1
|
Krauklis SA, Towers AE, York JM, Baynard T, Gainey SJ, Freund GG, Steelman AJ. Mouse Testing Methods in Psychoneuroimmunology: Measuring Behavioral Responses. Methods Mol Biol 2025; 2868:163-203. [PMID: 39546231 DOI: 10.1007/978-1-0716-4200-9_10] [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: 11/17/2024]
Abstract
The field of psychoneuroimmunology (PNI) aims to uncover the processes and consequences of nervous, immune, and endocrine system relationships. Behavior is a consequence of such interactions and manifests from a complex interweave of factors including immune-to-neural and neural-to-immune communication. Often the signaling molecules involved during a particular episode of neuroimmune activation are not known, but behavioral response provides evidence that bioactives such as neurotransmitters and cytokines are perturbed. Immunobehavioral phenotyping is a first-line approach when examining the neuroimmune system and its reaction to immune stimulation or suppression. Behavioral response is significantly more sensitive than direct measurement of a single specific bioactive and can quickly and efficiently rule in or out relevance of a particular immune challenge or therapeutic to neuroimmunity. Classically, immunobehavioral research was focused on sickness symptoms related to bacterial infection, but neuroimmune activation is now a recognized complication of diseases and disorders ranging from cancer to diabesity to Alzheimer's. Immunobehaviors include lethargy, loss of appetite, and disinterest in social activity/surrounding environment. In addition, neuroimmune activation can diminish physical activity, precipitate feelings of depression and anxiety, and impair cognitive and executive function. Provided is a detailed overview of behavioral tests frequently used to examine neuroimmune activation in mice with a special emphasis on pre-experimental conditions that can confound or prevent successful immunobehavioral experimentation.
Collapse
Affiliation(s)
- Steven A Krauklis
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Albert E Towers
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Jason M York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Tracy Baynard
- Academic Affairs, University of Massachusetts-Boston, Boston, MA, USA
| | - Stephen J Gainey
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Gregory G Freund
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Andrew J Steelman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Champaign, IL, USA.
| |
Collapse
|
2
|
Courault P, Bouvard S, Bouillot C, Zimmer L, Lancelot S. Preclinical investigation of the effect of stress on the binding of [ 18F]F13640, a 5-HT 1A radiopharmaceutical. Nucl Med Biol 2024; 138-139:108942. [PMID: 39151306 DOI: 10.1016/j.nucmedbio.2024.108942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND [18F]F13640 is a new PET radiopharmaceutical for brain molecular imaging of serotonin 5-HT1A receptors. Since we intend to use this radiopharmaceutical in psychiatric studies, it is crucial to establish possible sensitivity modification of 5-HT1A receptors availability during an acute stress exposure. In this study, we first assessed the cerebrometabolic effects of a new animal model of stress with [18F]FDG and then proceeded to test for effects of this model on the cerebral binding of [18F]F13640, a 5-HT1A receptors PET radiopharmaceutical. METHODS Four groups of male Sprague-Dawley were used to identify the optimal model: "stressed group" (n = 10), "post-traumatic stress disorder (PTSD) group" (n = 9) and "restraint group" (n = 8), compared with a control group (n = 8). All rats performed neuroimaging [18F]FDG μPET-CT to decipher which model was the most appropriate to test effects of stress on radiotracer binding. Subsequently, a group of rats (n = 10) underwent two PET imaging acquisitions (baseline and PTSD condition) using the PET radiopharmaceutical [18F]F13640 to assess influence of stress on its binding. Voxel-based analysis was performed to assess [18F]FDG or [18F]F13640 changes. RESULTS In [18F]FDG experiments, the PTSD group showed a pattern of cerebrometabolic activation in various brain regions previously implicated in stress (amygdala, perirhinal cortex, olfactory bulb and caudate). [18F]F13640 PET scans showed increased radiotracer binding in the PTSD condition in caudate nucleus and brainstem. CONCLUSIONS The present study demonstrated stress-induced cerebrometabolic activation or inhibition of various brain regions involved in stress model. Applying this model to our radiotracer, [18F]F13640 showed few influence of stress on its binding. This will enable to rule out any confounding effect of stress during imaging studies.
Collapse
Affiliation(s)
- Pierre Courault
- Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, Univ. Lyon 1, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP-Imaging platform, Groupement Hospitalier Est, Bron, France.
| | - Sandrine Bouvard
- Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, Univ. Lyon 1, Lyon, France
| | | | - Luc Zimmer
- Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, Univ. Lyon 1, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP-Imaging platform, Groupement Hospitalier Est, Bron, France
| | - Sophie Lancelot
- Lyon Neuroscience Research Center (CRNL), CNRS UMR5292, INSERM U1028, Univ. Lyon 1, Lyon, France; Hospices Civils de Lyon (HCL), Lyon, France; CERMEP-Imaging platform, Groupement Hospitalier Est, Bron, France
| |
Collapse
|
3
|
Bonanno SL, Sanfilippo P, Eamani A, Sampson MM, Kandagedon B, Li K, Burns GD, Makar ME, Zipursky SL, Krantz DE. Constitutive and Conditional Epitope Tagging of Endogenous G-Protein-Coupled Receptors in Drosophila. J Neurosci 2024; 44:e2377232024. [PMID: 38937100 PMCID: PMC11326870 DOI: 10.1523/jneurosci.2377-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/30/2024] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
To visualize the cellular and subcellular localization of neuromodulatory G-protein-coupled receptors in Drosophila, we implement a molecular strategy recently used to add epitope tags to ionotropic receptors at their endogenous loci. Leveraging evolutionary conservation to identify sites more likely to permit insertion of a tag, we generated constitutive and conditional tagged alleles for Drosophila 5-HT1A, 5-HT2A, 5-HT2B, Oct β 1R, Oct β 2R, two isoforms of OAMB, and mGluR The conditional alleles allow for the restricted expression of tagged receptor in specific cell types, an option not available for any previous reagents to label these proteins. We show expression patterns for these receptors in female brains and that 5-HT1A and 5-HT2B localize to the mushroom bodies (MBs) and central complex, respectively, as predicted by their roles in sleep. By contrast, the unexpected enrichment of Octβ1R in the central complex and of 5-HT1A and 5-HT2A to nerve terminals in lobular columnar cells in the visual system suggest new hypotheses about their functions at these sites. Using an additional tagged allele of the serotonin transporter, a marker of serotonergic tracts, we demonstrate diverse spatial relationships between postsynaptic 5-HT receptors and presynaptic 5-HT neurons, consistent with the importance of both synaptic and volume transmission. Finally, we use the conditional allele of 5-HT1A to show that it localizes to distinct sites within the MBs as both a postsynaptic receptor in Kenyon cells and a presynaptic autoreceptor.
Collapse
Affiliation(s)
- Shivan L Bonanno
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Piero Sanfilippo
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095
- Howard Hughes Medical Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095
| | - Aditya Eamani
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Maureen M Sampson
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Binu Kandagedon
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Kenneth Li
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Giselle D Burns
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Marylyn E Makar
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - S Lawrence Zipursky
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California 90095
- Howard Hughes Medical Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California 90095
| | - David E Krantz
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| |
Collapse
|
4
|
Francis B, Ganasan VA, Sulaiman ARB. Brexpiprazole Attenuates Aggression, Suicidality and Substance Use in Borderline Personality Disorder: A Case Series. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:283. [PMID: 38399570 PMCID: PMC10890360 DOI: 10.3390/medicina60020283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/30/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024]
Abstract
Background: Borderline personality disorder (BPD) is a heterogeneous and highly comorbid disorder. Suicidality, aggression and substance abuse are common presentations of BPD. Our case series is the first to highlight the role of brexpiprazole in improving these symptoms in patients with BPD. Case presentation: We describe three cases demonstrating the role of brexpiprazole in improving BPD's prominent features and comorbidities. All cases improved when brexpiprazole was added to their treatment regime. Case 1: A 26-year-old woman who was diagnosed with BPD and cyclothymia, presented to the psychiatric emergency unit with impulsive suicidal behaviour. Case 2: A 43-year-old woman suffering from BPD sought help due to her violent behaviour and emotional dysregulation. Case 3: A 22-year-old woman with underlying attention deficit and hyperactivity disorder, polysubstance use disorder and BPD presented with dysregulated emotions. Conclusions: Our case series provides anecdotal evidence of the potential role of brexpiprazole in attenuating suicidality, aggression and substance abuse in patients with BPD. We postulate that brexpiprazole's high affinity for the 5HT1A/5HT2A receptors, coupled with its low intrinsic effect on the D2/D3 receptor system, is fundamental in its actions to stabilise the aberrant dopaminergic and serotonergic signalling in BPD. Future research should focus on well-designed clinical trials investigating the efficacy of brexpiprazole in patients with BPD.
Collapse
Affiliation(s)
- Benedict Francis
- Department of Psychiatry, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Vijay A/L Ganasan
- Department of Psychiatry and Mental Health, Hospital Tuanku Ja’afar, Seremban 70300, Malaysia;
| | | |
Collapse
|
5
|
Bonanno SL, Sanfilippo P, Eamani A, Sampson MM, Binu K, Li K, Burns GD, Makar ME, Zipursky SL, Krantz DE. Constitutive and conditional epitope-tagging of endogenous G protein coupled receptors in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.27.573472. [PMID: 38234787 PMCID: PMC10793450 DOI: 10.1101/2023.12.27.573472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
To visualize the cellular and subcellular localization of neuromodulatory G-protein coupled receptors (GPCRs) in Drosophila , we implement a molecular strategy recently used to add epitope tags to ionotropic receptors at their endogenous loci. Leveraging evolutionary conservation to identify sites more likely to permit insertion of a tag, we generated constitutive and conditional tagged alleles for Drosophila 5-HT1A, 5-HT2A, 5-HT2B, Octβ1R, Octβ2R, two isoforms of OAMB, and mGluR. The conditional alleles allow for the restricted expression of tagged receptor in specific cell types, an option not available for any previous reagents to label these proteins. We show that 5-HT1A and 5-HT2B localize to the mushroom bodies and central complex respectively, as predicted by their roles in sleep. By contrast, the unexpected enrichment of Octβ1R in the central complex and of 5-HT1A and 5-HT2A to nerve terminals in lobular columnar cells in the visual system suggest new hypotheses about their function at these sites. Using an additional tagged allele of the serotonin transporter, a marker of serotonergic tracts, we demonstrate diverse spatial relationships between postsynaptic 5-HT receptors and presynaptic 5-HT neurons, consistent with the importance of both synaptic and volume transmission. Finally, we use the conditional allele of 5-HT1A to show that it localizes to distinct sites within the mushroom bodies as both a postsynaptic receptor in Kenyon cells and a presynaptic autoreceptor. Significance Statement In Drosophila , despite remarkable advances in both connectomic and genomic studies, antibodies to many aminergic GPCRs are not available. We have overcome this obstacle using evolutionary conservation to identify loci in GPCRs amenable to epitope-tagging, and CRISPR/Cas9 genome editing to generated eight novel lines. This method also may be applied to other GPCRs and allows cell-specific expression of the tagged locus. We have used the tagged alleles we generated to address several questions that remain poorly understood. These include the relationship between pre- and post-synaptic sites that express the same receptor, and the use of relatively distant targets by pre-synaptic release sites that may employ volume transmission as well as standard synaptic signaling.
Collapse
|
6
|
Alabdali R, Franchini L, Orlandi C. G α Protein Signaling Bias at Serotonin 1A Receptor. Mol Pharmacol 2023; 104:230-238. [PMID: 37567783 PMCID: PMC10586511 DOI: 10.1124/molpharm.123.000722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/11/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Serotonin 1A receptor (5-HT1AR) is a clinically relevant target because of its involvement in several central and peripheral functions, including sleep, temperature homeostasis, processing of emotions, and response to stress. As a G protein coupled receptor (GPCR) activating numerous Gα i/o/z family members, 5-HT1AR can potentially modulate multiple intracellular signaling pathways in response to different therapeutics. Here, we applied a cell-based bioluminescence resonance energy transfer assay to quantify how ten structurally diverse 5-HT1AR agonists exert biased signaling by differentially stimulating Gα i/o/z family members. Our concentration-response analysis of the activation of each Gα i/o/z protein revealed unique potency and efficacy profiles of selected agonists when compared with the reference 5-hydroxytryptamine, serotonin. Overall, our analysis of signaling bias identified groups of ligands sharing comparable G protein activation selectivity and also drugs with unique selectivity profiles. We observed, for example, a strong bias of F-15599 toward the activation of Gα i3 that was unique among the agonists tested: we found a biased factor of +2.19 when comparing the activation of Gα i3 versus Gα i2 by F-15599, while it was -0.29 for 8-hydroxy-2-(di-n-propylamino) tetralin. Similarly, vortioxetine showed a biased factor of +1.06 for Gα z versus Gα oA, while it was -1.38 for vilazodone. Considering that alternative signaling pathways are regulated downstream of each Gα protein, our data suggest that the unique pharmacological properties of the tested agonists could result in multiple unrelated cellular outcomes. Further investigation is needed to reveal how this type of ligand bias could affect cellular responses and to illuminate the molecular mechanisms underlying therapeutic profile and side effects of each drug. SIGNIFICANCE STATEMENT: Serotonin 1a receptor (5-HT1AR) activates several members of the Gi/o/z protein family. Here, we examined ten structurally diverse and clinically relevant agonists acting on 5-HT1AR and identified distinctive bias patterns among G proteins. Considering the diversity of their intracellular effectors and signaling properties, this data reveal novel mechanisms underlying both therapeutic and undesirable effects.
Collapse
Affiliation(s)
- Rana Alabdali
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY
| | - Luca Franchini
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
7
|
Yu H, Yu B, Qin X, Shan W. A unique inflammation-related mechanism by which high-fat diets induce depression-like behaviors in mice. J Affect Disord 2023; 339:180-193. [PMID: 37437725 DOI: 10.1016/j.jad.2023.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/03/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
BACKGROUND High-fat diet (HFD) consumption is an important reason for promoting depression, but the mechanism is unclear. The present study aims to explore the relationship between metabolic disturbance and HFD-induced depression-like behaviors. METHODS Depression models were established by HFD consumption and chronic unpredictable mild stress (CUMS) in mice. Enzyme-linked immunosorbent assay, western blotting, real-time polymerase chain reaction, gas chromatography and metabolomic analysis were undertaken to investigate the 5-hydroxytryptamine (5-HT) system, neuroinflammation and to identify altered lipid metabolic pathways. RESULTS Depression-like behaviors, impaired 5-HT neurotransmission and disordered lipid metabolism were observed upon HFD consumption. Despite a similar reduction of high-density lipoprotein cholesterol in CUMS and HFD group, high levels of body low-density lipoprotein cholesterol in the HFD group could help distinguish HFD from CUMS. Levels of interleukin (IL)-1β, IL-6, tumor necrosis factor (TNF)-α and inflammation-related metabolites were increased in HFD mice, so a link between depression and inflammation was postulated. Different metabolites were enriched in the two groups. The linoleic acid (LA) metabolic pathway and expression of fatty acid desaturase (FADS)1 and FADS2 (key enzymes in LA metabolic pathway) were enhanced significantly in HFD mice compared with the control group. LIMITATIONS Causality analyses for HFD and inflammation-related features were not undertaken. CONCLUSIONS HFD-induced depression-like behaviors was characterized by more severely disordered metabolism of lipids (especially in the LA metabolic pathway) and increased levels of inflammatory mediators, which might be the reasons for the disturbance of serotonergic system in hippocampus.
Collapse
Affiliation(s)
- Haining Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, 310014, Hangzhou, China.
| | - Bixian Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, 310014, Hangzhou, China
| | - Xiuyuan Qin
- College of Pharmaceutical Science, Zhejiang University of Technology, 310014, Hangzhou, China
| | - Weiguang Shan
- College of Pharmaceutical Science, Zhejiang University of Technology, 310014, Hangzhou, China
| |
Collapse
|
8
|
Hou W, Huang S, Li L, Guo X, He Z, Shang S, Jia Z, Zhang L, Qu Y, Huang C, Li Y, Li Y, Lv Z, Tai F. Oxytocin treatments or activation of the paraventricular nucleus-the shell of nucleus accumbens pathway reduce adverse effects of chronic social defeat stress on emotional and social behaviors in Mandarin voles. Neuropharmacology 2023; 230:109482. [PMID: 36893984 DOI: 10.1016/j.neuropharm.2023.109482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 02/12/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023]
Abstract
Chronic social stress can cause psychological disease. Although oxytocin (OT) has been showed to modulate effects of chronic social defeat stress (CSDS) on emotional and social behaviors, however, how OT circuits mediate effects of CSDS on emotional and social abnormalities remains unclear. Here, we found that repeated intraperitoneal OT administration in the process of CSDS buffered adverse effects of CSDS on emotional and social behaviors in mandarin voles (Microtus mandarinus) of both sexes except no effect on depression-like behavior of males. Repeated OT treatments during CSDS prevented decrease of oxytocin receptors in nucleus accumbens (NAc) in females, but produced no effects on males. Furthermore, using designer receptors exclusively activated by designer drugs (DREADDs)-based chemogenetic tools, we determined that the activation of the paraventricular nucleus (PVN)-the shell of NAc (NAcs) projections before social defeat during CSDS process significantly prevented the increase of the anxiety-like behaviors and social avoidance induced by CSDS in both sexes, and reversed the depressive-like behaviors induced by CSDS only in females. Besides, optogenetic activation of PVN-NAcs projections after CSDS reduced anxiety-like behaviors and increased levels of sociality. Collectively, we suggest that PVN-NAcs projections modulate emotional and social behaviors during or after the process of CSDS sex-specifically, although AAV viruses did not specifically infect OT neurons. These findings offer potential targets for preventing or treating emotional and social disorders induced by chronic stress.
Collapse
Affiliation(s)
- Wenjuan Hou
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Shuying Huang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Lu Li
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Xing Guo
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Zhixiong He
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Shufeng Shang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China; College of Bioscience and Engineering, Shaanxi University of Technology, Hanzhong, 723000, China
| | - Ziyan Jia
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Lizi Zhang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Yishan Qu
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Caihong Huang
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Yin Li
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Yitong Li
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Zijian Lv
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China
| | - Fadao Tai
- Institute of Brain and Behavioral Sciences, College of Life Sciences, Shaanxi Normal University, Xi'an, 710062, China.
| |
Collapse
|
9
|
Siddique R, Awan FM, Nabi G, Khan S, Xue M. Chronic jet lag-like conditions dysregulate molecular profiles of neurological disorders in nucleus accumbens and prefrontal cortex. Front Neuroinform 2022; 16:1031448. [PMID: 36582489 PMCID: PMC9792783 DOI: 10.3389/fninf.2022.1031448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/22/2022] [Indexed: 12/14/2022] Open
Abstract
Background Patients with neurological disorders often display altered circadian rhythms. The disrupted circadian rhythms through chronic jetlag or shiftwork are thought to increase the risk and severity of human disease including, cancer, psychiatric, and related brain diseases. Results In this study, we investigated the impact of shiftwork or chronic jetlag (CJL) like conditions on mice's brain. Transcriptome profiling based on RNA sequencing revealed that genes associated with serious neurological disorders were differentially expressed in the nucleus accumbens (NAc) and prefrontal cortex (PFC). According to the quantitative PCR (qPCR) analysis, several key regulatory genes associated with neurological disorders were significantly altered in the NAc, PFC, hypothalamus, hippocampus, and striatum. Serotonin levels and the expression levels of serotonin transporters and receptors were significantly altered in mice treated with CJL. Conclusion Overall, these results indicate that CJL may increase the risk of neurological disorders by disrupting the key regulatory genes, biological functions, serotonin, and corticosterone. These molecular linkages can further be studied to investigate the mechanism underlying CJL or shiftwork-mediated neurological disorders in order to develop treatment strategies.
Collapse
Affiliation(s)
- Rabeea Siddique
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - Faryal Mehwish Awan
- Department of Medical Lab Technology, The University of Haripur, Haripur, Pakistan
| | - Ghulam Nabi
- Institute of Nature Conservation, Polish Academy of Sciences, Kraków, Poland
| | - Suliman Khan
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China,Department of Medical Lab Technology, The University of Haripur, Haripur, Pakistan,*Correspondence: Suliman Khan, ;
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China,Mengzhou Xue,
| |
Collapse
|
10
|
Dual Targeting Ligands-Histamine H 3 Receptor Ligands with Monoamine Oxidase B Inhibitory Activity-In Vitro and In Vivo Evaluation. Pharmaceutics 2022; 14:pharmaceutics14102187. [PMID: 36297622 PMCID: PMC9607599 DOI: 10.3390/pharmaceutics14102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/03/2022] [Accepted: 10/09/2022] [Indexed: 11/17/2022] Open
Abstract
The clinical symptoms of Parkinson’s disease (PD) appear when dopamine (DA) concentrations in the striatum drops to around 20%. Simultaneous inhibitory effects on histamine H3 receptor (H3R) and MAO B can increase DA levels in the brain. A series of compounds was designed and tested in vitro for human H3R (hH3R) affinity and inhibitory activity to human MAO B (hMAO B). Results showed different activity of the compounds towards the two biological targets. Most compounds had poor affinity for hH3R (Ki > 500 nM), but very good inhibitory potency for hMAO B (IC50 < 50 nM). After further in vitro testing (modality of MAO B inhibition, permeability in PAMPA assay, cytotoxicity on human astrocyte cell lines), the most promising dual-acting ligand, 1-(3-(4-(tert-butyl)phenoxy)propyl)-2-methylpyrrolidine (13: hH3R: Ki = 25 nM; hMAO B IC50 = 4 nM) was selected for in vivo evaluation. Studies in rats of compound 13, in a dose of 3 mg/kg of body mass, confirmed its antagonistic effects for H3R (decline in food and a water consumption), decline in MAO B activity (>90%) in rat cerebral cortex (CTX), and an increase in DA content in CTX and striatum. Moreover, compound 13 caused a slight increase in noradrenaline, but a reduction in serotonin concentration in CTX. Thus, compound 13 is a promising dual-active ligand for the potential treatment of PD although further studies are needed to confirm this.
Collapse
|
11
|
Barbee BR, Gourley SL. Brain systems in cocaine abstinence-induced anxiety-like behavior in rodents: A review. ADDICTION NEUROSCIENCE 2022; 2:100012. [PMID: 37485439 PMCID: PMC10361393 DOI: 10.1016/j.addicn.2022.100012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Cocaine use disorder (CUD) is a significant public health issue that generates substantial personal, familial, and economic burdens. Still, there are no FDA-approved pharmacotherapies for CUD. Cocaine-dependent individuals report anxiety during withdrawal, and alleviation of anxiety and other negative affective states may be critical for maintaining drug abstinence. However, the neurobiological mechanisms underlying abstinence-related anxiety in humans or anxiety-like behavior in rodents are not fully understood. This review summarizes investigations regarding anxiety-like behavior in mice and rats undergoing cocaine abstinence, as assessed using four of the most common anxiety-related assays: the elevated plus (or its derivative, the elevated zero) maze, open field test, light-dark transition test, and defensive burying task. We first summarize available evidence that cocaine abstinence generates anxiety-like behavior that persists throughout protracted abstinence. Then, we examine investigations concerning neuropeptide, neurotransmitter, and neuromodulator systems in cocaine abstinence-induced anxiety-like behavior. Throughout, we discuss how differences in sex, rodent strain, cocaine dose and dosing strategy and abstinence duration interact to generate anxiety-like behavior.
Collapse
Affiliation(s)
- Britton R. Barbee
- Graduate Program in Molecular and Systems Pharmacology,
Emory University
- Department of Pediatrics, Emory University School of
Medicine; Yerkes National Primate Research Center
| | - Shannon L. Gourley
- Graduate Program in Molecular and Systems Pharmacology,
Emory University
- Department of Pediatrics, Emory University School of
Medicine; Yerkes National Primate Research Center
| |
Collapse
|
12
|
Chemogenetics as a neuromodulatory approach to treating neuropsychiatric diseases and disorders. Mol Ther 2022; 30:990-1005. [PMID: 34861415 PMCID: PMC8899595 DOI: 10.1016/j.ymthe.2021.11.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/12/2021] [Accepted: 11/29/2021] [Indexed: 01/01/2023] Open
Abstract
Chemogenetics enables precise, non-invasive, and reversible modulation of neural activity via the activation of engineered receptors that are pharmacologically selective to endogenous or exogenous ligands. With recent advances in therapeutic gene delivery, chemogenetics is poised to support novel interventions against neuropsychiatric diseases and disorders. To evaluate its translational potential, we performed a scoping review of applications of chemogenetics that led to the reversal of molecular and behavioral deficits in studies relevant to neuropsychiatric diseases and disorders. In this review, we present these findings and discuss the potential and challenges for using chemogenetics as a precision medicine-based neuromodulation strategy.
Collapse
|
13
|
Baek SJ, Park JS, Kim J, Yamamoto Y, Tanaka-Yamamoto K. VTA-projecting cerebellar neurons mediate stress-dependent depression-like behaviors. eLife 2022; 11:72981. [PMID: 35156922 PMCID: PMC8843095 DOI: 10.7554/elife.72981] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 01/31/2022] [Indexed: 12/16/2022] Open
Abstract
Although cerebellar alterations have been implicated in stress symptoms, the exact contribution of the cerebellum to stress symptoms remains to be elucidated. Here, we demonstrated the crucial role of cerebellar neurons projecting to the ventral tegmental area (VTA) in the development of chronic stress-induced behavioral alterations in mice. Chronic chemogenetic activation of inhibitory Purkinje cells in crus I suppressed c-Fos expression in the DN and an increase in immobility in the tail suspension test or forced swimming test, which were triggered by chronic stress application. The combination of adeno-associated virus-based circuit mapping and electrophysiological recording identified network connections from crus I to the VTA via the dentate nucleus (DN) of the deep cerebellar nuclei. Furthermore, chronic inhibition of specific neurons in the DN that project to the VTA prevented stressed mice from showing such depression-like behavior, whereas chronic activation of these neurons alone triggered behavioral changes that were comparable with the depression-like behaviors triggered by chronic stress application. Our results indicate that the VTA-projecting cerebellar neurons proactively regulate the development of depression-like behavior, raising the possibility that cerebellum may be an effective target for the prevention of depressive disorders in human.
Collapse
Affiliation(s)
- Soo Ji Baek
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Jin Sung Park
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Jinhyun Kim
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Yukio Yamamoto
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Keiko Tanaka-Yamamoto
- Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| |
Collapse
|
14
|
Neural serotonergic circuits for controlling long-term voluntary alcohol consumption in mice. Mol Psychiatry 2022; 27:4599-4610. [PMID: 36195637 PMCID: PMC9531213 DOI: 10.1038/s41380-022-01789-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/05/2022] [Accepted: 09/09/2022] [Indexed: 12/14/2022]
Abstract
Alcohol-use-disorders are chronic relapsing illnesses, often co-morbid with anxiety. We have previously shown using the "drinking-in-the-dark" model in mice that the stimulation of the serotonin receptor 1A (5-HT1A) reduces ethanol binge-drinking behaviour and withdrawal-induced anxiety. The 5-HT1A receptor is located either on Raphe neurons as autoreceptors, or on target neurons as heteroreceptors. By combining a pharmacological approach with biased agonists targeting the 5-HT1A auto- or heteroreceptor and a chemogenetic approach (DREADDs), here we identified that ethanol-binge drinking behaviour is dependent on 5-HT1A autoreceptors and 5-HT neuronal function, with a transition from DRN-dependent regulation of short-term (6 weeks) ethanol intake, to MRN-dependent regulation after longer ethanol exposure (12 weeks). We further identified a serotonergic microcircuit (5-HTMRN→DG) originating from the MRN and projecting to the dentate gyrus (DG) of the hippocampus, that is specifically affected by, and modulates long-term ethanol consumption. The present study indicates that targeting Raphe nuclei 5-HT1A autoreceptors with agonists might represent an innovative pharmacotherapeutic strategy to combat alcohol abuse.
Collapse
|
15
|
Kang S, Jun S, Baek SJ, Park H, Yamamoto Y, Tanaka-Yamamoto K. Recent Advances in the Understanding of Specific Efferent Pathways Emerging From the Cerebellum. Front Neuroanat 2021; 15:759948. [PMID: 34975418 PMCID: PMC8716603 DOI: 10.3389/fnana.2021.759948] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/15/2021] [Indexed: 11/13/2022] Open
Abstract
The cerebellum has a long history in terms of research on its network structures and motor functions, yet our understanding of them has further advanced in recent years owing to technical developments, such as viral tracers, optogenetic and chemogenetic manipulation, and single cell gene expression analyses. Specifically, it is now widely accepted that the cerebellum is also involved in non-motor functions, such as cognitive and psychological functions, mainly from studies that have clarified neuronal pathways from the cerebellum to other brain regions that are relevant to these functions. The techniques to manipulate specific neuronal pathways were effectively utilized to demonstrate the involvement of the cerebellum and its pathways in specific brain functions, without altering motor activity. In particular, the cerebellar efferent pathways that have recently gained attention are not only monosynaptic connections to other brain regions, including the periaqueductal gray and ventral tegmental area, but also polysynaptic connections to other brain regions, including the non-primary motor cortex and hippocampus. Besides these efferent pathways associated with non-motor functions, recent studies using sophisticated experimental techniques further characterized the historically studied efferent pathways that are primarily associated with motor functions. Nevertheless, to our knowledge, there are no articles that comprehensively describe various cerebellar efferent pathways, although there are many interesting review articles focusing on specific functions or pathways. Here, we summarize the recent findings on neuronal networks projecting from the cerebellum to several brain regions. We also introduce various techniques that have enabled us to advance our understanding of the cerebellar efferent pathways, and further discuss possible directions for future research regarding these efferent pathways and their functions.
Collapse
Affiliation(s)
- Seulgi Kang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| | - Soyoung Jun
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| | - Soo Ji Baek
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| | - Heeyoun Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Yukio Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
| | - Keiko Tanaka-Yamamoto
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, South Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, South Korea
| |
Collapse
|
16
|
Vanderschuren LJMJ, Ahmed SH. Animal Models of the Behavioral Symptoms of Substance Use Disorders. Cold Spring Harb Perspect Med 2021; 11:cshperspect.a040287. [PMID: 32513674 PMCID: PMC8327824 DOI: 10.1101/cshperspect.a040287] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To more effectively manage substance use disorders, it is imperative to understand the neural, genetic, and psychological underpinnings of addictive behavior. To contribute to this understanding, considerable efforts have been made to develop translational animal models that capture key behavioral characteristics of addiction on the basis of DSM5 criteria of substance use disorders. In this review, we summarize empirical evidence for the occurrence of addiction-like behavior in animals. These symptoms include escalation of drug use, neurocognitive deficits, resistance to extinction, exaggerated motivation for drugs, increased reinstatement of drug seeking after extinction, preference for drugs over nondrug rewards, and resistance to punishment. The occurrence of addiction-like behavior in laboratory animals has opened the opportunity to investigate the neural, genetic, and psychological background of key aspects of addiction, which may ultimately contribute to the prevention and treatment of substance use disorders.
Collapse
Affiliation(s)
- Louk J M J Vanderschuren
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, the Netherlands
| | - Serge H Ahmed
- Université de Bordeaux, Bordeaux Neurocampus, Institut des Maladies Neurodégénératives, CNRS UMR 5293, F-33000 Bordeaux, France
| |
Collapse
|
17
|
Ozawa A, Arakawa H. Chemogenetics drives paradigm change in the investigation of behavioral circuits and neural mechanisms underlying drug action. Behav Brain Res 2021; 406:113234. [PMID: 33741409 PMCID: PMC8110310 DOI: 10.1016/j.bbr.2021.113234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
Recent developments in chemogenetic approaches to the investigation of brain function have ushered in a paradigm change in the strategy for drug and behavior research and clinical drug-based medications. As the nature of the drug action is based on humoral regulation, it is a challenge to identify the neuronal mechanisms responsible for the expression of certain targeted behavior induced by drug application. The development of chemogenetic approaches has allowed researchers to control neural activities in targeted neurons through a toolbox, including engineered G protein-coupled receptors or ligand-gated ion channels together with exogenously inert synthetic ligands. This review provides a brief overview of the chemogenetics toolbox with an emphasis on the DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) technique used in rodent models, which is applicable to the investigation of how specific neural circuits regulate behavioral processes. The use of chemogenetics has had a significant impact on basic neuroscience for a better understanding of the relationships between brain activity and the expression of behaviors with cell- and circuit-specific orders. Furthermore, chemogenetics is potentially a useful tool to deconstruct the neuropathological mechanisms of mental diseases and its regulation by drug, and provide us with transformative therapeutics with medication. We also review recent findings in the use of chemogenetic techniques to uncover functional circuit connections of serotonergic neurons in rodent models.
Collapse
Affiliation(s)
- Akihiko Ozawa
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, USA
| | - Hiroyuki Arakawa
- Department of Psychology, Tokiwa University, Mito, Ibaraki, Japan; Department of Systems Physiology, University of Ryukyus, Faculty of Medicine, Nakagami District, Okinawa, Japan.
| |
Collapse
|
18
|
Navarrete F, García-Gutiérrez MS, Gasparyan A, Austrich-Olivares A, Manzanares J. Role of Cannabidiol in the Therapeutic Intervention for Substance Use Disorders. Front Pharmacol 2021; 12:626010. [PMID: 34093179 PMCID: PMC8173061 DOI: 10.3389/fphar.2021.626010] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/04/2021] [Indexed: 01/04/2023] Open
Abstract
Drug treatments available for the management of substance use disorders (SUD) present multiple limitations in efficacy, lack of approved treatments or alarming relapse rates. These facts hamper the clinical outcome and the quality of life of the patients supporting the importance to develop new pharmacological agents. Lately, several reports suggest that cannabidiol (CBD) presents beneficial effects relevant for the management of neurological disorders such as epilepsy, multiple sclerosis, Parkinson's, or Alzheimer's diseases. Furthermore, there is a large body of evidence pointing out that CBD improves cognition, neurogenesis and presents anxiolytic, antidepressant, antipsychotic, and neuroprotective effects suggesting potential usefulness for the treatment of neuropsychiatric diseases and SUD. Here we review preclinical and clinical reports regarding the effects of CBD on the regulation of the reinforcing, motivational and withdrawal-related effects of different drugs of abuse such as alcohol, opioids (morphine, heroin), cannabinoids, nicotine, and psychostimulants (cocaine, amphetamine). Furthermore, a special section of the review is focused on the neurobiological mechanisms that might be underlying the 'anti-addictive' action of CBD through the regulation of dopaminergic, opioidergic, serotonergic, and endocannabinoid systems as well as hippocampal neurogenesis. The multimodal pharmacological profile described for CBD and the specific regulation of addictive behavior-related targets explains, at least in part, its therapeutic effects on the regulation of the reinforcing and motivational properties of different drugs of abuse. Moreover, the remarkable safety profile of CBD, its lack of reinforcing properties and the existence of approved medications containing this compound (Sativex®, Epidiolex®) increased the number of studies suggesting the potential of CBD as a therapeutic intervention for SUD. The rising number of publications with substantial results on the valuable therapeutic innovation of CBD for treating SUD, the undeniable need of new therapeutic agents to improve the clinical outcome of patients with SUD, and the upcoming clinical trials involving CBD endorse the relevance of this review.
Collapse
Affiliation(s)
- Francisco Navarrete
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - María Salud García-Gutiérrez
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | - Ani Gasparyan
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| | | | - Jorge Manzanares
- Instituto de Neurociencias, Universidad Miguel Hernández-CSIC, San Juan de Alicante, Spain
- Red Temática de Investigación Cooperativa en Salud (RETICS), Red de Trastornos Adictivos, Instituto de Salud Carlos III, MICINN and FEDER, Madrid, Spain
| |
Collapse
|
19
|
Delcourte S, Etievant A, Haddjeri N. Role of central serotonin and noradrenaline interactions in the antidepressants' action: Electrophysiological and neurochemical evidence. PROGRESS IN BRAIN RESEARCH 2021; 259:7-81. [PMID: 33541681 DOI: 10.1016/bs.pbr.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of antidepressant drugs, in the last 6 decades, has been associated with theories based on a deficiency of serotonin (5-HT) and/or noradrenaline (NA) systems. Although the pathophysiology of major depression (MD) is not fully understood, numerous investigations have suggested that treatments with various classes of antidepressant drugs may lead to an enhanced 5-HT and/or adapted NA neurotransmissions. In this review, particular morpho-physiological aspects of these systems are first considered. Second, principal features of central 5-HT/NA interactions are examined. In this regard, the effects of the acute and sustained antidepressant administrations on these systems are discussed. Finally, future directions including novel therapeutic strategies are proposed.
Collapse
Affiliation(s)
- Sarah Delcourte
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France
| | - Adeline Etievant
- Integrative and Clinical Neurosciences EA481, University of Bourgogne Franche-Comté, Besançon, France
| | - Nasser Haddjeri
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute U1208, Bron, France.
| |
Collapse
|
20
|
Siemann JK, Grueter BA, McMahon DG. Rhythms, Reward, and Blues: Consequences of Circadian Photoperiod on Affective and Reward Circuit Function. Neuroscience 2020; 457:220-234. [PMID: 33385488 DOI: 10.1016/j.neuroscience.2020.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/01/2023]
Abstract
Circadian disruptions, along with altered affective and reward states, are commonly associated with psychiatric disorders. In addition to genetics, the enduring influence of environmental factors in programming neural networks is of increased interest in assessing the underpinnings of mental health. The duration of daylight or photoperiod is known to impact both the serotonin and dopamine systems, which are implicated in mood and reward-based disorders. This review first examines the effects of circadian disruption and photoperiod in the serotonin system in both human and preclinical studies. We next highlight how brain regions crucial for the serotoninergic system (i.e., dorsal raphe nucleus; DRN), and dopaminergic (i.e., nucleus accumbens; NAc and ventral tegmental area; VTA) system are intertwined in overlapping circuitry, and play influential roles in the pathology of mood and reward-based disorders. We then focus on human and animal studies that demonstrate the impact of circadian factors on the dopaminergic system. Lastly, we discuss how environmental factors such as circadian photoperiod can impact the neural circuits that are responsible for regulating affective and reward states, offering novel insights into the biological mechanisms underlying the pathophysiology, systems, and therapeutic treatments necessary for mood and reward-based disorders.
Collapse
Affiliation(s)
- Justin K Siemann
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA; Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Brad A Grueter
- Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA; Department of Anesthesiology, Vanderbilt University, Nashville, TN 37235, USA; Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN 37235, USA; Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA
| | - Douglas G McMahon
- Department of Biological Sciences, Vanderbilt University, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University, Nashville, TN 37235, USA; Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, TN 37235, USA.
| |
Collapse
|
21
|
Galaj E, Xi ZX. Possible Receptor Mechanisms Underlying Cannabidiol Effects on Addictive-like Behaviors in Experimental Animals. Int J Mol Sci 2020; 22:ijms22010134. [PMID: 33374481 PMCID: PMC7795330 DOI: 10.3390/ijms22010134] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/19/2020] [Accepted: 12/20/2020] [Indexed: 12/24/2022] Open
Abstract
Substance use disorder (SUD) is a serious public health problem worldwide for which available treatments show limited effectiveness. Since the legalization of cannabis and the approval of cannabidiol (CBD) by the US Food and Drug Administration, therapeutic potential of CBD for the treatment of SUDs and other diseases has been widely explored. In this mini-review article, we first review the history and evidence supporting CBD as a potential pharmacotherapeutic. We then focus on recent progress in preclinical research regarding the pharmacological efficacy of CBD and the underlying receptor mechanisms on addictive-like behavior. Growing evidence indicates that CBD has therapeutic potential in reducing drug reward, as assessed in intravenous drug self-administration, conditioned place preference and intracranial brain-stimulation reward paradigms. In addition, CBD is effective in reducing relapse in experimental animals. Both in vivo and in vitro receptor mechanism studies indicate that CBD may act as a negative allosteric modulator of type 1 cannabinoid (CB1) receptor and an agonist of type 2 cannabinoid (CB2), transient receptor potential vanilloid 1 (TRPV1), and serotonin 5-HT1A receptors. Through these multiple-receptor mechanisms, CBD is believed to modulate brain dopamine in response to drugs of abuse, leading to attenuation of drug-taking and drug-seeking behavior. While these findings suggest that CBD is a promising therapeutic candidate, further investigation is required to verify its safety, pharmacological efficacy and the underlying receptor mechanisms in both experimental animals and humans.
Collapse
|
22
|
Chu J, Deyama S, Li X, Motono M, Otoda A, Saito A, Esaki H, Nishitani N, Kaneda K. Role of 5-HT 1A receptor-mediated serotonergic transmission in the medial prefrontal cortex in acute restraint stress-induced augmentation of rewarding memory of cocaine in mice. Neurosci Lett 2020; 743:135555. [PMID: 33352288 DOI: 10.1016/j.neulet.2020.135555] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/28/2020] [Accepted: 12/02/2020] [Indexed: 11/27/2022]
Abstract
Stress enhances cocaine craving. We recently reported that acute restraint stress increases cocaine conditioned place preference (CPP) in mice; however, the underlying mechanisms remain unclear. This study aimed to examine the role of serotonergic transmission in the medial prefrontal cortex (mPFC) in cocaine CPP enhancement by acute restraint stress, which increases extracellular serotonin (5-HT) levels in the mPFC. Intra-mPFC infusion of the selective serotonin reuptake inhibitor (S)-citalopram prior to the test session significantly increased the cocaine CPP score under non-stressed conditions. This is indicative of the substantial role of increased mPFC 5-HT levels in cocaine CPP enhancement. Moreover, intra-mPFC and systemic administration of the 5-HT1A receptor antagonist WAY100635 immediately before restraint stress exposure significantly attenuated stress-induced cocaine CPP enhancement. Our findings suggest that enhanced serotonergic transmission via 5-HT1A receptors in the mPFC is involved in acute stress-induced augmentation of rewarding memory of cocaine; moreover, the 5-HT1A receptor could be a therapeutic target for stress-induced cocaine craving.
Collapse
Affiliation(s)
- Jinling Chu
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Satoshi Deyama
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Xueting Li
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Mei Motono
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Atsuki Otoda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Atsushi Saito
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Hirohito Esaki
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Naoya Nishitani
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Katsuyuki Kaneda
- Laboratory of Molecular Pharmacology, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
23
|
Psychedelics as an emerging novel intervention in the treatment of substance use disorder: a review. Mol Biol Rep 2020; 47:9791-9799. [PMID: 33231817 DOI: 10.1007/s11033-020-06009-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022]
Abstract
Classical psychedelics are a group of drugs characterized by their activation of the serotonin-2A (5-hydroxytryptamine-2A; 5-HT2A) receptor and the unique hallucinogenic and mystical-type experiences that result. After a substantial period of restrictions limiting investigations into the therapeutic potential of psychedelics, a relatively recent recommencement of interest has sparked the burgeoning possibility for these drugs to play a part in the treatment of a wide array of psychopathologies. One of the most promising is in the study of addiction. Evidence has emerged that psychedelic agents may provide a novel avenue for the clinical treatment of patients dealing with substance use disorders (SUD). These serotonergic hallucinogens have displayed remarkable and enduring positive outcomes in this area, even when administered as one or two doses. The neural targets for these psychedelics are varied and underlie a complex mechanism of action-modulating multiple neural networks. It is believed that these agents allow for the reorganization of disordered neural pathways in the default mode network and attenuate maladaptive signaling in mesolimbic reward circuitry. The aim of this review is to examine the current standing of evidence regarding psychedelic psychopharmacology and to provide an overview of the use and effectiveness of these drugs in the treatment of SUD, alcohol use disorder, and for smoking cessation.
Collapse
|
24
|
Baskin BM, Mai JJ, Dymecki SM, Kantak KM. Cocaine reward and memory after chemogenetic inhibition of distinct serotonin neuron subtypes in mice. Psychopharmacology (Berl) 2020; 237:2633-2648. [PMID: 32494973 PMCID: PMC8057056 DOI: 10.1007/s00213-020-05560-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/18/2020] [Indexed: 01/23/2023]
Abstract
RATIONALE We probed serotonin neurons, those denoted by their developmental gene expression as r2Hoxa2-Pet1 (experiment 1) and Drd1a-Pet1 (experiment 2), for differential modulation of cocaine reward and memory as revealed by the expression and development of conditioned place preference (CPP) in transgenic mice. OBJECTIVES To query roles in CPP, we inhibited neurons cell autonomously in vivo by activating the transgenically expressed, synthetic DREADD receptor hM4Di (Di) with the exogenous ligand clozapine-N-oxide (CNO). METHODS To examine CPP expression, mice were conditioned using behaviorally active doses of cocaine (10.0 or 17.8 mg/kg) vs. saline followed by CPP assessment, first without neuron inhibition (post-conditioning session 1), and then with CNO-mediated neuron inhibition (post-conditioning session 2), followed by 4 more post-conditioning sessions. To examine CPP development, we administered CNO during conditioning sessions and then assayed CPP across 6 post-conditioning sessions. RESULTS In r2Hoxa2-Pet1-Di mice, post-conditioning CNO administration did not impact cocaine CPP expression, but after CNO administration during conditioning, cocaine CPP (17.8 mg/kg) persisted across post-conditioning sessions compared with that in controls, suggesting a deficit in extinguishing cocaine memory. Drd1a-Pet1-Di mice, prior to CNO-Di-triggered neuronal inhibition, unexpectedly expressed heightened cocaine CPP (10.0 and 17.8 mg/kg) compared with controls, and this basal phenotype was transiently blocked by acute post-conditioning CNO administration and persistently blocked by repeated CNO administration during conditioning. CONCLUSION Cocaine reward and memory likely map to distinct serotonergic Pet1 neuron subtypes. r2Hoxa2-Pet1 neurons normally may limit the durability of cocaine memory, without impacting initial cocaine reward magnitude. Drd1a-Pet1 neurons normally may help to promote cocaine reward.
Collapse
Affiliation(s)
- Britahny M. Baskin
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| | - Jia Jia Mai
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Susan M. Dymecki
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Kathleen M. Kantak
- Department of Psychological and Brain Sciences, Boston University, 64 Cummington Mall, Boston, MA 02215, USA
| |
Collapse
|
25
|
Sizemore TR, Hurley LM, Dacks AM. Serotonergic modulation across sensory modalities. J Neurophysiol 2020; 123:2406-2425. [PMID: 32401124 PMCID: PMC7311732 DOI: 10.1152/jn.00034.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/04/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
The serotonergic system has been widely studied across animal taxa and different functional networks. This modulatory system is therefore well positioned to compare the consequences of neuromodulation for sensory processing across species and modalities at multiple levels of sensory organization. Serotonergic neurons that innervate sensory networks often bidirectionally exchange information with these networks but also receive input representative of motor events or motivational state. This convergence of information supports serotonin's capacity for contextualizing sensory information according to the animal's physiological state and external events. At the level of sensory circuitry, serotonin can have variable effects due to differential projections across specific sensory subregions, as well as differential serotonin receptor type expression within those subregions. Functionally, this infrastructure may gate or filter sensory inputs to emphasize specific stimulus features or select among different streams of information. The near-ubiquitous presence of serotonin and other neuromodulators within sensory regions, coupled with their strong effects on stimulus representation, suggests that these signaling pathways should be considered integral components of sensory systems.
Collapse
Affiliation(s)
- Tyler R Sizemore
- Department of Biology, West Virginia University, Morgantown, West Virginia
| | - Laura M Hurley
- Department of Biology, Indiana University, Bloomington, Indiana
| | - Andrew M Dacks
- Department of Biology, West Virginia University, Morgantown, West Virginia
- Department of Neuroscience, West Virginia University, Morgantown, West Virginia
| |
Collapse
|
26
|
Galaj E, Bi GH, Yang HJ, Xi ZX. Cannabidiol attenuates the rewarding effects of cocaine in rats by CB2, 5-HT 1A and TRPV1 receptor mechanisms. Neuropharmacology 2020; 167:107740. [PMID: 31437433 PMCID: PMC7493134 DOI: 10.1016/j.neuropharm.2019.107740] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/16/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022]
Abstract
Cocaine abuse continues to be a serious health problem worldwide. Despite intense research there is currently no FDA-approved medication to treat cocaine use disorder. The recent search has been focused on agents targeting primarily the dopamine system, while limited success has been achieved at the clinical level. Cannabidiol (CBD) is a U.S. FDA-approved cannabinoid for the treatment of epilepsy and recently was reported to have therapeutic potential for other disorders. Here we systemically evaluated its potential utility for the treatment of cocaine use disorder and explored the underlying receptor mechanisms in experimental animals. Systemic administration (10-40 mg/kg) of CBD dose-dependently inhibited cocaine self-administration, shifted a cocaine dose-response curve downward, and lowered break-points for cocaine self-administration under a progressive-ratio schedule of reinforcement. CBD inhibited cocaine self-administration maintained by low, but not high, doses of cocaine. In addition, CBD (3-20 mg/kg) dose-dependently attenuated cocaine-enhanced brain-stimulation reward (BSR) in rats. Strikingly, this reduction in both cocaine self-administration and BSR was blocked by AM630 (a cannabinoid CB2 receptor antagonist), WAY100135 (a 5-HT1A receptor antagonist), or capsazepine (a TRPV1 channel blocker), but not by AM251 (a CB1 receptor antagonist), CID16020046 (a GPR55 antagonist), or naloxone (an opioid receptor antagonist), suggesting the involvement of CB2, 5-HT1A, and TRPV1 receptors in CBD action. In vivo microdialysis indicated that pretreatment with CBD (10-20 mg/kg) attenuated cocaine-induced increases in extracellular dopamine (DA) in the nucleus accumbens, while CBD alone failed to alter extracellular DA. These findings suggest that CBD may have certain therapeutic utility by blunting the acute rewarding effects of cocaine via a DA-dependent mechanism.
Collapse
Affiliation(s)
- Ewa Galaj
- Addiction Biology Unit, Molecular Targets and Medication Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Guo-Hua Bi
- Addiction Biology Unit, Molecular Targets and Medication Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Hong-Ju Yang
- Addiction Biology Unit, Molecular Targets and Medication Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA
| | - Zheng-Xiong Xi
- Addiction Biology Unit, Molecular Targets and Medication Discovery Branch, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD, 21224, USA.
| |
Collapse
|
27
|
Karel P, Van der Toorn A, Vanderschuren L, Guo C, Sadighi Alvandi M, Reneman L, Dijkhuizen R, Verheij MMM, Homberg JR. Ultrahigh-resolution MRI reveals structural brain differences in serotonin transporter knockout rats after sucrose and cocaine self-administration. Addict Biol 2020; 25:e12722. [PMID: 30748070 PMCID: PMC6916608 DOI: 10.1111/adb.12722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 11/21/2018] [Accepted: 01/08/2019] [Indexed: 12/16/2022]
Abstract
Excessive use of cocaine is known to induce changes in brain white and gray matter. It is unknown whether the extent of these changes is related to individual differences in vulnerability to cocaine addiction. One factor increasing vulnerability involves reduced expression of the serotonin transporter (5-HTT). Human studies have shown that inherited 5-HTT downregulation is associated with structural changes in the brain. These genotype-related structural changes may contribute to risk for cocaine addiction. Here, we tested this idea by using ultrahigh-resolution structural magnetic resonance imaging (MRI) on postmortem tissue of 5-HTT-/- and wild-type (5-HTT+/+ ) rats with a history of long access to cocaine or sucrose (control) self-administration. We found that 5-HTT-/- rats, compared with wild-type control animals, self-administered more cocaine, but not sucrose, under long-access conditions. Ultrahigh-resolution structural MRI subsequently revealed that, independent of sucrose or cocaine self-administration, 5-HTT-/- rats had a smaller amygdala. Moreover, we found an interaction between genotype and type of reward for dorsal raphe nucleus volume. The data point to an important but differential role of the amygdala and dorsal raphe nucleus in 5-HTT genotype-dependent vulnerability to cocaine addiction.
Collapse
Affiliation(s)
- Peter Karel
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| | - Annette Van der Toorn
- Biomedical MR Imaging and Spectroscopy Group, Center for Image SciencesUniversity Medical Center Utrecht and Utrecht UniversityUtrechtThe Netherlands
| | - Louk Vanderschuren
- Department of Animals in Science and Society, Division of Behavioural Neuroscience, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Chao Guo
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| | - Mina Sadighi Alvandi
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| | - Liesbeth Reneman
- Department of Radiology and Nuclear Medicine, Academic Medical CenterUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Brain and CognitionUniversity of AmsterdamAmsterdamThe Netherlands
| | - Rick Dijkhuizen
- Biomedical MR Imaging and Spectroscopy Group, Center for Image SciencesUniversity Medical Center Utrecht and Utrecht UniversityUtrechtThe Netherlands
| | - Michel M. M. Verheij
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| | - Judith R. Homberg
- Department of Cognitive Neuroscience, Centre for Neuroscience, Donders Institute for Brain, Cognition and BehaviourRadboudumcNijmegenThe Netherlands
| |
Collapse
|
28
|
5-HT1A autoreceptor in dorsal raphe nucleus mediates sensitization of conditioned place preference to cocaine in mice experienced with chronic pain. Neuroreport 2019; 30:681-687. [DOI: 10.1097/wnr.0000000000001260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Muir J, Lopez J, Bagot RC. Wiring the depressed brain: optogenetic and chemogenetic circuit interrogation in animal models of depression. Neuropsychopharmacology 2019; 44:1013-1026. [PMID: 30555161 PMCID: PMC6461994 DOI: 10.1038/s41386-018-0291-6] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/26/2018] [Indexed: 12/19/2022]
Abstract
The advent of optogenetics and chemogenetics has revolutionized the study of neural circuit mechanisms of behavioral dysregulation in psychiatric disease. These powerful technologies allow manipulation of specific neurons to determine causal relationships between neuronal activity and behavior. Optogenetic tools have been key to mapping the circuitry underlying depression-like behavior in animal models, clarifying the contribution of the ventral tegmental area, nucleus accumbens, medial prefrontal cortex, ventral hippocampus, and other limbic areas, to stress susceptibility. In comparison, chemogenetics have been relatively underutilized, despite offering unique advantages for probing long-term effects of manipulating neuronal activity. The ongoing development of optogenetic tools to probe in vivo function of ever-more specific circuits, combined with greater integration of chemogenetic tools and recent advances in vivo imaging techniques will continue to advance our understanding of the circuit mechanisms of depression.
Collapse
Affiliation(s)
- Jessie Muir
- 0000 0004 1936 8649grid.14709.3bIntegrated Program in Neuroscience, McGill University, Montréal, QC Canada
| | - Joëlle Lopez
- 0000 0004 1936 8649grid.14709.3bDepartment of Psychology, McGill University, Montréal, QC Canada
| | - Rosemary C. Bagot
- 0000 0004 1936 8649grid.14709.3bDepartment of Psychology, McGill University, Montréal, QC Canada ,Ludmer Center for Neuroinformatics and Mental Health, Montréal, QC Canada
| |
Collapse
|
30
|
Klawonn AM, Malenka RC. Nucleus Accumbens Modulation in Reward and Aversion. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2019; 83:119-129. [PMID: 30674650 PMCID: PMC6650377 DOI: 10.1101/sqb.2018.83.037457] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The nucleus accumbens (NAc) is a key node of the brain’s circuitry that is responsible for translating motivation into action. It has been implicated in playing critical roles in virtually all forms of adaptive and pathological motivated behaviors. It is subject to modulation by a broad array of inputs that influence NAc activity in complex ways that are still poorly understood. Here, we briefly review current knowledge about the behavioral consequences of NAc modulation, focusing on recent studies that use novel techniques developed and implemented over the last decade.
Collapse
Affiliation(s)
- Anna M Klawonn
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Robert C Malenka
- Department of Psychiatry and Behavioral Sciences, Nancy Pritzker Laboratory, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
31
|
Haleem DJ, Nawaz S, Salman T. Dopamine and serotonin metabolism associated with morphine reward and its inhibition with buspirone: A study in the rat striatum. Pharmacol Biochem Behav 2018; 170:71-78. [PMID: 29782941 DOI: 10.1016/j.pbb.2018.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 05/17/2018] [Accepted: 05/17/2018] [Indexed: 02/06/2023]
Abstract
Adaptations within the nucleus accumbens (NAc) and caudate nucleus (CN) dopamine neurotransmission are involved in behavioral sensitization and enhanced incentive motivation towards drug paired stimuli which lead to drug addiction. Serotonin (5-hydroxytryptamine; 5-HT) can modulate dopamine neurotransmission to reduce rewarding effects of drugs of abuse. A recent study from our laboratory shows that rewarding effects of morphine are inhibited in rats co-treated with buspirone. To understand the neurochemical mechanism involved in morphine addiction and its inhibition with buspirone, present study determines the effects of buspirone, morphine and their co-administration on the metabolism of serotonin and dopamine in the NAc and CN. We find that rewarding effects of morphine are associated with an enhancement and attenuation of dopamine metabolism, respectively in the CN and NAc. Serotonin metabolism is enhanced in both regions. Co-administration of buspirone not only prevents rewarding effects of morphine, but its effects on the metabolism of dopamine and serotonin in the NAc and CN are also reversed. Results suggest that 5-HT1A receptor dependent modulation of dopamine neurotransmission in the CN and NAc is involved in the modulation of the rewarding effects of morphine in buspirone co-treated animals. The findings documenting an important role of 5-HT1A receptors in drug addiction suggest that synthetic opioid drugs with agonist activity of 5-HT1A receptors may prove non addictive analgesics.
Collapse
Affiliation(s)
- Darakhshan Jabeen Haleem
- Neuroscience Research Laboratory, Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science (ICCBS), University of Karachi, Karachi 75270, Pakistan.
| | - Shazia Nawaz
- Neuroscience Research Laboratory, Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science (ICCBS), University of Karachi, Karachi 75270, Pakistan
| | - Tabinda Salman
- Neuroscience Research Laboratory, Dr Panjwani Center for Molecular Medicine & Drug Research (PCMD), International Center for Chemical and Biological Science (ICCBS), University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
32
|
Resolving Behavioral Output via Chemogenetic Designer Receptors Exclusively Activated by Designer Drugs. J Neurosci 2017; 36:9268-82. [PMID: 27605603 DOI: 10.1523/jneurosci.1333-16.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 07/13/2016] [Indexed: 12/26/2022] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) have proven to be highly effective neuromodulatory tools for the investigation of neural circuits underlying behavioral outputs. They exhibit a number of advantages: they rely on cell-specific manipulations through canonical intracellular signaling pathways, they are easy and cost-effective to implement in a laboratory setting, and they are easily scalable for single-region or full-brain manipulations. On the other hand, DREADDs rely on ligand-G-protein-coupled receptor interactions, leading to coarse temporal dynamics. In this review we will provide a brief overview of DREADDs, their implementation, and the advantages and disadvantages of their use in animal systems. We also will provide numerous examples of their use across a broad variety of biomedical research fields.
Collapse
|
33
|
Simmler LD, Anacker AMJ, Levin MH, Vaswani NM, Gresch PJ, Nackenoff AG, Anastasio NC, Stutz SJ, Cunningham KA, Wang J, Zhang B, Henry LK, Stewart A, Veenstra‐VanderWeele J, Blakely RD. Blockade of the 5-HT transporter contributes to the behavioural, neuronal and molecular effects of cocaine. Br J Pharmacol 2017; 174:2716-2738. [PMID: 28585320 PMCID: PMC5522997 DOI: 10.1111/bph.13899] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/05/2017] [Accepted: 05/26/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE The psychostimulant cocaine induces complex molecular, cellular and behavioural responses as a consequence of inhibiting presynaptic dopamine, noradrenaline and 5-HT transporters. To elucidate 5-HT transporter (SERT)-specific contributions to cocaine action, we evaluated cocaine effects in the SERT Met172 knock-in mouse, which expresses a SERT coding substitution that eliminates high-affinity cocaine recognition. EXPERIMENTAL APPROACH We measured the effects of SERT Met172 on cocaine antagonism of 5-HT re-uptake using ex vivo synaptosome preparations and in vivo microdialysis. We assessed SERT dependence of cocaine actions behaviourally through acute and chronic locomotor activation, sensitization, conditioned place preference (CPP) and oral cocaine consumption. We used c-Fos, quantitative RT-PCR and RNA sequencing methods for insights into cellular and molecular networks supporting SERT-dependent cocaine actions. KEY RESULTS SERT Met172 mice demonstrated functional insensitivity for cocaine at SERT. Although they displayed wild-type levels of acute cocaine-induced hyperactivity or chronic sensitization, the pattern of acute motor activation was different, with a bias toward thigmotaxis. CPP was increased, and a time-dependent elevation in oral cocaine consumption was observed. SERT Met172 mice displayed relatively higher levels of neuronal activation in the hippocampus, piriform cortex and prelimbic cortex (PrL), accompanied by region-dependent changes in immediate early gene expression. Distinct SERT-dependent gene expression networks triggered by acute and chronic cocaine administration were identified, including PrL Akt and nucleus accumbens ERK1/2 signalling. CONCLUSION AND IMPLICATIONS Our studies reveal distinct SERT contributions to cocaine action, reinforcing the possibility of targeting specific aspects of cocaine addiction by modulation of 5-HT signalling.
Collapse
Affiliation(s)
- Linda D Simmler
- Department of PharmacologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Allison M J Anacker
- Department of PsychiatryColumbia University; New York State Psychiatric InstituteNew YorkNYUSA
- Sackler Institute for Developmental PsychobiologyColumbia University; New York State Psychiatric InstituteNew YorkNYUSA
| | - Michael H Levin
- Department of PharmacologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Nina M Vaswani
- Department of PharmacologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Paul J Gresch
- Department of PharmacologyVanderbilt University School of MedicineNashvilleTNUSA
- Department of Biomedical ScienceCharles E. Schmidt College of Medicine and Brain InstituteJupiterFLUSA
| | - Alex G Nackenoff
- Department of PharmacologyVanderbilt University School of MedicineNashvilleTNUSA
| | - Noelle C Anastasio
- Center for Addiction ResearchUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Pharmacology and ToxicologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Sonja J Stutz
- Center for Addiction ResearchUniversity of Texas Medical BranchGalvestonTXUSA
| | - Kathryn A Cunningham
- Center for Addiction ResearchUniversity of Texas Medical BranchGalvestonTXUSA
- Department of Pharmacology and ToxicologyUniversity of Texas Medical BranchGalvestonTXUSA
| | - Jing Wang
- Department of Biomedical InformaticsVanderbilt University School of MedicineNashvilleTNUSA
- Lester and Sue Smith Breast CenterBaylor College of MedicineHoustonTXUSA
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - Bing Zhang
- Department of Biomedical InformaticsVanderbilt University School of MedicineNashvilleTNUSA
- Lester and Sue Smith Breast CenterBaylor College of MedicineHoustonTXUSA
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTXUSA
| | - L Keith Henry
- Department of Biomedical SciencesUniversity of North Dakota School of Medicine and Health SciencesGrand ForksNDUSA
| | - Adele Stewart
- Department of PharmacologyVanderbilt University School of MedicineNashvilleTNUSA
- Department of Biomedical ScienceCharles E. Schmidt College of Medicine and Brain InstituteJupiterFLUSA
| | - Jeremy Veenstra‐VanderWeele
- Department of PsychiatryColumbia University; New York State Psychiatric InstituteNew YorkNYUSA
- Sackler Institute for Developmental PsychobiologyColumbia University; New York State Psychiatric InstituteNew YorkNYUSA
| | - Randy D Blakely
- Department of PharmacologyVanderbilt University School of MedicineNashvilleTNUSA
- Department of PsychiatryVanderbilt University School of MedicineNashvilleTNUSA
- Department of Biomedical ScienceCharles E. Schmidt College of Medicine and Brain InstituteJupiterFLUSA
| |
Collapse
|
34
|
Dobrzanski G, Kossut M. Application of the DREADD technique in biomedical brain research. Pharmacol Rep 2017; 69:213-221. [DOI: 10.1016/j.pharep.2016.10.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/21/2016] [Accepted: 10/24/2016] [Indexed: 12/25/2022]
|
35
|
Reilly MT, Noronha A, Goldman D, Koob GF. Genetic studies of alcohol dependence in the context of the addiction cycle. Neuropharmacology 2017; 122:3-21. [PMID: 28118990 DOI: 10.1016/j.neuropharm.2017.01.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/13/2017] [Accepted: 01/19/2017] [Indexed: 12/16/2022]
Abstract
Family, twin and adoption studies demonstrate clearly that alcohol dependence and alcohol use disorders are phenotypically complex and heritable. The heritability of alcohol use disorders is estimated at approximately 50-60% of the total phenotypic variability. Vulnerability to alcohol use disorders can be due to multiple genetic or environmental factors or their interaction which gives rise to extensive and daunting heterogeneity. This heterogeneity makes it a significant challenge in mapping and identifying the specific genes that influence alcohol use disorders. Genetic linkage and (candidate gene) association studies have been used now for decades to map and characterize genomic loci and genes that underlie the genetic vulnerability to alcohol use disorders. These approaches have been moderately successful in identifying several genes that contribute to the complexity of alcohol use disorders. Recently, genome-wide association studies have become one of the major tools for identifying genes for alcohol use disorders by examining correlations between millions of common single-nucleotide polymorphisms with diagnosis status. Genome-wide association studies are just beginning to uncover novel biology; however, the functional significance of results remains a matter of extensive debate and uncertainty. In this review, we present a select group of genome-wide association studies of alcohol dependence, as one example of a way to generate functional hypotheses, within the addiction cycle framework. This analysis may provide novel directions for validating the functional significance of alcohol dependence candidate genes. This article is part of the Special Issue entitled "Alcoholism".
Collapse
Affiliation(s)
- Matthew T Reilly
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Division of Neuroscience and Behavior, 5635 Fishers Lane, Bethesda, MD 20852, USA.
| | - Antonio Noronha
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Division of Neuroscience and Behavior, 5635 Fishers Lane, Bethesda, MD 20852, USA
| | - David Goldman
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Chief, Laboratory of Neurogenetics, 5635 Fishers Lane, Bethesda, MD 20852, USA
| | - George F Koob
- National Institutes of Health (NIH), National Institute on Alcohol Abuse and Alcoholism (NIAAA), Director NIAAA, 5635 Fishers Lane, Bethesda, MD 20852, USA
| |
Collapse
|
36
|
Abstract
Classic hallucinogens share pharmacology as serotonin 5-HT2A, 5-HT2B, and 5-HT2C receptor agonists. Unique among most other Schedule 1 drugs, they are generally non-addictive and can be effective tools in the treatment of addiction. Mechanisms underlying these attributes are largely unknown. However, many preclinical studies show that 5-HT2C agonists counteract the addictive effects of drugs from several classes, suggesting this pharmacological property of classic hallucinogens may be significant. Drawing from a comprehensive analysis of preclinical behavior, neuroanatomy, and neurochemistry studies, this review builds rationale for this hypothesis, and also proposes a testable, neurobiological framework. 5-HT2C agonists work, in part, by modulating dopamine neuron activity in the ventral tegmental area-nucleus accumbens (NAc) reward pathway. We argue that activation of 5-HT2C receptors on NAc shell, GABAergic, medium spiny neurons inhibits potassium Kv1.x channels, thereby enhancing inhibitory activity via intrinsic mechanisms. Together with experiments that show that addictive drugs, such as cocaine, potentiate Kv1.x channels, thereby suppressing NAc shell GABAergic activity, this hypothesis provides a mechanism by which classic hallucinogen-mediated stimulation of 5-HT2C receptors could thwart addiction. It also provides a potential reason for the non-addictive nature of classic hallucinogens.
Collapse
Affiliation(s)
- Clinton E Canal
- Center for Drug Discovery, Department of Pharmaceutical Sciences, Northeastern University, Boston, USA
| | - Kevin S Murnane
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Mercer University Health Sciences Center, Atlanta, USA
| |
Collapse
|
37
|
de Veen BTH, Schellekens AFA, Verheij MMM, Homberg JR. Psilocybin for treating substance use disorders? Expert Rev Neurother 2016; 17:203-212. [PMID: 27684102 DOI: 10.1080/14737175.2016.1220834] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Evidence based treatment for Substance use disorders (SUD) includes psychotherapy and pharmacotherapy. However, these are only partially effective. Hallucinogens, such as psilocybin, may represent potential new treatment options for SUD. This review provides a summary of (human) studies on the putative therapeutic effects of psilocybin, and discusses the receptor systems, brain regions and cognitive and emotional processes mediating psilocybin's effects. Psilocybin's chemical structure is similar to that of serotonin. Dysregulations in the serotonin system are associated with alterations in stress hormones, such as cortisol, and mood disorders. After psilocybin administration cortisol levels spike and activate the executive control network, with subsequent increased control over emotional processes, and relief of negative thinking and persistent negative emotions. Preliminary data of ongoing alcohol and smoking addiction studies in humans shows promising effects of psilocybin administration on substance use. Importantly, psilocybin has a low risk of toxicity and dependence and can be used safely under controlled clinical conditions. Areas covered: This paper is a narrative review based on the search terms: psilocybin, substance use disorder, addiction, depression, serotonin. Literature on potential efficacy and mechanisms of action of psilocybin in SUD is discussed. Expert commentary: Recent positive findings with psilocybin need confirmation in well-designed placebo controlled randomized trials employing a large sample size.
Collapse
Affiliation(s)
- Bas T H de Veen
- a Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Cognitive Neuroscience , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Arnt F A Schellekens
- b Department of Psychiatry , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Michel M M Verheij
- a Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Cognitive Neuroscience , Radboud University Medical Center , Nijmegen , The Netherlands
| | - Judith R Homberg
- a Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, Department of Cognitive Neuroscience , Radboud University Medical Center , Nijmegen , The Netherlands
| |
Collapse
|