a monoamine-depleting agent, enhanced the head-twitching, but did not influence the effect of isorhynchophylline on the behavioural response. Isocorynoxeine, an isorhynchophylline-related alkaloid in which the configuration of the oxindole moiety is the same as in isorhynchophylline, also reduced the head-twitch response in reserpinized mice over the same dose range as isorhynchophylline, while both rhynchophylline and corynoxeine, stereoisomers of isorhynchophylline and isocorynoxeine, did not. None of the alkaloids tested had an effect on meta-chlorophenylpiperazine-induced hypolocomotion, a 5-HT2C receptor-mediated behavioural response. In experiments in vitro, isorhynchophylline and isocorynoxeine dose-dependently and competitively inhibited 5-HT-evoked currents in Xenopus oocytes expressing 5-HT2A receptors, but had less of a suppressive effect on those in oocytes expressing 5-HT2C receptors. These results indicate that isorhynchophylline and isocorynoxeine preferentially suppress 5-HT2A receptor function in the brain probably via a competitive antagonism at 5-HT2A receptor sites and that the configuration of the oxindole moiety of isorhynchophylline is essential for their antagonistic activity at the 5-HT2A receptor.
Collapse
MESH Headings
- Alkaloids/pharmacology
- Aminopyridines/pharmacology
- Animals
- Behavior, Animal/drug effects
- Brain/drug effects
- Brain/physiology
- Dose-Response Relationship, Drug
- Female
- Indole Alkaloids
- Ketanserin/pharmacology
- Male
- Membrane Potentials/drug effects
- Methoxydimethyltryptamines/pharmacology
- Mianserin/pharmacology
- Mice
- Mice, Inbred ICR
- Motor Activity/drug effects
- Oocytes/drug effects
- Oocytes/physiology
- Oxindoles
- Patch-Clamp Techniques
- Piperazines/pharmacology
- Rats
- Receptor, Serotonin, 5-HT2A/genetics
- Receptor, Serotonin, 5-HT2A/physiology
- Receptor, Serotonin, 5-HT2C/genetics
- Receptor, Serotonin, 5-HT2C/physiology
- Serotonin/pharmacology
- Serotonin 5-HT2 Receptor Antagonists
- Serotonin Antagonists/pharmacology
- Xenopus
Collapse
Affiliation(s)
- Kinzo Matsumoto
- Division of Medicinal Pharmacology, Institute of Natural Medicine, Toyama Medical and Pharmaceutical University, 2630 Sugitani, Toyama 930-0194, Japan.
| | | | | | | | | | | | | |
Collapse
|
44
|
Tordera RM, Pei Q, Sharp T. Evidence for increased expression of the vesicular glutamate transporter, VGLUT1, by a course of antidepressant treatment. J Neurochem 2005; 94:875-83. [PMID: 15992385 DOI: 10.1111/j.1471-4159.2005.03192.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The therapeutic effect of a course of antidepressant treatment is believed to involve a cascade of neuroadaptive changes in gene expression leading to increased neural plasticity. Because glutamate is linked to mechanisms of neural plasticity, this transmitter may play a role in these changes. This study investigated the effect of antidepressant treatment on expression of the vesicular glutamate transporters, VGLUT1-3 in brain regions of the rat. Repeated treatment with fluoxetine, paroxetine or desipramine increased VGLUT1 mRNA abundance in frontal, orbital, cingulate and parietal cortices, and regions of the hippocampus. Immunoautoradiography analysis showed that repeated antidepressant drug treatment increased VGLUT1 protein expression. Repeated electroconvulsive shock (ECS) also increased VGLUT1 mRNA abundance in regions of the cortex and hippocampus compared to sham controls. The antidepressant drugs and ECS did not alter VGLUT1 mRNA abundance after acute administration, and no change was detected after repeated treatment with the antipsychotic agents, haloperidol and chlorpromazine. In contrast to VGLUT1, the different antidepressant treatments did not commonly increase the expression of VGLUT2 or VGLUT3 mRNA. These data suggest that a course of antidepressant drug or ECS treatment increases expression of VGLUT1, a key gene involved in the regulation of glutamate secretion.
Collapse
|
45
|
Millan MJ. The role of monoamines in the actions of established and "novel" antidepressant agents: a critical review. Eur J Pharmacol 2005; 500:371-84. [PMID: 15464046 DOI: 10.1016/j.ejphar.2004.07.038] [Citation(s) in RCA: 195] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2004] [Indexed: 11/19/2022]
Abstract
Monoaminergic pathways are highly responsive to aversive stimuli and play a crucial role in the control of affect, cognition, endocrine secretion, chronobiotic rhythms, appetite, and motor function, all of which are profoundly disrupted in depressive states. Accordingly, a perturbation of monoaminergic transmission is implicated in the aetiology of depressive disorders, and all clinically available antidepressants increase corticolimbic availability of monoamines. However, their limited efficacy, delayed onset of action, and undesirable side effects underlie ongoing efforts to identify improved therapeutic agents. Sequencing the human genome has raised the hope not only of better symptomatic control of depression, but even of the prevention or cure of depressive states. In the pursuit of these goals, there is currently a tendency to focus on selective ligands of "novel" nonmonoaminergic targets. However, certain classes of novel agent (such as neurokinin(1) receptor antagonists) indirectly modulate the activity of monoaminergic networks. Others may act "downstream" of them, converging onto common cellular substrates controlling gene expression, synaptic plasticity, and neurogenesis. Further, by analogy to the broad-based actions of currently employed drugs, multitarget agents may be better adapted than selective agents to the management of depression-a complex disorder with hereditary, developmental, and environmental origins. It is, thus, important to continue the creative exploration of clinically validated and innovative monoaminergic strategies within a multitarget framework. In this light, drugs combining monoaminergic and nonmonoaminergic mechanisms of action may be of particular interest. The present article provides a critical overview of monoaminergic strategies for the treatment of depressive states, both established and under development, and discusses interactions of novel "nonmonoaminergic" antidepressants with monoaminergic mechanisms.
Collapse
Affiliation(s)
- Mark J Millan
- Psychopharmacology Department, IdR Servier, 125 Chemin de Ronde, Croissy/Seine, Paris 78290, France.
| |
Collapse
|