1
|
Butelman ER, Huang Y, King SG, Gaudreault PO, Ceceli AO, Kronberg G, Cathomas F, Roussos P, Russo SJ, Garland EL, Goldstein RZ, Alia-Klein N. Peripheral Blood Cytokines as Markers of Longitudinal Change in White Matter Microstructure Following Inpatient Treatment for Opioid Use Disorders. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100480. [PMID: 40248275 PMCID: PMC12005283 DOI: 10.1016/j.bpsgos.2025.100480] [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: 12/13/2024] [Revised: 01/24/2025] [Accepted: 02/20/2025] [Indexed: 04/19/2025] Open
Abstract
Background Opioid use disorder (OUD) causes major public health morbidity and mortality. Although standard-of-care treatment with medications for OUD (MOUDs) is available, there are few biological markers of the clinical process of recovery. Neurobiological aspects of recovery can include normalization of brain white matter (WM) microstructure, which is sensitive to cytokine signaling. Here, we determined whether blood-based cytokines can be markers of change in WM microstructure following MOUD. Methods Inpatient individuals with heroin use disorder (iHUDs) (n = 21) with methadone or buprenorphine MOUD underwent magnetic resonance imaging (MRI) scans with diffusion tensor imaging (DTI) and provided ratings of drug cue-induced craving, arousal, and valence earlier in treatment (MRI1) and ≈14 weeks thereafter (MRI2). Healthy control participants (HCs) (n = 24) also underwent 2 MRI scans during a similar time interval. At MRI2, participants provided a peripheral blood sample for multiplex quantification of serum cytokines. We analyzed the correlation of a multitarget biomarker score (from a principal component analysis of 19 cytokines that differed significantly between iHUDs and HCs) with treatment-related change in DTI metrics (ΔDTI; MRI2 - MRI1). Results The cytokine biomarker score was negatively correlated with ΔDTI metrics in frontal, frontoparietal, and corticolimbic WM tracts in iHUDs but not in HCs. Also, serum levels of specific cytokines in the cytokine biomarker score, including the interleukin-related oncostatin M (OSM), similarly correlated with ΔDTI metrics in iHUDs but not in HCs. Serum levels of other specific cytokines were negatively correlated with changes in cue-induced craving and arousal in the iHUDs. Conclusions Specific serum cytokines, studied alone or as a group, may serve as accessible biomarkers of WM microstructure changes and potential recovery in iHUDs undergoing treatment with MOUD.
Collapse
Affiliation(s)
- Eduardo R. Butelman
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Yuefeng Huang
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Sarah G. King
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pierre-Olivier Gaudreault
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ahmet O. Ceceli
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Greg Kronberg
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Flurin Cathomas
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Genetics and Genomic Science, Icahn School of Medicine at Mount Sinai, New York, New York
- Mental Illness Research, Education, and Clinical Center (VISN 2 South), James J. Peters Veterans Affairs, Medical Center, Bronx, New York
- Center for Precision Medicine and Translational Therapeutics, James J. Peters Veterans Affairs, Medical Center, Bronx, New York
| | - Scott J. Russo
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai, New York, New York
- Center of Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric L. Garland
- Department of Psychiatry, University of California San Diego, La Jolla, California
- Sanford Institute for Empathy and Compassion, University of California San Diego, La Jolla, California
| | - Rita Z. Goldstein
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Center of Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nelly Alia-Klein
- Neuropsychoimaging of Addictions and Related Conditions Research Program, Icahn School of Medicine at Mount Sinai, Departments of Psychiatry and Neuroscience, New York, New York
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
- Center of Affective Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
2
|
Zhang Q, Wang R, Zhang L, Li M, Lin J, Lu X, Tian Y, Lin Y, Liu T, Chen Y, Li Y, Cao J, Wu Q, Wang J, Lu Z, Hong Z. A humanized Gs-coupled DREADD for circuit and behavior modulation. Front Cell Neurosci 2025; 19:1577117. [PMID: 40271540 PMCID: PMC12015759 DOI: 10.3389/fncel.2025.1577117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Designer receptors exclusively activated by designer drugs (DREADDs) play important roles in neuroscience research and show great promise for future clinical interventions in neurological diseases. The Gs-coupled DREADD, rM3Ds, modulates excitability in neuron subsets that are sensitive to downstream effectors of Gs protein. However, given the non-human nature of the rM3Ds backbone, risks about potential immunogenicity and tolerability exist when considering clinical translation. Here, we report the development of a whole sequence-humanized Gs-coupled DREADD, hM3Ds. We found that hM3Ds has a comparable DREADD ligand response profile to rM3Ds. We then selectively expressed hM3Ds in D1 medium spiny neurons (D1-MSNs) and found that hM3Ds was able to activate the D1-MSNs-mediated basal ganglia direct pathway and alleviate Parkinsonian phenotypes in a Parkinson's disease mouse model. In conclusion, this engineered humanized Gs-coupled DREADD is suitable as an effective, and likely safer, DREADD tool for both research and future clinical applications.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ruiqi Wang
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Liang Zhang
- Department of Anesthesiology, The Third People’s Hospital of Shenzhen, Shenzhen, China
| | - Mengqi Li
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Jianbang Lin
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoyang Lu
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yixuan Tian
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yunping Lin
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Taian Liu
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yefei Chen
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yuantao Li
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Jun Cao
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Qiang Wu
- Department of Anesthesiology, The Third People’s Hospital of Shenzhen, Shenzhen, China
- State Key Laboratory of Biomedical Imaging Science and System, Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Jinhui Wang
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, China
| | - Zhonghua Lu
- Research Center for Primate Neuromodulation and Neuroimaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Technological Research Center for Primate Translational Medicine, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Biomedical Imaging Science and System, Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Zexuan Hong
- Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
3
|
Zhu R, Wang D, Fan F, Fu F, Wei D, Tang S, Tian Y, Chen J, Li Y, Zhou H, Wang L, Zhang X. Differences in the prevalence and clinical correlates of depressive symptoms in male patients with methamphetamine and heroin use disorder in a Chinese Han population. Am J Addict 2024; 33:48-57. [PMID: 37644677 DOI: 10.1111/ajad.13476] [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: 04/07/2023] [Revised: 08/02/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Comorbidity of substance use disorders and depression is a common phenomenon. It is well known that opioid addiction is neurobiologically distinct from psychostimulant addiction. However, direct comparisons of comorbid depressive symptoms in patients with methamphetamine (METH) use disorder (MAUD) and heroin use disorders (HUD) have been lacking until now. METHODS A total of 353 patients with methamphetamine use disorder, 76 patients with HUD, and 203 healthy controls were recruited. The Beck Depression Inventory (BDI-SF), the Desires for Drug Questionnaire (DDQ) and the short form of the Childhood Trauma Questionnaire (CTQ-SF) were used to measure participants' depressive symptoms, drug craving, and childhood abuse or neglect, respectively. RESULTS The prevalence of depressive symptoms was 35.41% (125/353) in MAUD and 56.57% (43/76) in MAUD, significantly higher than the 22.66% (46/203) in healthy controls. Furthermore, there was a significant difference in the total BDI score between the MAUD and HUD groups (F = 5.02, df = 1, 372, p = .026). Among MAUD, years of education, history of incarceration, month of abstinence and negative reinforcement scores were associated with depressive symptoms (all p < .05). Among HUD, duration of drug use, childhood emotional abuse and sexual abuse were associated with depressive symptoms (all p < .05). CONCLUSIONS AND SCIENTIFIC SIGNIFICANCE The prevalence and correlates of depressive symptoms differ between MAUD and HUD, adding to the existing literature. Therefore, treatment and intervention programs should be designed to address these unique correlates in HUD and MAUD patients.
Collapse
Affiliation(s)
- Rongrong Zhu
- CAS Key Laboratory of Mental Health, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Dongmei Wang
- CAS Key Laboratory of Mental Health, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Fusheng Fan
- Psychological Health Center, Xin Hua Drug Rehabilitation Center, Sichuan, China
| | - Fabing Fu
- Psychological Health Center, Xin Hua Drug Rehabilitation Center, Sichuan, China
| | - Dejun Wei
- Psychological Health Center, Xin Hua Drug Rehabilitation Center, Sichuan, China
| | - Shanshan Tang
- Psychological Health Center, Xin Hua Drug Rehabilitation Center, Sichuan, China
| | - Yang Tian
- CAS Key Laboratory of Mental Health, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jiajing Chen
- CAS Key Laboratory of Mental Health, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Yuqing Li
- CAS Key Laboratory of Mental Health, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Huixia Zhou
- CAS Key Laboratory of Mental Health, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Li Wang
- CAS Key Laboratory of Mental Health, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Xiangyang Zhang
- CAS Key Laboratory of Mental Health, Chinese Academy of Sciences, Beijing, China
- Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
4
|
Adzic M, Lukic I, Mitic M, Glavonic E, Dragicevic N, Ivkovic S. Contribution of the opioid system to depression and to the therapeutic effects of classical antidepressants and ketamine. Life Sci 2023:121803. [PMID: 37245840 DOI: 10.1016/j.lfs.2023.121803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 05/22/2023] [Accepted: 05/22/2023] [Indexed: 05/30/2023]
Abstract
Major depressive disorder (MDD) afflicts approximately 5 % of the world population, and about 30-50 % of patients who receive classical antidepressant medications do not achieve complete remission (treatment resistant depressive patients). Emerging evidence suggests that targeting opioid receptors mu (MOP), kappa (KOP), delta (DOP), and the nociceptin/orphanin FQ receptor (NOP) may yield effective therapeutics for stress-related psychiatric disorders. As depression and pain exhibit significant overlap in their clinical manifestations and molecular mechanisms involved, it is not a surprise that opioids, historically used to alleviate pain, emerged as promising and effective therapeutic options in the treatment of depression. The opioid signaling is dysregulated in depression and numerous preclinical studies and clinical trials strongly suggest that opioid modulation can serve as either an adjuvant or even an alternative to classical monoaminergic antidepressants. Importantly, some classical antidepressants require the opioid receptor modulation to exert their antidepressant effects. Finally, ketamine, a well-known anesthetic whose extremely efficient antidepressant effects were recently discovered, was shown to mediate its antidepressant effects via the endogenous opioid system. Thus, although opioid system modulation is a promising therapeutical venue in the treatment of depression further research is warranted to fully understand the benefits and weaknesses of such approach.
Collapse
Affiliation(s)
- Miroslav Adzic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| | - Iva Lukic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milos Mitic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Emilija Glavonic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Nina Dragicevic
- Department of Pharmacy, Singidunum University, Belgrade, Serbia
| | - Sanja Ivkovic
- Department of Molecular Biology and Endocrinology, Vinca - Institute for Nuclear Sciences, National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
5
|
Quintanilla B, Medeiros GC, Greenstein D, Yuan P, Johnston JN, Park LT, Goes F, Gould TD, Zarate CA. κ-Opioid Receptor Plasma Levels Are Associated With Sex and Diagnosis of Major Depressive Disorder But Not Response to Ketamine. J Clin Psychopharmacol 2023; 43:89-96. [PMID: 36821406 PMCID: PMC9992159 DOI: 10.1097/jcp.0000000000001663] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
BACKGROUND Preclinical evidence indicates that the κ-opioid receptor (KOR)/dynorphin pathway is implicated in depressive-like behaviors. Ketamine is believed to partly exert its antidepressant effects by modulating the opioid system. This post hoc study examined the following research questions: (1) at baseline, were there differences in KOR or dynorphin plasma levels between individuals with major depressive disorder (MDD) and healthy volunteers (HVs) or between men and women? (2) in individuals with MDD, did KOR or dynorphin baseline plasma levels moderate ketamine's therapeutic effects or adverse effects? and (3) in individuals with MDD, were KOR or dynorphin plasma levels affected after treatment with ketamine compared with placebo? METHODS Thirty-nine unmedicated individuals with MDD (23 women) and 25 HVs (16 women) received intravenous ketamine (0.5 mg/kg) and placebo in a randomized, crossover, double-blind trial. Blood was obtained from all participants at baseline and at 3 postinfusion time points (230 minutes, day 1, day 3). Linear mixed model regressions were used. RESULTS At baseline, participants with MDD had lower KOR plasma levels than HVs ( F1,60 = 13.16, P < 0.001), and women (MDD and HVs) had higher KOR plasma levels than men ( F1,60 = 4.98, P = 0.03). Diagnosis and sex had no significant effects on baseline dynorphin levels. Baseline KOR and dynorphin levels did not moderate ketamine's therapeutic or adverse effects. Compared with placebo, ketamine was not associated with postinfusion changes in KOR or dynorphin levels. CONCLUSIONS In humans, diagnosis of MDD and biological sex are involved with changes in components of the KOR/dynorphin pathway. Neither KOR nor dynorphin levels consistently moderated ketamine's therapeutic effects or adverse effects, nor were levels altered after ketamine infusion. TRIAL REGISTRATION NCT00088699 ( ClinicalTrials.gov ).
Collapse
Affiliation(s)
- Brandi Quintanilla
- Experimental Therapeutics & Pathophysiology Branch, NIMH-NIH, Bethesda, MD, USA
| | - Gustavo C. Medeiros
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Dede Greenstein
- Experimental Therapeutics & Pathophysiology Branch, NIMH-NIH, Bethesda, MD, USA
| | - Peixiong Yuan
- Experimental Therapeutics & Pathophysiology Branch, NIMH-NIH, Bethesda, MD, USA
| | - Jenessa N. Johnston
- Experimental Therapeutics & Pathophysiology Branch, NIMH-NIH, Bethesda, MD, USA
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lawrence T. Park
- Experimental Therapeutics & Pathophysiology Branch, NIMH-NIH, Bethesda, MD, USA
| | - Fernando Goes
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Todd D. Gould
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, MD, USA
- Departments of Pharmacology and Anatomy & Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
- Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Carlos A. Zarate
- Experimental Therapeutics & Pathophysiology Branch, NIMH-NIH, Bethesda, MD, USA
| |
Collapse
|
6
|
Limoges A, Yarur HE, Tejeda HA. Dynorphin/kappa opioid receptor system regulation on amygdaloid circuitry: Implications for neuropsychiatric disorders. Front Syst Neurosci 2022; 16:963691. [PMID: 36276608 PMCID: PMC9579273 DOI: 10.3389/fnsys.2022.963691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Amygdaloid circuits are involved in a variety of emotional and motivation-related behaviors and are impacted by stress. The amygdala expresses several neuromodulatory systems, including opioid peptides and their receptors. The Dynorphin (Dyn)/kappa opioid receptor (KOR) system has been implicated in the processing of emotional and stress-related information and is expressed in brain areas involved in stress and motivation. Dysregulation of the Dyn/KOR system has also been implicated in various neuropsychiatric disorders. However, there is limited information about the role of the Dyn/KOR system in regulating amygdala circuitry. Here, we review the literature on the (1) basic anatomy of the amygdala, (2) functional regulation of synaptic transmission by the Dyn/KOR system, (3) anatomical architecture and function of the Dyn/KOR system in the amygdala, (4) regulation of amygdala-dependent behaviors by the Dyn/KOR system, and (5) future directions for the field. Future work investigating how the Dyn/KOR system shapes a wide range of amygdala-related behaviors will be required to increase our understanding of underlying circuitry modulation by the Dyn/KOR system. We anticipate that continued focus on the amygdala Dyn/KOR system will also elucidate novel ways to target the Dyn/KOR system to treat neuropsychiatric disorders.
Collapse
Affiliation(s)
- Aaron Limoges
- Unit on Neuromodulation and Synaptic Integration, Bethesda, MD, United States
- NIH-Columbia University Individual Graduate Partnership Program, National Institutes of Health, Bethesda, MD, United States
- Department of Biological Sciences, Columbia University, New York, NY, United States
| | - Hector E. Yarur
- Unit on Neuromodulation and Synaptic Integration, Bethesda, MD, United States
| | - Hugo A. Tejeda
- Unit on Neuromodulation and Synaptic Integration, Bethesda, MD, United States
- *Correspondence: Hugo A. Tejeda,
| |
Collapse
|
7
|
Kaya-Akyüzlü D, Özkan-Kotiloğlu S, Yalçın-Şahiner Ş, Ağtaş-Ertan E, Özgür-İlhan İ. Association of PDYN 68-bp VNTR polymorphism with sublingual buprenorphine/naloxone treatment and with opioid or alcohol use disorder: Effect on craving, depression, anxiety and age onset of first use. Eur J Pharmacol 2022; 921:174862. [PMID: 35271823 DOI: 10.1016/j.ejphar.2022.174862] [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: 12/18/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 11/03/2022]
Abstract
In this case-control study (423 Turkish subjects), the functional pro-dynorphin (PDYN) 68-bp VNTR polymorphism was genotyped in opioid users receiving sublingual buprenorphine/naloxone treatment (SBNT; n = 129, 119 males and 10 females), in opioid users (OUD; n = 99, 90 males and 9 females), in alcohol users (AUD; n = 75, 75 males) and in controls (n = 120, 109 males and 11 females) to determine the effect of this polymorphism on different treatment responses, heroin or alcohol dependence as well as age onset of first use. The PDYN 68-bp alleles were determined based on the number of repeats and genotypes were classified as "short/short (SS)", "short-long (SL)" and "long-long (LL)". The intensity of craving, withdrawal, depression and anxiety were measured by the Substance Craving Scale (SCS), the Clinical Opiate Withdrawal Scale (COWS), the Beck Depression Inventory-II (BDI-II) and Beck Anxiety Inventory (BAI), respectively. Healthy controls (5.5 ± 5.8) had significantly lower levels of depressive symptoms compared to OUD (25.4 ± 13.5), AUD (22.5 ± 11.3) and SBNT (19.29 ± 12.2) groups. In OUD group, the LL genotype was associated with decreased intensity of anxiety and depressive symptoms than the SS+SL genotype. The BDI-II scores for PDYN VNTR genotypes within the 4 groups were analysed by two-way ANOVA and statistical differences were found for the groups. SBNT group had significantly lower COWS score than OUD group (1.00 versus 3.00). There were statistically significant differences in the median BAI (11 versus 24) and BDI-II scores (17.5 versus 25) between OUD and SBNT groups, supporting the antidepressant and anxiolytic effects of SBNT in persons with OUD.
Collapse
Affiliation(s)
| | - Selin Özkan-Kotiloğlu
- Kırşehir Ahi Evran University, Faculty of Science and Art, Department of Molecular Biology and Genetics, Kırşehir, Turkey
| | | | - Ece Ağtaş-Ertan
- Ankara University, Faculty of Medicine, Department of Mental Health and Diseases, Ankara, Turkey
| | - İnci Özgür-İlhan
- Ankara University, Faculty of Medicine, Department of Mental Health and Diseases, Ankara, Turkey
| |
Collapse
|
8
|
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: 16] [Impact Index Per Article: 5.3] [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
|
9
|
Lin S, Du Y, Xia Y, Xie Y, Xiao L, Wang G. Advances in optogenetic studies of depressive-like behaviors and underlying neural circuit mechanisms. Front Psychiatry 2022; 13:950910. [PMID: 36159933 PMCID: PMC9492959 DOI: 10.3389/fpsyt.2022.950910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUNDS The neural circuit mechanisms underlying depression remain unclear. Recently optogenetics has gradually gained recognition as a novel technique to regulate the activity of neurons with light stimulation. Scientists are now transferring their focus to the function of brain regions and neural circuits in the pathogenic progress of depression. Deciphering the circuitry mechanism of depressive-like behaviors may help us better understand the symptomatology of depression. However, few studies have summarized current progress on optogenetic researches into the neural circuit mechanisms of depressive-like behaviors. AIMS This review aimed to introduce fundamental characteristics and methodologies of optogenetics, as well as how this technique achieves specific neuronal control with spatial and temporal accuracy. We mainly summarized recent progress in neural circuit discoveries in depressive-like behaviors using optogenetics and exhibited the potential of optogenetics as a tool to investigate the mechanism and possible optimization underlying antidepressant treatment such as ketamine and deep brain stimulation. METHODS A systematic review of the literature published in English mainly from 2010 to the present in databases was performed. The selected literature is then categorized and summarized according to their neural circuits and depressive-like behaviors. CONCLUSIONS Many important discoveries have been made utilizing optogenetics. These findings support optogenetics as a powerful and potential tool for studying depression. And our comprehension to the etiology of depression and other psychiatric disorders will also be more thorough with this rapidly developing technique in the near future.
Collapse
Affiliation(s)
- Shanshan Lin
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiwei Du
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yujie Xia
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yumeng Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Noto T, Zhou G, Yang Q, Lane G, Zelano C. Human Primary Olfactory Amygdala Subregions Form Distinct Functional Networks, Suggesting Distinct Olfactory Functions. Front Syst Neurosci 2021; 15:752320. [PMID: 34955769 PMCID: PMC8695617 DOI: 10.3389/fnsys.2021.752320] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/08/2021] [Indexed: 12/02/2022] Open
Abstract
Three subregions of the amygdala receive monosynaptic projections from the olfactory bulb, making them part of the primary olfactory cortex. These primary olfactory areas are located at the anterior-medial aspect of the amygdala and include the medial amygdala (MeA), cortical amygdala (CoA), and the periamygdaloid complex (PAC). The vast majority of research on the amygdala has focused on the larger basolateral and basomedial subregions, which are known to be involved in implicit learning, threat responses, and emotion. Fewer studies have focused on the MeA, CoA, and PAC, with most conducted in rodents. Therefore, our understanding of the functions of these amygdala subregions is limited, particularly in humans. Here, we first conducted a review of existing literature on the MeA, CoA, and PAC. We then used resting-state fMRI and unbiased k-means clustering techniques to show that the anatomical boundaries of human MeA, CoA, and PAC accurately parcellate based on their whole-brain resting connectivity patterns alone, suggesting that their functional networks are distinct, relative both to each other and to the amygdala subregions that do not receive input from the olfactory bulb. Finally, considering that distinct functional networks are suggestive of distinct functions, we examined the whole-brain resting network of each subregion and speculated on potential roles that each region may play in olfactory processing. Based on these analyses, we speculate that the MeA could potentially be involved in the generation of rapid motor responses to olfactory stimuli (including fight/flight), particularly in approach/avoid contexts. The CoA could potentially be involved in olfactory-related reward processing, including learning and memory of approach/avoid responses. The PAC could potentially be involved in the multisensory integration of olfactory information with other sensory systems. These speculations can be used to form the basis of future studies aimed at clarifying the olfactory functions of these under-studied primary olfactory areas.
Collapse
Affiliation(s)
- Torben Noto
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Guangyu Zhou
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Qiaohan Yang
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Gregory Lane
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Christina Zelano
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
11
|
Abstract
After participating in this activity, learners should be better able to:• Identify the effects of dysregulated opioid signalling in depression• Evaluate the use of opioid compounds and ketamine in patients with depression ABSTRACT: Major depressive disorder (MDD) remains one of the leading causes of disability and functional impairment worldwide. Current antidepressant therapeutics require weeks to months of treatment prior to the onset of clinical efficacy on depressed mood but remain ineffective in treating suicidal ideation and cognitive impairment. Moreover, 30%-40% of individuals fail to respond to currently available antidepressant medications. MDD is a heterogeneous disorder with an unknown etiology; novel strategies must be developed to treat MDD more effectively. Emerging evidence suggests that targeting one or more of the four opioid receptors-mu (MOR), kappa (KOR), delta (DOR), and the nociceptin/orphanin FQ receptor (NOP)-may yield effective therapeutics for stress-related psychiatric disorders. Furthermore, the effects of the rapidly acting antidepressant ketamine may involve opioid receptors. This review highlights dysregulated opioid signaling in depression, evaluates clinical trials with opioid compounds, and considers the role of opioid mechanisms in rapidly acting antidepressants.
Collapse
|
12
|
Boehm MA, Bonaventura J, Gomez JL, Solís O, Stein EA, Bradberry CW, Michaelides M. Translational PET applications for brain circuit mapping with transgenic neuromodulation tools. Pharmacol Biochem Behav 2021; 204:173147. [PMID: 33549570 PMCID: PMC8297666 DOI: 10.1016/j.pbb.2021.173147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 02/08/2023]
Abstract
Transgenic neuromodulation tools have transformed the field of neuroscience over the past two decades by enabling targeted manipulation of neuronal populations and circuits with unprecedented specificity. Chemogenetic and optogenetic neuromodulation systems are among the most widely used and allow targeted control of neuronal activity through the administration of a selective compound or light, respectively. Innovative genetic targeting strategies are utilized to transduce specific cells to express transgenic receptors and opsins capable of manipulating neuronal activity. These allow mapping of neuroanatomical projection sites and link cellular manipulations with brain circuit functions and behavior. As these tools continue to expand knowledge of the nervous system in preclinical models, developing translational applications for human therapies is becoming increasingly possible. However, new strategies for implementing and monitoring transgenic tools are needed for safe and effective use in translational research and potential clinical applications. A major challenge for such applications is the need to track the location and function of chemogenetic receptors and opsins in vivo, and new developments in positron emission tomography (PET) imaging techniques offer promising solutions. The goal of this review is to summarize current research combining transgenic tools with PET for in vivo mapping and manipulation of brain circuits and to propose future directions for translational applications.
Collapse
Affiliation(s)
- Matthew A Boehm
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States; Department of Neuroscience, Brown University, Providence, RI 02906, United States.
| | - Jordi Bonaventura
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Juan L Gomez
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Oscar Solís
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Elliot A Stein
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Charles W Bradberry
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States.
| | - Michael Michaelides
- National Institute on Drug Abuse Intramural Research Program, 251 Bayview Blvd, Baltimore, MD 21224, United States; Department of Psychiatry & Behavioral Sciences, Johns Hopkins Medicine, Baltimore, MD, 21205, United States.
| |
Collapse
|
13
|
Kappa Opioid Receptors in the Pathology and Treatment of Major Depressive Disorder. Handb Exp Pharmacol 2021; 271:493-524. [PMID: 33580854 DOI: 10.1007/164_2020_432] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The kappa opioid receptor (KOR) is thought to regulate neural systems associated with anhedonia and aversion and mediate negative affective states that are associated with a number of psychiatric disorders, but especially major depressive disorder (MDD). Largely because KOR antagonists mitigate the effects of stress in preclinical studies, KOR antagonists have been recommended as novel drugs for treating MDD. The purpose of this review is to examine the role of KORs and its endogenous ligand dynorphins (DYNs) in the pathology and treatment of MDD derived from different types of clinical studies. Evidence pertaining to the role of KOR and MDD will be reviewed from (1) post mortem mRNA expression patterns in MDD, (2) the utility of KOR neuroimaging agents and serum biomarkers in MDD, and (3) evidence from the recent Fast Fail clinical trial that established KOR antagonism as a potential therapeutic strategy for the alleviation of anhedonia, a core feature of MDD. These findings are compared with a focused evaluation of stress-induced alterations in OPRK and PDYN mRNA expression. Finally, the current status of the effects of KOR antagonists on behavioral phenotypes of stress in preclinical studies related to MDD is summarized.
Collapse
|
14
|
Gold MS, Baron D, Bowirrat A, Blum K. Neurological correlates of brain reward circuitry linked to opioid use disorder (OUD): Do homo sapiens acquire or have a reward deficiency syndrome? J Neurol Sci 2020; 418:117137. [PMID: 32957037 PMCID: PMC7490287 DOI: 10.1016/j.jns.2020.117137] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/19/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
The extant literature confirms that an array of polymorphic genes related to- neurotransmitters and second messengers govern the net release of dopamine in the Nucleus Accumbens (NAc) in the mesolimbic region of the brain. They are linked predominantly to motivation, anti-stress, incentive salience (wanting), and wellbeing. Notably, in 2000 the Nobel Prize was awarded to Carlsson, Greengard, and Kandel for their work on the molecular and cellular function of dopaminergic activity at neurons. This historical psychopharmacological work involved neurotransmission of serotonin, endorphins, glutamate, and dopamine, and the seminal work of Blum, Gold, Volkow, Nestler, and others related to neurotransmitter function and related behaviors. Currently, Americans are facing their second and worst opioid epidemic, prescribed opioids, and easy access drive this epidemic of overdoses, and opioid use disorders (OUDs). Presently the clinical consensus is to treat OUD, as if it were an opioid deficiency syndrome, with long-term to life-long opioid substitution therapy. Opioid agonist administration is seen as necessary to replace missing opioids, treat OUD, and prevent overdoses, like insulin is used to treat diabetes. Treatment of OUD and addiction, in general, is similar to the endocrinopathy conceptualization in that it views opioid agonist MATs as an essential core to therapy. Is this approach logical? Other than as harm reduction, is using opioids to treat OUD therapeutic or harmful in the long term? This historical Trieste provides a molecular framework to understand the current underpinnings of endorphinergic/dopaminergic mechanisms related to opioid deficiency syndrome and generalized reward processing depletion. WC 249.
Collapse
Affiliation(s)
- Mark S Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States.
| | - David Baron
- Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
| | - Abdalla Bowirrat
- Department of Neuroscience and Genetics, Interdisciplinary Center Herzliya, Israel
| | - Kenneth Blum
- Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
| |
Collapse
|
15
|
Peeters LM, Missault S, Keliris AJ, Keliris GA. Combining designer receptors exclusively activated by designer drugs and neuroimaging in experimental models: A powerful approach towards neurotheranostic applications. Br J Pharmacol 2020; 177:992-1002. [PMID: 31658365 PMCID: PMC7042113 DOI: 10.1111/bph.14885] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 11/30/2022] Open
Abstract
The combination of chemogenetics targeting specific brain cell populations with in vivo imaging techniques provides scientists with a powerful new tool to study functional neural networks at the whole-brain scale. A number of recent studies indicate the potential of this approach to increase our understanding of brain function in health and disease. In this review, we discuss the employment of a specific chemogenetic tool, designer receptors exclusively activated by designer drugs, in conjunction with non-invasive neuroimaging techniques such as PET and MRI. We highlight the utility of using this multiscale approach in longitudinal studies and its ability to identify novel brain circuits relevant to behaviour that can be monitored in parallel. In addition, some identified shortcomings in this technique and more recent efforts to overcome them are also presented. Finally, we discuss the translational potential of designer receptors exclusively activated by designer drugs in neuroimaging and the promise it holds for future neurotheranostic applications.
Collapse
|
16
|
Jacobson ML, Browne CA, Lucki I. Kappa Opioid Receptor Antagonists as Potential Therapeutics for Stress-Related Disorders. Annu Rev Pharmacol Toxicol 2020; 60:615-636. [DOI: 10.1146/annurev-pharmtox-010919-023317] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Exposure to stressful stimuli activates kappa opioid receptor (KOR) signaling, a process known to produce aversion and dysphoria in humans and other species. This endogenous opioid system is dysregulated in stress-related disorders, specifically in major depressive disorder (MDD). These findings serve as the foundation for a growing interest in the therapeutic potential of KOR antagonists as novel antidepressants. In this review, data supporting the hypothesis of dysregulated KOR function in MDD are considered. The clinical data demonstrating the therapeutic efficacy and safety of selective and mixed opioid antagonists are then presented. Finally, the preclinical evidence illustrating the induction of behaviors relevant to the endophenotypes of MDD and KOR antagonist activity in stress-naïve and stress-exposed animals is evaluated. Overall, this review highlights the emergent literature supporting the pursuit of KOR antagonists as novel therapeutics for MDD and other stress-related disorders.
Collapse
Affiliation(s)
- Moriah L. Jacobson
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Caroline A. Browne
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| | - Irwin Lucki
- Department of Pharmacology & Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
- Department of Psychiatry, Uniformed Services University of the Health Sciences, Bethesda, Maryland 20814, USA
| |
Collapse
|
17
|
Burke NN, Li Y, Deaver DR, Finn DP, Roche M, Eyerman DJ, Sanchez C, Kelly JP. Chronic administration of buprenorphine in combination with samidorphan produces sustained effects in olfactory bulbectomised rats and Wistar-Kyoto rats. J Psychopharmacol 2019; 33:1620-1627. [PMID: 31512988 DOI: 10.1177/0269881119872203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The combination of buprenorphine, a partial mu-opioid receptor agonist and a functional kappa-opioid receptor antagonist, with samidorphan, a functional mu-opioid receptor antagonist, is being developed as an adjunct therapy for major depressive disorder, in order to harness the mood-enhancing effects of opioids without unwanted side-effects such as a risk of addiction. Acute and subacute administration of the combination of buprenorphine and samidorphan is effective in reducing forced swim immobility in the Wistar-Kyoto rat, but the chronic effects have not been examined. AIMS AND METHODS The purpose of this study was to assess if chronic (14-day) administration of buprenorphine (0.1 mg/kg, subcutaneous) alone or in combination with samidorphan (0.3 mg/kg, subcutaneous) maintains antidepressant-like activity in the olfactory bulbectomised rat model and the Wistar-Kyoto rat, two models that exhibit ongoing behavioural deficits in tests commonly used to study effects of antidepressants. RESULTS Olfactory bulbectomised-induced hyperactivity was attenuated by chronic administration of buprenorphine alone and in combination with samidorphan, to that of sham control activity levels. Neither buprenorphine nor samidorphan altered stress-associated defecation in sham or olfactory bulbectomised rats in the open field. In Wistar-Kyoto rats, buprenorphine alone significantly reduced forced swim immobility and increased locomotor activity three hours post-final dosing. Buprenorphine plus samidorphan significantly reduced forced swim immobility without changing locomotor activity at this time point. Buprenorphine alone also significantly reduced forced swim immobility 24 h post-final dosing. CONCLUSION Chronic treatment of buprenorphine alone or buprenorphine plus samidorphan is effective in reversing behavioural deficits in distinct non-clinical paradigms. These non-clinical results complement the antidepressant effect of this combination observed in clinical studies.
Collapse
Affiliation(s)
- Nikita N Burke
- Pharmacology and Therapeutics, National University of Ireland, Galway, Ireland
| | - Yan Li
- Alkermes Inc., Waltham, MA, USA
| | | | - David P Finn
- Pharmacology and Therapeutics, National University of Ireland, Galway, Ireland
| | - Michelle Roche
- Physiology, National University of Ireland, Galway, Ireland
| | | | | | - John P Kelly
- Pharmacology and Therapeutics, National University of Ireland, Galway, Ireland
| |
Collapse
|
18
|
Biselli T, Lange SS, Sablottny L, Steffen J, Walther A. Optogenetic and chemogenetic insights into the neurocircuitry of depression-like behaviour: A systematic review. Eur J Neurosci 2019; 53:9-38. [PMID: 31633833 DOI: 10.1111/ejn.14603] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 09/13/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Abstract
Major depressive disorder (MDD) and its treatment are challenges for global health. Optogenetics and chemogenetics are driving MDD research forward by unveiling causal relations between cell-type-specific control of neurons and depressive-like behaviour in rodents. Using a systematic search process, in this review, a set of 43 original studies applying optogenetic or chemogenetic techniques in rodent models of depression was identified. Our aim was to provide an examination of all available studies elucidating central neuronal mechanisms leading to depressive-like behaviour in rodents and thereby unveiling the most promising routes for future research. A complex interacting network of relevant structures, in which central circuitries causally related to depressive-like behaviour are implicated, has been identified. As most relevant structures emerge: medial prefrontal cortex, anterior cingulate cortex, amygdala, nucleus accumbens, ventral tegmental area, hippocampus and raphe nuclei. Further evidence, though examined by only few studies, emerges for structures like the lateral habenula, or medial dorsal thalamus. Most of the identified brain areas have previously been associated with MDD neuropathology, but now evidence can be provided for causal pathological mechanisms within a complex cortico-limbic reward circuitry. However, the studies also show conflicting results concerning the mechanisms underlying the causal involvement of specific circuitries. Comparability of studies is partly limited since even small deviations in methodological approaches lead to different outcomes. Factors influencing study outcomes were identified and need to be considered in future studies (e.g. frequency used for stimulation, time and duration of stimulation, limitations of applied animal models of MDD).
Collapse
Affiliation(s)
- Tom Biselli
- Biological Psychology, TU Dresden, Dresden, Germany
| | | | | | | | - Andreas Walther
- Biological Psychology, TU Dresden, Dresden, Germany.,Clinical Psychology and Psychotherapy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Yun H, Park ES, Choi S, Shin B, Yu J, Yu J, Amarasekara DS, Kim S, Lee N, Choi JS, Choi Y, Rho J. TDAG51 is a crucial regulator of maternal care and depressive-like behavior after parturition. PLoS Genet 2019; 15:e1008214. [PMID: 31251738 PMCID: PMC6599150 DOI: 10.1371/journal.pgen.1008214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 05/27/2019] [Indexed: 12/11/2022] Open
Abstract
Postpartum depression is a severe emotional and mental disorder that involves maternal care defects and psychiatric illness. Postpartum depression is closely associated with a combination of physical changes and physiological stress during pregnancy or after parturition in stress-sensitive women. Although postpartum depression is relatively well known to have deleterious effects on the developing fetus, the influence of genetic risk factors on the development of postpartum depression remains unclear. In this study, we discovered a novel function of T cell death-associated gene 51 (TDAG51/PHLDA1) in the regulation of maternal and depressive-like behavior. After parturition, TDAG51-deficient dams showed impaired maternal behavior in pup retrieving, nursing and nest building tests. In contrast to the normal dams, the TDAG51-deficient dams also exhibited more sensitive depressive-like behaviors after parturition. Furthermore, changes in the expression levels of various maternal and depressive-like behavior-associated genes regulating neuroendocrine factor and monoamine neurotransmitter levels were observed in TDAG51-deficient postpartum brain tissues. These findings indicate that TDAG51 plays a protective role against maternal care defects and depressive-like behavior after parturition. Thus, TDAG51 is a maternal care-associated gene that functions as a crucial regulator of maternal and depressive-like behavior after parturition. Postpartum depression is a severe emotional and mental disease that can affect women typically after parturition. However, the genetic risk factors associated with the development of postpartum depression are still largely unknown. We discovered a novel function of T cell death-associated gene 51 (TDAG51) in the regulation of maternal behavior and postpartum depression. We report that TDAG51 deficiency induces depressive-like and abnormal maternal behavior after parturition. The loss of TDAG51 in postpartum brain tissues induces changes in the expression levels of various maternal and depressive-like behavior-associated genes that regulate the levels of neuroendocrine factors and monoamine neurotransmitters. TDAG51 is a maternal care-associated gene that functions as a crucial regulator of maternal and depressive-like behavior after parturition.
Collapse
Affiliation(s)
- Hyeongseok Yun
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Eui-Soon Park
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Seunga Choi
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Bongjin Shin
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jungeun Yu
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jiyeon Yu
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | | | - Sumi Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Nari Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jong-Soon Choi
- Division of Life Science, Korea Basic Science Institute, Daejeon, Korea
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
- * E-mail:
| |
Collapse
|
20
|
Egervari G, Kozlenkov A, Dracheva S, Hurd YL. Molecular windows into the human brain for psychiatric disorders. Mol Psychiatry 2019; 24:653-673. [PMID: 29955163 PMCID: PMC6310674 DOI: 10.1038/s41380-018-0125-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/14/2018] [Accepted: 06/05/2018] [Indexed: 12/20/2022]
Abstract
Delineating the pathophysiology of psychiatric disorders has been extremely challenging but technological advances in recent decades have facilitated a deeper interrogation of molecular processes in the human brain. Initial candidate gene expression studies of the postmortem brain have evolved into genome wide profiling of the transcriptome and the epigenome, a critical regulator of gene expression. Here, we review the potential and challenges of direct molecular characterization of the postmortem human brain, and provide a brief overview of recent transcriptional and epigenetic studies with respect to neuropsychiatric disorders. Such information can now be leveraged and integrated with the growing number of genome-wide association databases to provide a functional context of trait-associated genetic variants linked to psychiatric illnesses and related phenotypes. While it is clear that the field is still developing and challenges remain to be surmounted, these recent advances nevertheless hold tremendous promise for delineating the neurobiological underpinnings of mental diseases and accelerating the development of novel medication strategies.
Collapse
Affiliation(s)
- Gabor Egervari
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Addiction Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, School of Medicine at Mount Sinai, New York, NY, USA
- Epigenetics Institute and Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alexey Kozlenkov
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Stella Dracheva
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, School of Medicine at Mount Sinai, New York, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Yasmin L Hurd
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Addiction Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
21
|
Browne CA, Lucki I. Targeting opioid dysregulation in depression for the development of novel therapeutics. Pharmacol Ther 2019; 201:51-76. [PMID: 31051197 DOI: 10.1016/j.pharmthera.2019.04.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 04/23/2019] [Indexed: 02/07/2023]
Abstract
Since the serendipitous discovery of the first class of modern antidepressants in the 1950's, all pharmacotherapies approved by the Food and Drug Administration for major depressive disorder (MDD) have shared a common mechanism of action, increased monoaminergic neurotransmission. Despite the widespread availability of antidepressants, as many as 50% of depressed patients are resistant to these conventional therapies. The significant length of time required to produce meaningful symptom relief with these medications, 4-6 weeks, indicates that other mechanisms are likely involved in the pathophysiology of depression which may yield more viable targets for drug development. For decades, no viable candidate target with a different mechanism of action to that of conventional therapies proved successful in clinical studies. Now several exciting avenues for drug development are under intense investigation. One of these emerging targets is modulation of endogenous opioid tone. This review will evaluate preclinical and clinical evidence pertaining to opioid dysregulation in depression, focusing on the role of the endogenous ligands endorphin, enkephalin, dynorphin, and nociceptin/orphanin FQ (N/OFQ) and their respective receptors, mu (MOR), delta (DOR), kappa (KOR), and the N/OFQ receptor (NOP) in mediating behaviors relevant to depression and anxiety. Finally, putative opioid based antidepressants that are under investigation in clinical trials, ALKS5461, JNJ-67953964 (formerly LY2456302 and CERC-501) and BTRX-246040 (formerly LY-2940094) will be discussed. This review will illustrate the potential therapeutic value of targeting opioid dysregulation in developing novel therapies for MDD.
Collapse
Affiliation(s)
- Caroline A Browne
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America
| | - Irwin Lucki
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, United States of America.
| |
Collapse
|
22
|
Burns JA, Kroll DS, Feldman DE, Kure Liu C, Manza P, Wiers CE, Volkow ND, Wang GJ. Molecular Imaging of Opioid and Dopamine Systems: Insights Into the Pharmacogenetics of Opioid Use Disorders. Front Psychiatry 2019; 10:626. [PMID: 31620026 PMCID: PMC6759955 DOI: 10.3389/fpsyt.2019.00626] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/05/2019] [Indexed: 12/21/2022] Open
Abstract
Opioid use in the United States has steadily risen since the 1990s, along with staggering increases in addiction and overdose fatalities. With this surge in prescription and illicit opioid abuse, it is paramount to understand the genetic risk factors and neuropsychological effects of opioid use disorder (OUD). Polymorphisms disrupting the opioid and dopamine systems have been associated with increased risk for developing substance use disorders. Molecular imaging studies have revealed how these polymorphisms impact the brain and contribute to cognitive and behavioral differences across individuals. Here, we review the current molecular imaging literature to assess how genetic variations in the opioid and dopamine systems affect function in the brain's reward, cognition, and stress pathways, potentially resulting in vulnerabilities to OUD. Continued research of the functional consequences of genetic variants and corresponding alterations in neural mechanisms will inform prevention and treatment of OUD.
Collapse
Affiliation(s)
- Jamie A Burns
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Danielle S Kroll
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Dana E Feldman
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | | | - Peter Manza
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States.,National Institute on Drug Abuse, Bethesda, MD, United States
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| |
Collapse
|
23
|
Abstract
The failure of traditional antidepressant medications to adequately target cognitive impairment is associated with poor treatment response, increased risk of relapse, and greater lifetime disability. Opioid receptor antagonists are currently under development as novel therapeutics for major depressive disorder (MDD) and other stress-related illnesses. Although it is known that dysregulation of the endogenous opioid system is observed in patients diagnosed with MDD, the impact of opioidergic neurotransmission on cognitive impairment has not been systematically evaluated. Here we review the literature indicating that opioid manipulations can alter cognitive functions in humans. Furthermore, we detail the preclinical studies that demonstrate the ability of mu-opioid receptor and kappa-opioid receptor ligands to modulate several cognitive processes. Specifically, this review focuses on domains within higher order cognitive processing, including attention and executive functioning, which can differentiate cognitive processes influenced by motivational state.
Collapse
|
24
|
Lee KH, Yen CF. The relationships between depression, neuroticism, and attitudes (NDA model) in heroin abusers in Taiwan. Am J Addict 2018; 27:139-143. [DOI: 10.1111/ajad.12691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 12/28/2017] [Accepted: 02/06/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Kun-Hua Lee
- Ministry of Health and Welfare; Yuli Hospital; Yuli Township Hualien County Taiwan
- Department of Counseling and Clinical Psychology; National Dong Hwa University; Shoufeng Hualien Taiwan
| | - Cheng-Fang Yen
- Department of Psychiatry; Kaohsiung Medical University Hospital; Kaohsiung City Taiwan
- Department of Psychiatry; School of Medicine and Graduate Institute of Medicine; College of Medicine; Kaohsiung Medical University; Kaohsiung City Taiwan
| |
Collapse
|
25
|
Epigenetic regulation of the kappa opioid receptor gene by an insertion-deletion in the promoter region. Eur Neuropsychopharmacol 2018; 28:334-340. [PMID: 29336871 DOI: 10.1016/j.euroneuro.2017.12.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/05/2017] [Accepted: 12/08/2017] [Indexed: 12/28/2022]
Abstract
Preclinical and clinical studies have demonstrated that the kappa opioid receptor (KOR) regulates reward, hedonic tone and emotions. At therapeutic level, on-going clinical trials are assessing the potential of targeting the KOR for the management of depression, anxiety disorders and substance use disorders. However, genetic polymorphisms in the KOR gene that potentially contribute to its implication in these phenotypes have been poorly studied. Here we investigated an insertion-deletion in the promoter region of KOR (rs35566036), recently associated with alcohol addiction, in a cohort of depressed subjects who died by suicide, as well as psychiatrically healthy individuals. Focusing on 3 brain regions (anterior insula, anterior cingulate cortex, and mediodorsal thalamus), we characterized the functional impact of this structural variant on the expression and patterns of DNA methylation of the KOR gene, using qPCR and targeted Bisulfite-Sequencing, respectively. While there was no significant change in the expression of KOR as a function of the insertion-deletion, or as a function of disease status in any brain region, we found that this variant strongly determines DNA methylation in KOR promoter, leading to a significant decrease in methylation levels of 8 nearby CpG dinucleotides located approximately 500 base pairs upstream the transcription start site. In addition, our results suggest a possible association between the insertion-deletion and depression; however, this result should be tested in larger populations. In sum, in this study we uncovered an epigenetic mechanism potentially contributing to KOR dysfunction in carriers of the insertion-deletion.
Collapse
|
26
|
Mazzone CM, Pati D, Michaelides M, DiBerto J, Fox JH, Tipton G, Anderson C, Duffy K, McKlveen JM, Hardaway JA, Magness ST, Falls WA, Hammack SE, McElligott ZA, Hurd YL, Kash TL. Acute engagement of G q-mediated signaling in the bed nucleus of the stria terminalis induces anxiety-like behavior. Mol Psychiatry 2018; 23:143-153. [PMID: 27956747 PMCID: PMC5468515 DOI: 10.1038/mp.2016.218] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 09/21/2016] [Accepted: 10/11/2016] [Indexed: 01/23/2023]
Abstract
The bed nucleus of the stria terminalis (BNST) is a brain region important for regulating anxiety-related behavior in both humans and rodents. Here we used a chemogenetic strategy to investigate how engagement of G protein-coupled receptor (GPCR) signaling cascades in genetically defined GABAergic BNST neurons modulates anxiety-related behavior and downstream circuit function. We saw that stimulation of vesicular γ-aminobutyric acid (GABA) transporter (VGAT)-expressing BNST neurons using hM3Dq, but neither hM4Di nor rM3Ds designer receptors exclusively activated by a designer drug (DREADD), promotes anxiety-like behavior. Further, we identified that activation of hM3Dq receptors in BNST VGAT neurons can induce a long-term depression-like state of glutamatergic synaptic transmission, indicating DREADD-induced changes in synaptic plasticity. Further, we used DREADD-assisted metabolic mapping to profile brain-wide network activity following activation of Gq-mediated signaling in BNST VGAT neurons and saw increased activity within ventral midbrain structures, including the ventral tegmental area and hindbrain structures such as the locus coeruleus and parabrachial nucleus. These results highlight that Gq-mediated signaling in BNST VGAT neurons can drive downstream network activity that correlates with anxiety-like behavior and points to the importance of identifying endogenous GPCRs within genetically defined cell populations. We next used a microfluidics approach to profile the receptorome of single BNST VGAT neurons. This approach yielded multiple Gq-coupled receptors that are associated with anxiety-like behavior and several potential novel candidates for regulation of anxiety-like behavior. From this, we identified that stimulation of the Gq-coupled receptor 5-HT2CR in the BNST is sufficient to elevate anxiety-like behavior in an acoustic startle task. Together, these results provide a novel profile of receptors within genetically defined BNST VGAT neurons that may serve as therapeutic targets for regulating anxiety states and provide a blueprint for examining how G-protein-mediated signaling in a genetically defined cell type can be used to assess behavior and brain-wide circuit function.
Collapse
Affiliation(s)
- Christopher M. Mazzone
- Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC 27599
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Dipanwita Pati
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Michael Michaelides
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, Neuroimaging Research Branch, National Institute on Drug Abuse, Baltimore, MD 21224
| | - Jeffrey DiBerto
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - James H. Fox
- Department of Psychology, University of Vermont, Burlington, VT 05405
| | - Gregory Tipton
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Carlton Anderson
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, NC 27599
| | - Kelly Duffy
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Jessica M. McKlveen
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - J. Andrew Hardaway
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
| | - Scott T. Magness
- Department of Medicine, University of North Carolina, Chapel Hill, NC 27599
- Department of Cell Biology and Physiology, and Biomedical Engineering, University of North Carolina, Chapel Hill, NC 27599
| | - William A. Falls
- Department of Psychology, University of Vermont, Burlington, VT 05405
| | | | - Zoe A. McElligott
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC 27599
| | - Yasmin L. Hurd
- Fishberg Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Department of Psychiatry, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Thomas L. Kash
- Bowles Center for Alcohol Studies, University of North Carolina School of Medicine, Chapel Hill, NC 27599
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, NC, 27599
| |
Collapse
|
27
|
Browne CA, Falcon E, Robinson SA, Berton O, Lucki I. Reversal of Stress-Induced Social Interaction Deficits by Buprenorphine. Int J Neuropsychopharmacol 2017; 21:164-174. [PMID: 29020387 PMCID: PMC5793841 DOI: 10.1093/ijnp/pyx079] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/22/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Patients with post-traumatic stress disorder frequently report persistent problems with social interactions, emerging after a traumatic experience. Chronic social defeat stress is a widely used rodent model of stress that produces robust and sustained social avoidance behavior. The avoidance of other rodents can be reversed by 28 days of treatment with selective serotonin reuptake inhibitors, the only pharmaceutical class approved by the U.S. Food and Drug Administration for treating post-traumatic stress disorder. In this study, the sensitivity of social interaction deficits evoked by 10 days of chronic social defeat stress to prospective treatments for post-traumatic stress disorder was examined. METHODS The effects of acute and repeated treatment with a low dose of buprenorphine (0.25 mg/kg/d) on social interaction deficits in male C57BL/6 mice by chronic social defeat stress were studied. Another cohort of mice was used to determine the effects of the selective serotonin reuptake inhibitor fluoxetine (10 mg/kg/d), the NMDA antagonist ketamine (10 mg/kg/d), and the selective kappa opioid receptor antagonist CERC-501 (1 mg/kg/d). Changes in mRNA expression of Oprm1 and Oprk1 were assessed in a separate cohort. RESULTS Buprenorphine significantly reversed social interaction deficits produced by chronic social defeat stress following 7 days of administration, but not after acute injection. Treatment with fluoxetine for 7 days, but not 24 hours, also reinstated social interaction behavior in mice that were susceptible to chronic social defeat. In contrast, CERC-501 and ketamine failed to reverse social avoidance. Gene expression analysis found: (1) Oprm1 mRNA expression was reduced in the hippocampus and increased in the frontal cortex of susceptible mice and (2) Oprk1 mRNA expression was reduced in the amygdala and increased in the frontal cortex of susceptible mice compared to non-stressed controls and stress-resilient mice. CONCLUSIONS Short-term treatment with buprenorphine and fluoxetine normalized social interaction after chronic social defeat stress. In concert with the changes in opioid receptor expression produced by chronic social defeat stress, we speculate that buprenorphine's efficacy in this model of post-traumatic stress disorder may be associated with the ability of this compound to engage multiple opioid receptors.
Collapse
Affiliation(s)
| | | | | | | | - Irwin Lucki
- Departments of Psychiatry, Philadelphia, Pennsylvania,Systems Pharmacology and Translational Therapeutics, Philadelphia, Pennsylvania,University of Pennsylvania, Philadelphia, Pennsylvania,Correspondence: Irwin Lucki, PhD, Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, 4301 Jones Bridge Road, Bethesda, MD 20814 ()
| |
Collapse
|
28
|
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
|
29
|
Kozlenkov A, Jaffe AE, Timashpolsky A, Apontes P, Rudchenko S, Barbu M, Byne W, Hurd YL, Horvath S, Dracheva S. DNA Methylation Profiling of Human Prefrontal Cortex Neurons in Heroin Users Shows Significant Difference between Genomic Contexts of Hyper- and Hypomethylation and a Younger Epigenetic Age. Genes (Basel) 2017; 8:genes8060152. [PMID: 28556790 PMCID: PMC5485516 DOI: 10.3390/genes8060152] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/21/2017] [Accepted: 05/25/2017] [Indexed: 12/30/2022] Open
Abstract
We employed Illumina 450 K Infinium microarrays to profile DNA methylation (DNAm) in neuronal nuclei separated by fluorescence-activated sorting from the postmortem orbitofrontal cortex (OFC) of heroin users who died from heroin overdose (N = 37), suicide completers (N = 22) with no evidence of heroin use and from control subjects who did not abuse illicit drugs and died of non-suicide causes (N = 28). We identified 1298 differentially methylated CpG sites (DMSs) between heroin users and controls, and 454 DMSs between suicide completers and controls (p < 0.001). DMSs and corresponding genes (DMGs) in heroin users showed significant differences in the preferential context of hyper and hypo DM. HyperDMSs were enriched in gene bodies and exons but depleted in promoters, whereas hypoDMSs were enriched in promoters and enhancers. In addition, hyperDMGs showed preference for genes expressed specifically by glutamatergic as opposed to GABAergic neurons and enrichment for axonogenesis- and synaptic-related gene ontology categories, whereas hypoDMGs were enriched for transcription factor activity- and gene expression regulation-related terms. Finally, we found that the DNAm-based “epigenetic age” of neurons from heroin users was younger than that in controls. Suicide-related results were more difficult to interpret. Collectively, these findings suggest that the observed DNAm differences could represent functionally significant marks of heroin-associated plasticity in the OFC.
Collapse
Affiliation(s)
- Alexey Kozlenkov
- James J. Peters VA Medical Center, Bronx, NY 10468, USA.
- The Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Andrew E Jaffe
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21205, USA.
- Department of Biostatistics and Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, 21205, USA.
| | | | - Pasha Apontes
- James J. Peters VA Medical Center, Bronx, NY 10468, USA.
| | | | - Mihaela Barbu
- Hospital for Special Surgery, New York, NY 10021, USA.
| | - William Byne
- James J. Peters VA Medical Center, Bronx, NY 10468, USA.
- The Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Yasmin L Hurd
- The Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.
- Department of Biostatistics, School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Stella Dracheva
- James J. Peters VA Medical Center, Bronx, NY 10468, USA.
- The Friedman Brain Institute and Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
30
|
Ganesana M, Lee ST, Wang Y, Venton BJ. Analytical Techniques in Neuroscience: Recent Advances in Imaging, Separation, and Electrochemical Methods. Anal Chem 2017; 89:314-341. [PMID: 28105819 PMCID: PMC5260807 DOI: 10.1021/acs.analchem.6b04278] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | | | | | - B. Jill Venton
- Department of Chemistry, PO Box 400319, University of Virginia, Charlottesville, VA 22904
| |
Collapse
|
31
|
Clozapine N-Oxide Administration Produces Behavioral Effects in Long-Evans Rats: Implications for Designing DREADD Experiments. eNeuro 2016; 3:eN-NWR-0219-16. [PMID: 27822508 PMCID: PMC5089539 DOI: 10.1523/eneuro.0219-16.2016] [Citation(s) in RCA: 232] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/06/2016] [Accepted: 10/06/2016] [Indexed: 12/18/2022] Open
Abstract
Clozapine N-oxide (CNO) is a ligand for a powerful chemogenetic system that can selectively inhibit or activate neurons; the so-called Designer Receptors Exclusively Activated by Designer Drugs (DREADD) system. This system consists of synthetic G-protein-coupled receptors, which are not believed to be activated by any endogenous ligand, but are activated by the otherwise inert CNO. However, it has previously been shown that the administration of CNO in humans and rats leads to detectable levels of the bioactive compounds clozapine and N-desmethylclozapine (N-Des). As a follow-up, experiments were conducted to investigate the effects of CNO in male Long–Evans rats. It was found that 1 mg/kg CNO reduced the acoustic startle reflex but had no effect on prepulse inhibition (PPI; a measure of sensorimotor gating). CNO (2 and 5 mg/kg) had no effect on the disruption to PPI induced by the NMDA antagonist phencyclidine or the muscarinic antagonist scopolamine. In locomotor studies, CNO alone (at 1, 2, and 5 mg/kg) had no effect on spontaneous locomotion, but 5 mg/kg CNO pretreatment significantly attenuated d-amphetamine-induced hyperlocomotion. In line with the behavioral results, fast-scan cyclic voltammetry found that 5 mg/kg CNO significantly attenuated the d-amphetamine-induced increase in evoked dopamine. However, the effects seen after CNO administration cannot be definitively ascribed to CNO because biologically relevant levels of clozapine and N-Des were found in plasma after CNO injection. Our results show that CNO has multiple dose-dependent effects in vivo and is converted to clozapine and N-Des emphasizing the need for a CNO-only DREADD-free control group when designing DREADD-based experiments.
Collapse
|
32
|
Sizemore RJ, Seeger-Armbruster S, Hughes SM, Parr-Brownlie LC. Viral vector-based tools advance knowledge of basal ganglia anatomy and physiology. J Neurophysiol 2016; 115:2124-46. [PMID: 26888111 PMCID: PMC4869490 DOI: 10.1152/jn.01131.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/16/2016] [Indexed: 01/07/2023] Open
Abstract
Viral vectors were originally developed to deliver genes into host cells for therapeutic potential. However, viral vector use in neuroscience research has increased because they enhance interpretation of the anatomy and physiology of brain circuits compared with conventional tract tracing or electrical stimulation techniques. Viral vectors enable neuronal or glial subpopulations to be labeled or stimulated, which can be spatially restricted to a single target nucleus or pathway. Here we review the use of viral vectors to examine the structure and function of motor and limbic basal ganglia (BG) networks in normal and pathological states. We outline the use of viral vectors, particularly lentivirus and adeno-associated virus, in circuit tracing, optogenetic stimulation, and designer drug stimulation experiments. Key studies that have used viral vectors to trace and image pathways and connectivity at gross or ultrastructural levels are reviewed. We explain how optogenetic stimulation and designer drugs used to modulate a distinct pathway and neuronal subpopulation have enhanced our mechanistic understanding of BG function in health and pathophysiology in disease. Finally, we outline how viral vector technology may be applied to neurological and psychiatric conditions to offer new treatments with enhanced outcomes for patients.
Collapse
Affiliation(s)
- Rachel J Sizemore
- Department of Anatomy, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Sonja Seeger-Armbruster
- Department of Physiology, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand; and
| | - Stephanie M Hughes
- Department of Biochemistry, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Louise C Parr-Brownlie
- Department of Anatomy, Otago School of Medical Sciences, Brain Health Research Centre, Brain Research New Zealand, University of Otago, Dunedin, New Zealand;
| |
Collapse
|
33
|
Smith KS, Bucci DJ, Luikart BW, Mahler SV. DREADDS: Use and application in behavioral neuroscience. Behav Neurosci 2016; 130:137-55. [PMID: 26913540 DOI: 10.1037/bne0000135] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Technological advances over the last decade are changing the face of behavioral neuroscience research. Here we review recent work on the use of one such transformative tool in behavioral neuroscience research, chemogenetics (or Designer Receptors Exclusively Activated by Designer Drugs, DREADDS). As transformative technologies such as DREADDs are introduced, applied, and refined, their utility in addressing complex questions about behavior and cognition becomes clear and exciting. In the behavioral neuroscience field, remarkable new findings now regularly appear as a result of the ability to monitor and intervene in neural processes with high anatomical precision as animals behave in complex task environments. As these new tools are applied to behavioral questions, individualized procedures for their use find their way into diverse labs. Thus, "tips of the trade" become important for wide dissemination not only for laboratories that are using the tools but also for those who are interested in incorporating them into their own work. Our aim is to provide an up-to-date perspective on how the DREADD technique is being used for research on learning and memory, decision making, and goal-directed behavior, as well as to provide suggestions and considerations for current and future users based on our collective experience. (PsycINFO Database Record
Collapse
Affiliation(s)
- Kyle S Smith
- Department of Psychological and Brain Sciences, Dartmouth College
| | - David J Bucci
- Department of Psychological and Brain Sciences, Dartmouth College
| | - Bryan W Luikart
- Department of Physiology and Neurobiology, Geisel School of Medicine, Dartmouth College
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California Irvine
| |
Collapse
|
34
|
Chang L, Hu J, Chen F, Chen Z, Shi J, Yang Z, Li Y, Lee LJ. Nanoscale bio-platforms for living cell interrogation: current status and future perspectives. NANOSCALE 2016; 8:3181-3206. [PMID: 26745513 DOI: 10.1039/c5nr06694h] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The living cell is a complex entity that dynamically responds to both intracellular and extracellular environments. Extensive efforts have been devoted to the understanding intracellular functions orchestrated with mRNAs and proteins in investigation of the fate of a single-cell, including proliferation, apoptosis, motility, differentiation and mutations. The rapid development of modern cellular analysis techniques (e.g. PCR, western blotting, immunochemistry, etc.) offers new opportunities in quantitative analysis of RNA/protein expression up to a single cell level. The recent entries of nanoscale platforms that include kinds of methodologies with high spatial and temporal resolution have been widely employed to probe the living cells. In this tutorial review paper, we give insight into background introduction and technical innovation of currently reported nanoscale platforms for living cell interrogation. These highlighted technologies are documented in details within four categories, including nano-biosensors for label-free detection of living cells, nanodevices for living cell probing by intracellular marker delivery, high-throughput platforms towards clinical current, and the progress of microscopic imaging platforms for cell/tissue tracking in vitro and in vivo. Perspectives for system improvement were also discussed to solve the limitations remains in current techniques, for the purpose of clinical use in future.
Collapse
Affiliation(s)
- Lingqian Chang
- NSF Nanoscale Science and Engineering Center (NSEC), The Ohio State University, Columbus, OH 43212, USA.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Icick R, Peoc'h K, Karsinti E, Ksouda K, Hajj A, Bloch V, Prince N, Mouly S, Bellivier F, Lépine JP, Laplanche JL, Vorspan F. A cannabinoid receptor 1 polymorphism is protective against major depressive disorder in methadone-maintained outpatients. Am J Addict 2015; 24:613-20. [PMID: 26331953 DOI: 10.1111/ajad.12273] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 07/23/2015] [Accepted: 08/08/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND OBJECTIVES The burden of opiate dependence not only relies on somatic complications such as infectious diseases or acute intoxication but also on frequent psychiatric events such as major depressive disorder (MDD) and suicidal behavior (SB). Given the preclinical and clinical evidence regarding the associations between cannabinoid systems and both opiate dependence and psychiatric disorders, we chose to address whether one single nucleotide polymorphism (SNP) of the cannabinoid receptor type 1 gene (CNR1) named rs2023239 would be associated with lifetime MDD and SB in a population of opiate-dependent outpatients remitted under stable methadone treatment. METHODS Sociodemographic and clinical data were included as independent factors in two logistic regression models aimed at predicting SB and MDD, respectively, performed with 85 Caucasian individuals. RESULTS The minor C allele of rs2023239 showed an independent protective effect against lifetime MDD after adjustment for potential confounders. It was not associated with variables related to suicidal behavior. CONCLUSIONS AND SCIENTIFIC SIGNIFICANCE Despite limitations due to the modest sample size, our results are consistent with previous research on the endocannabinoid system and suggest new leads for detecting subjects at risk of MDD, which remains insufficiently diagnosed and treated in patients suffering from severe addictive disorders.
Collapse
Affiliation(s)
- Romain Icick
- Department of Psychiatry and Addiction Medicine, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Inserm UMR-S1144, Paris, France.,Paris Descartes University, Paris, France
| | - Katell Peoc'h
- Inserm UMR-S1144, Paris, France.,Paris Descartes University, Paris, France.,Paris Diderot University, Paris, France.,Department of Biochemistry and Molecular Biology, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, AP-HP, Paris, France
| | - Emily Karsinti
- Department of Psychiatry and Addiction Medicine, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Inserm UMR-S1144, Paris, France
| | | | - Aline Hajj
- Inserm UMR-S1144, Paris, France.,Paris Descartes University, Paris, France
| | - Vanessa Bloch
- Inserm UMR-S1144, Paris, France.,Hospital Pharmacy, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, AP-HP, Paris, France
| | - Nathalie Prince
- Inserm UMR-S1144, Paris, France.,Department of Biochemistry and Molecular Biology, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, AP-HP, Paris, France
| | - Stéphane Mouly
- Inserm UMR-S1144, Paris, France.,Paris Descartes University, Paris, France.,Paris Diderot University, Paris, France.,Department of Internal Medicine, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Frank Bellivier
- Department of Psychiatry and Addiction Medicine, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Inserm UMR-S1144, Paris, France.,Paris Descartes University, Paris, France.,Paris Diderot University, Paris, France
| | - Jean-Pierre Lépine
- Department of Psychiatry and Addiction Medicine, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Inserm UMR-S1144, Paris, France.,Paris Descartes University, Paris, France.,Paris Diderot University, Paris, France
| | - Jean-Louis Laplanche
- Inserm UMR-S1144, Paris, France.,Paris Descartes University, Paris, France.,Paris Diderot University, Paris, France.,Department of Biochemistry and Molecular Biology, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, AP-HP, Paris, France
| | - Florence Vorspan
- Department of Psychiatry and Addiction Medicine, Groupe Hospitalier Saint-Louis-Lariboisière-Fernand Widal, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.,Inserm UMR-S1144, Paris, France.,Paris Descartes University, Paris, France.,Paris Diderot University, Paris, France
| |
Collapse
|
36
|
Deconstructing 5-HT6 receptor effects on striatal circuit function. Neuroscience 2015; 299:97-106. [PMID: 25934037 DOI: 10.1016/j.neuroscience.2015.04.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/21/2015] [Indexed: 11/23/2022]
Abstract
Medium spiny neurons (MSNs) constitute 95% of neurons in the dorsal striatum subdivided into direct (striatonigral) and indirect (striatopallidal) pathways. Whereas D1 and D2 receptors and several neuropeptides, including dynorphin and enkephalin, are differentially expressed in these neurons, 5-hydroxytryptamine 6 receptors (5-HT6) are expressed in both pathways. Previous results demonstrate that concurrent 5-HT6 receptor overexpression in MSNs of both pathways in the dorsomedial striatum (DMS) interferes with instrumental learning and that 5-HT6 overexpression in the dorsolateral striatum (DLS) relieves rats from inflexible habitual behaviors. We hypothesized that 5-HT6 receptor-mediated co-activation of both pathways interferes with the differential activation/inhibition of direct/indirect pathways by dopamine. To test this idea, we cloned novel viral vectors to selectively overexpress 5-HT6 receptors in direct or indirect pathway MSNs to deconstruct their role in modulating instrumental learning and habitual responding. We found that increasing 5-HT6 receptor expression in either direct or indirect pathway MSNs of the posterior DMS selectively enhanced or impaired initial acquisition of a discrete instrumental learning task respectively, though all rats were ultimately able to learn the task. In a separate set of experiments, 5-HT6 receptor overexpression in indirect pathway MSNs of the DLS facilitated behavioral flexibility in rats overtrained on a repetitive pressing task using a variable interval schedule of reinforcement, during an omission contingency training session and subsequent probe testing. Together these findings further the notion that 5-HT6 signaling causes balanced activation of opposing MSN pathways by serotonin in sub-regions of the dorsal striatum allowing for more reflective modalities of behavior.
Collapse
|
37
|
Epigenetic basis of opiate suppression of Bdnf gene expression in the ventral tegmental area. Nat Neurosci 2015; 18:415-22. [PMID: 25643298 PMCID: PMC4340719 DOI: 10.1038/nn.3932] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a crucial role in modulating neural and behavioral plasticity to drugs of abuse. Here, we demonstrate a persistent down-regulation of exon-specific Bdnf expression in the ventral tegmental area (VTA) in response to chronic opiate exposure, which is mediated by specific epigenetic modifications at the corresponding Bdnf gene promoters. Exposure to chronic morphine increases stalling of RNA polymerase II at these Bdnf promoters in VTA and alters permissive and repressive histone modifications and occupancy of their regulatory proteins at the specific promoters. Furthermore, we show that morphine suppresses binding of phospho-CREB (cAMP response element binding protein) to Bdnf promoters in VTA, which results from enrichment of trimethylated H3K27 at the promoters, and that decreased NURR1 (nuclear receptor related-1) expression also contributes to Bdnf repression and associated behavioral plasticity to morphine. These studies reveal novel epigenetic mechanisms of morphine-induced molecular and behavioral neuroadaptations.
Collapse
|
38
|
DREAMM: a biobehavioral imaging methodology for dynamic in vivo whole-brain mapping of cell type-specific functional networks. Neuropsychopharmacology 2015; 40:239-40. [PMID: 25482170 PMCID: PMC4262913 DOI: 10.1038/npp.2014.233] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
39
|
Abstract
This paper is the thirty-sixth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2013 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia; stress and social status; tolerance and dependence; learning and memory; eating and drinking; alcohol and drugs of abuse; sexual activity and hormones, pregnancy, development and endocrinology; mental illness and mood; seizures and neurologic disorders; electrical-related activity and neurophysiology; general activity and locomotion; gastrointestinal, renal and hepatic functions; cardiovascular responses; respiration and thermoregulation; and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
40
|
Antidepressant-like effects of buprenorphine in rats are strain dependent. Behav Brain Res 2014; 278:385-92. [PMID: 25453747 DOI: 10.1016/j.bbr.2014.10.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/06/2014] [Accepted: 10/11/2014] [Indexed: 01/01/2023]
Abstract
The prevalence of major depressive disorder and the limited efficacy of conventional drug treatments provide significant impetus to develop novel and more rapidly acting antidepressants for individuals with treatment resistant forms of depression. The primary goal of these studies was to ascertain whether buprenorphine (BPN), a medically available drug with mixed effects at opioid receptors, was effective in behavioral tests using the Wistar Kyoto (WKY) rat strain, a rodent model of exaggerated depressive and anxiety behaviors that demonstrates resistance to certain antidepressants. As WKY rats are maintained by different sources, we assessed the behavioral effects of BPN using the modified rat forced swim test (FST) and the emergence test in WKY rat colonies obtained from different vendors. BPN dose-dependently reduced immobility and increased swimming behavior in the FST and reduced emergence latencies in two WKY lines (Charles River (WKY/NCrl) and Harlan laboratories (WKY/NHsd)) that also showed high baseline immobility in the FST. WKY rats from Taconic (WKY/NTac) did not show high baseline immobility in the FST or anxiety as had been previously reported, suggesting a drift in the phenotype of rats from this supplier. Furthermore, BPN did not reduce immobility in the FST or reduce latencies in the emergence test in WKY rats from Taconic. BPN also failed to produce antidepressant-like effects in Wistar and Sprague-Dawley rats. These results indicate a striking strain-selectivity for the effects of BPN, producing antidepressant and anxiolytic-like responses in WKY/NCrl and WKY/NHsd lines but not in the normosensitive control Wistar and Sprague-Dawley strains.
Collapse
|
41
|
Urban DJ, Roth BL. DREADDs (designer receptors exclusively activated by designer drugs): chemogenetic tools with therapeutic utility. Annu Rev Pharmacol Toxicol 2014; 55:399-417. [PMID: 25292433 DOI: 10.1146/annurev-pharmtox-010814-124803] [Citation(s) in RCA: 483] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the past decade, emerging synthetic biology technologies such as chemogenetics have dramatically transformed how pharmacologists and systems biologists deconstruct the involvement of G protein-coupled receptors (GPCRs) in a myriad of physiological and translational settings. Here we highlight a specific chemogenetic application that extends the utility of the concept of RASSLs (receptors activated solely by synthetic ligands): We have dubbed it DREADDs (designer receptors exclusively activated by designer drugs). As we show in this review, DREADDs are now used ubiquitously to modulate GPCR activity noninvasively in vivo. Results from these studies have directly implicated GPCR signaling in a large number of therapeutically relevant contexts. We also highlight recent applications of DREADD technology that have illuminated GPCR signaling processes that control pathways relevant to the treatment of eating disorders, obesity, and obesity-associated metabolic abnormalities. Additionally, we provide an overview of the potential utility of chemogenetic technologies for transformative therapeutics.
Collapse
|
42
|
Abstract
Drug addiction is characterized by compulsive drug-seeking and drug-taking, and a high propensity for relapse. Although the brain regions involved in regulating addiction processes have long been identified, the ways in which individual cell types govern addiction behaviors remain elusive. New technologies for modulating the activity of defined cell types have recently emerged that are allowing us to address these important questions. Here, we review how one such technology, DREADDs (Designer Receptors Exclusively Activated by Designer Drugs), can be used to refine our knowledge of addiction circuitry. These engineered receptors modulate cellular activity by acting on G protein coupled signaling cascades and in this review we pay particular attention to how this slower-onset modulation preferentially regulates behaviors that develop over time.
Collapse
Affiliation(s)
- Susan M Ferguson
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, United States; Department of Psychiatry and Behavioral Sciences, Seattle, WA, United States
| | - John F Neumaier
- Department of Psychiatry and Behavioral Sciences, Seattle, WA, United States; Department of Pharmacology, University of Washington, Seattle, WA, United States
| |
Collapse
|