1
|
Chmiel J, Stępień-Słodkowska M, Ramik-Mażewska I. Efficacy of Transcranial Direct Current Stimulation (tDCS) on Neuropsychiatric Symptoms in Substance Use Disorder (SUD)-A Review and Insights into Possible Mechanisms of Action. J Clin Med 2025; 14:1337. [PMID: 40004867 PMCID: PMC11856849 DOI: 10.3390/jcm14041337] [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: 01/23/2025] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Introduction: Substance use disorder (SUD) is a significant global clinical issue marked by the excessive consumption of alcohol, nicotine, and various psychoactive substances, leading to impaired social, cognitive, and occupational functioning. Individuals with SUD frequently experience depression and anxiety disorders, which exacerbate their prognosis and contribute to substantial health and social burdens. The pathophysiology of SUD and its associated conditions is multifaceted, involving multiple dysfunctions in the brain. This complexity underscores an urgent need for the development of noninvasive treatments that can directly target the brain. One of them is transcranial direct current stimulation (tDCS), an intensively studied technique for safely modulating cortical excitability. The aim of this study is to investigate the effectiveness of tDCS in treating symptoms of depression and anxiety in SUD. Methods: With an emphasis on the underlying mechanisms of action, this mechanistic review investigates the effectiveness of tDCS in treating anxiety and depression in SUD patients. Literature searches were conducted using the PubMed/Medline, ResearchGate, Cochrane, and Google Scholar databases. Results: The review identified 12 relevant studies. The results showed that left dorsolateral prefrontal cortex (DLPFC) stimulation is an effective treatment option for depression in SUD. In anxiety disorders, left and right DLPFC stimulation is effective, with better results observed with right DLPFC stimulation. However, the included studies differed in their methodology, sample characteristics, and measurement methods, which could have influenced the final results of the analysis. The central focus of this mechanistic review is to discuss the potential mechanisms of action of tDCS in treating depression and anxiety in SUD. These mechanisms include the modulation of brain networks, a reduction in neuroinflammation, an enhancement in neuroplasticity, and an increase in P300 amplitude. We also discuss the limitations of the included studies and propose ways to address them in future research. Conclusions: This review provides evidence that tDCS is an effective treatment option for anxiety and depression in SUD. Stimulation of the left DLPFC reduces symptoms of depression, while stimulation of the right DLPFC reduces symptoms of anxiety. However, future research is required to confirm these findings and to deepen our understanding of the mechanisms through which tDCS exerts its effects in this context. Neuroimaging methods (fMRI and EEG) and blood tests could be particularly useful.
Collapse
Affiliation(s)
- James Chmiel
- Institute of Physical Culture Sciences, Faculty of Physical Culture and Health, University of Szczecin, Al. Piastów 40B, Block 6, 71-065 Szczecin, Poland
- Doctoral School, University of Szczecin, Mickiewicza 16, 70-384 Szczecin, Poland
| | - Marta Stępień-Słodkowska
- Institute of Physical Culture Sciences, Faculty of Physical Culture and Health, University of Szczecin, Al. Piastów 40B, Block 6, 71-065 Szczecin, Poland
| | - Irena Ramik-Mażewska
- Institute of Pedagogy, University of Szczecin, ul. Ogińskiego 16/17, 71-415 Szczecin, Poland
| |
Collapse
|
2
|
Tian X, Russo SJ, Li L. Behavioral Animal Models and Neural-Circuit Framework of Depressive Disorder. Neurosci Bull 2025; 41:272-288. [PMID: 39120643 PMCID: PMC11794861 DOI: 10.1007/s12264-024-01270-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/26/2024] [Indexed: 08/10/2024] Open
Abstract
Depressive disorder is a chronic, recurring, and potentially life-endangering neuropsychiatric disease. According to a report by the World Health Organization, the global population suffering from depression is experiencing a significant annual increase. Despite its prevalence and considerable impact on people, little is known about its pathogenesis. One major reason is the scarcity of reliable animal models due to the absence of consensus on the pathology and etiology of depression. Furthermore, the neural circuit mechanism of depression induced by various factors is particularly complex. Considering the variability in depressive behavior patterns and neurobiological mechanisms among different animal models of depression, a comparison between the neural circuits of depression induced by various factors is essential for its treatment. In this review, we mainly summarize the most widely used behavioral animal models and neural circuits under different triggers of depression, aiming to provide a theoretical basis for depression prevention.
Collapse
Affiliation(s)
- Xiangyun Tian
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of the Chinese Academy of Sciences, Beijing, 100049, China
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Long Li
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of the Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
3
|
Chen CY, Wang YF, Lei L, Zhang Y. MicroRNA-specific targets for neuronal plasticity, neurotransmitters, neurotrophic factors, and gut microbes in the pathogenesis and therapeutics of depression. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111186. [PMID: 39521033 DOI: 10.1016/j.pnpbp.2024.111186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/11/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Depression is of great concern because of the huge burden, and it is impacted by various epigenetic modifications, e.g., histone modification, covalent modifications in DNA, and silencing mechanisms of non-coding protein genes, e.g., microRNAs (miRNAs). MiRNAs are a class of endogenous non-coding RNAs. Alternations in specific miRNAs have been observed both in depressive patients and experimental animals. Also, miRNAs are highly expressed in the central nervous system and can be delivered to different tissues via tissue-specific exosomes. However, the mechanism of miRNAs' involvement in the pathological process of depression is not well understood. Therefore, we summarized and discussed the role of miRNAs in depression. Conclusively, miRNAs are involved in the pathology of depression by causing structural and functional changes in synapses, mediating neuronal regeneration, differentiation, and apoptosis, regulating the gut microbes and the expression of various neurotransmitters and BDNF, and mediating inflammatory and immune responses. Moreover, miRNAs can predict the efficacy of antidepressant medications and explain the mechanism of action of antidepressant drugs and aerobic exercise to prevent and assist in treating depression.
Collapse
Affiliation(s)
- Cong-Ya Chen
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yu-Fei Wang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lan Lei
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yi Zhang
- Department of Anatomy, School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
4
|
Paoletti I, Coccurello R. Irisin: A Multifaceted Hormone Bridging Exercise and Disease Pathophysiology. Int J Mol Sci 2024; 25:13480. [PMID: 39769243 PMCID: PMC11676223 DOI: 10.3390/ijms252413480] [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: 11/27/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/03/2025] Open
Abstract
The fibronectin domain-containing protein 5 (FNDC5), or irisin, is an adipo-myokine hormone produced during exercise, which shows therapeutic potential for conditions like metabolic disorders, osteoporosis, sarcopenia, obesity, type 2 diabetes, and neurodegenerative diseases, including Alzheimer's disease (AD). This review explores its potential across various pathophysiological processes that are often considered independent. Elevated in healthy states but reduced in diseases, irisin improves muscle-adipose communication, insulin sensitivity, and metabolic balance by enhancing mitochondrial function and reducing oxidative stress. It promotes osteogenesis and mitigates bone loss in osteoporosis and sarcopenia. Irisin exhibits anti-inflammatory effects by inhibiting NF-κB signaling and countering insulin resistance. In the brain, it reduces amyloid-β toxicity, inflammation, and oxidative stress, enhancing brain-derived neurotrophic factor (BDNF) signaling, which improves cognition and synaptic health in AD models. It also regulates dopamine pathways, potentially alleviating neuropsychiatric symptoms like depression and apathy. By linking physical activity to systemic health, irisin emphasizes its role in the muscle-bone-brain axis. Its multifaceted benefits highlight its potential as a therapeutic target for AD and related disorders, with applications in prevention, in treatment, and as a complement to exercise strategies.
Collapse
Affiliation(s)
- Ilaria Paoletti
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
| | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, European Center for Brain Research, 00143 Rome, Italy;
- Institute for Complex Systems (ISC), National Research Council (C.N.R.), 00185 Rome, Italy
| |
Collapse
|
5
|
Schmidt ME, Kezic I, Popova V, Melkote R, Van Der Ark P, Pemberton DJ, Mareels G, Canuso CM, Fava M, Drevets WC. Efficacy and safety of aticaprant, a kappa receptor antagonist, adjunctive to oral SSRI/SNRI antidepressant in major depressive disorder: results of a phase 2 randomized, double-blind, placebo-controlled study. Neuropsychopharmacology 2024; 49:1437-1447. [PMID: 38649428 PMCID: PMC11251157 DOI: 10.1038/s41386-024-01862-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024]
Abstract
This was a double-blind, randomized, phase 2 study of adults (18-64 years) with DSM-5 diagnosis of major depressive disorder (MDD), with moderate-to-severe episode severity (Montgomery-Åsberg Depression Rating Scale [MADRS] ≥25) despite an adequate course with ongoing antidepressant for ≥6 weeks to ≤12 months. Following a double-blind placebo lead-in period (up to 3 weeks), participants were randomized to receive once daily aticaprant 10 mg or continue placebo, added to their ongoing treatment, for 6 weeks. Of 184 participants enrolled, 169 were included in safety analyses (aticaprant n = 85, placebo n = 84) and 166 in full intent-to-treat (fITT) efficacy analyses; 121 placebo lead-in non-responders (<30% reduction in MADRS total score) in fITT were included in enriched ITT (eITT) analyses. Improvement (least squares mean difference [upper limit 1-sided 80% CI] versus placebo) in MADRS total score at week 6 for aticaprant was significant versus placebo (eITT: -2.1 [-1.09], 1-sided p = 0.044; effect size (ES) 0.23; fITT -3.1 [2.21], 1-sided p = 0.002; ES 0.36). The between-group difference was larger among participants with Snaith-Hamilton Pleasure Scale (SHAPS) score greater/equal to versus less than baseline median SHAPS. The most common treatment-emergent adverse events reported for aticaprant (versus placebo) were headache (11.8% versus 7.1%), diarrhea (8.2% versus 2.4%), nasopharyngitis (5.9% versus 2.4%), and pruritus (5.9% versus 0%). One participant (1.2%) in each arm discontinued treatment due to an adverse event. In this study of participants with MDD and inadequate response to SSRI/SNRI, adjunctive treatment with aticaprant significantly reduced depressive symptoms versus placebo, without resulting in significant safety signals, supporting further investigation in larger trials.
Collapse
Affiliation(s)
| | - Iva Kezic
- Janssen Research & Development, Beerse, Belgium
| | | | - Rama Melkote
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | | | | | - Guy Mareels
- Janssen Research & Development, Beerse, Belgium
| | - Carla M Canuso
- Janssen Research & Development, LLC, Titusville, NJ, USA
| | - Maurizio Fava
- Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
6
|
Taraku B, Loureiro JR, Sahib AK, Zavaliangos‐Petropulu A, Al‐Sharif N, Leaver AM, Wade B, Joshi S, Woods RP, Espinoza R, Narr KL. Modulation of habenular and nucleus accumbens functional connectivity by ketamine in major depression. Brain Behav 2024; 14:e3511. [PMID: 38894648 PMCID: PMC11187958 DOI: 10.1002/brb3.3511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/09/2024] [Accepted: 04/13/2024] [Indexed: 06/21/2024] Open
Abstract
INTRODUCTION Major depressive disorder (MDD) is associated with dysfunctional reward processing, which involves functional circuitry of the habenula (Hb) and nucleus accumbens (NAc). Since ketamine elicits rapid antidepressant and antianhedonic effects in MDD, this study sought to investigate how serial ketamine infusion (SKI) treatment modulates static and dynamic functional connectivity (FC) in Hb and NAc functional networks. METHODS MDD participants (n = 58, mean age = 40.7 years, female = 28) received four ketamine infusions (0.5 mg/kg) 2-3 times weekly. Resting-state functional magnetic resonance imaging (fMRI) scans and clinical assessments were collected at baseline and 24 h post-SKI. Static FC (sFC) and dynamic FC variability (dFCv) were calculated from left and right Hb and NAc seeds to all other brain regions. Changes in FC pre-to-post SKI, and correlations with changes with mood and anhedonia were examined. Comparisons of FC between patients and healthy controls (HC) at baseline (n = 55, mean age = 32.6, female = 31), and between HC assessed twice (n = 16) were conducted as follow-up analyses. RESULTS Following SKI, significant increases in left Hb-bilateral visual cortex FC, decreases in left Hb-left inferior parietal cortex FC, and decreases in left NAc-right cerebellum FC occurred. Decreased dFCv between left Hb and right precuneus and visual cortex, and decreased dFCv between right NAc and right visual cortex both significantly correlated with improvements in mood ratings. Decreased FC between left Hb and bilateral visual/parietal cortices as well as increased FC between left NAc and right visual/parietal cortices both significantly correlated with improvements in anhedonia. No differences were observed between HC at baseline or over time. CONCLUSION Subanesthetic ketamine modulates functional pathways linking the Hb and NAc with visual, parietal, and cerebellar regions in MDD. Overlapping effects between Hb and NAc functional systems were associated with ketamine's therapeutic response.
Collapse
Affiliation(s)
- Brandon Taraku
- Ahmanson‐Lovelace Brain Mapping Center, Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Joana R. Loureiro
- Ahmanson‐Lovelace Brain Mapping Center, Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Ashish K. Sahib
- Ahmanson‐Lovelace Brain Mapping Center, Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Artemis Zavaliangos‐Petropulu
- Ahmanson‐Lovelace Brain Mapping Center, Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Noor Al‐Sharif
- Ahmanson‐Lovelace Brain Mapping Center, Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Amber M. Leaver
- Department of RadiologyNorthwestern UniversityChicagoIllinoisUSA
| | - Benjamin Wade
- Division of Neuropsychiatry and NeuromodulationMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Shantanu Joshi
- Ahmanson‐Lovelace Brain Mapping Center, Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
- Department of Psychiatry and Biobehavioral SciencesUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Roger P. Woods
- Ahmanson‐Lovelace Brain Mapping Center, Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Randall Espinoza
- Department of Psychiatry and Biobehavioral SciencesUniversity of California Los AngelesLos AngelesCaliforniaUSA
| | - Katherine L. Narr
- Ahmanson‐Lovelace Brain Mapping Center, Department of NeurologyUniversity of California Los AngelesLos AngelesCaliforniaUSA
- Department of Psychiatry and Biobehavioral SciencesUniversity of California Los AngelesLos AngelesCaliforniaUSA
| |
Collapse
|
7
|
Spreen A, Alkhoury D, Walter H, Müller S. Optogenetic behavioral studies in depression research: A systematic review. iScience 2024; 27:109776. [PMID: 38726370 PMCID: PMC11079475 DOI: 10.1016/j.isci.2024.109776] [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: 07/27/2023] [Revised: 10/21/2023] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Optogenetics has made substantial contributions to our understanding of the mechanistic underpinnings of depression. This systematic review employs quantitative analysis to investigate the impact of optogenetic stimulation in mice and rats on behavioral alterations in social interaction, sucrose consumption, and mobility. The review analyses optogenetic behavioral studies using standardized behavioral tests to detect behavioral changes induced via optogenetic stimulation in stressed or stress-naive mice and rats. Behavioral changes were evaluated as either positive, negative, or not effective. The analysis comprises the outcomes of 248 behavioral tests of 168 studies described in 37 articles, including negative and null results. Test outcomes were compared for each behavior, depending on the animal cohort, applied type of stimulation and the stimulated neuronal circuit and cell type. The presented synthesis contributes toward a comprehensive picture of optogenetic behavioral research in the context of depression.
Collapse
Affiliation(s)
- Anika Spreen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
- Experimental Biophysics, Institute for Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dana Alkhoury
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| | - Henrik Walter
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| | - Sabine Müller
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Psychiatry and Neurosciences, CCM, Berlin, Germany
| |
Collapse
|
8
|
Abdelkawy YS, Elharoun M, Sheta E, Abdel-Raheem IT, Nematalla HA. Liraglutide and Naringenin relieve depressive symptoms in mice by enhancing Neurogenesis and reducing inflammation. Eur J Pharmacol 2024; 971:176525. [PMID: 38561101 DOI: 10.1016/j.ejphar.2024.176525] [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: 12/30/2023] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Depression is a debilitating mental disease that negatively impacts individuals' lives and society. Novel hypotheses have been recently proposed to improve our understanding of depression pathogenesis. Impaired neuroplasticity and upregulated neuro-inflammation add-on to the disturbance in monoamine neurotransmitters and therefore require novel anti-depressants to target them simultaneously. Recent reports demonstrate the antidepressant effect of the anti-diabetic drug liraglutide. Similarly, the natural flavonoid naringenin has shown both anti-diabetic and anti-depressant effects. However, the neuro-pharmacological mechanisms underlying their actions remain understudied. The study aims to evaluate the antidepressant effects and neuroprotective mechanisms of liraglutide, naringenin or a combination of both. Depression was induced in mice by administering dexamethasone (32 mcg/kg) for seven consecutive days. Liraglutide (200 mcg/kg), naringenin (50 mg/kg) and a combination of both were administered either simultaneously or after induction of depression for twenty-eight days. Behavioral and molecular assays were used to assess the progression of depressive symptoms and biomarkers. Liraglutide and naringenin alone or in combination alleviated the depressive behavior in mice, manifested by decrease in anxiety, anhedonia, and despair. Mechanistically, liraglutide and naringenin improved neurogenesis, decreased neuroinflammation and comparably restored the monoamines levels to that of the reference drug escitalopram. The drugs protected mice from developing depression when given simultaneously with dexamethasone. Collectively, the results highlight the usability of liraglutide and naringenin in the treatment of depression in mice and emphasize the different pathways that contribute to the pathogenesis of depression.
Collapse
Affiliation(s)
- Yara S Abdelkawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22514, Egypt
| | - Mona Elharoun
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22514, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria 21131, Egypt
| | - Ihab Talat Abdel-Raheem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22514, Egypt
| | - Hisham A Nematalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Damanhour University, Damanhour 22514, Egypt.
| |
Collapse
|
9
|
Desai S, Zundel CG, Evanski JM, Gowatch LC, Bhogal A, Ely S, Carpenter C, Shampine M, O'Mara E, Rabinak CA, Marusak HA. Genetic variation in endocannabinoid signaling: Anxiety, depression, and threat- and reward-related brain functioning during the transition into adolescence. Behav Brain Res 2024; 463:114925. [PMID: 38423255 PMCID: PMC10977105 DOI: 10.1016/j.bbr.2024.114925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/21/2024] [Accepted: 02/26/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND The endocannabinoid system modulates neural activity throughout the lifespan. In adults, neuroimaging studies link a common genetic variant in fatty acid amide hydrolase (FAAH C385A)-an enzyme that regulates endocannabinoid signaling-to reduced risk of anxiety and depression, and altered threat- and reward-related neural activity. However, limited research has investigated these associations during the transition into adolescence, a period of substantial neurodevelopment and increased psychopathology risk. METHODS This study included FAAH genotype and longitudinal neuroimaging and neurobehavioral data from 4811 youth (46% female; 9-11 years at Baseline, 11-13 years at Year 2) from the Adolescent Brain Cognitive DevelopmentSM Study. Linear mixed models examined the effects of FAAH and the FAAH x time interaction on anxiety and depressive symptoms, amygdala reactivity to threatening faces, and nucleus accumbens (NAcc) response to happy faces during the emotional n-back task. RESULTS A significant main effect of FAAH on depressive symptoms was observed, such that depressive symptoms were lower across both timepoints in those with the AA genotype compared to both AC and CC genotypes (p's<0.05). There were no significant FAAH x time interactions for anxiety, depression, or neural responses (p's>0.05). Additionally, there were no main effects of FAAH on anxiety or neural responses (p's>0.05). CONCLUSIONS Our findings add to emerging evidence linking the FAAH C385A variant to lower risk of psychopathology, and extend these findings to a developmental sample. In particular, we found lower depressive symptoms in FAAH AA genotypes compared to AC and CC genotypes. Future research is needed to characterize the role of the FAAH variant and the eCB system more broadly in neurodevelopment and psychiatric risk.
Collapse
Affiliation(s)
- Shreya Desai
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - Clara G Zundel
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - Julia M Evanski
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - Leah C Gowatch
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - Amanpreet Bhogal
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - Samantha Ely
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - Carmen Carpenter
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - MacKenna Shampine
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - Emilie O'Mara
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA
| | - Christine A Rabinak
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA; Dept. of Pharmacy Practice, Wayne State University, USA
| | - Hilary A Marusak
- Dept. of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, USA; Dept. of Pharmacology, Wayne State University School of Medicine, USA; Merrill Palmer Skillman Institute for Child and Family Development, Wayne State University, USA.
| |
Collapse
|
10
|
Nakayama K, Nemoto K, Arai T. Nucleus accumbens degeneration in spinocerebellar ataxia type 2: a preliminary study. Psychogeriatrics 2024; 24:345-354. [PMID: 38243757 DOI: 10.1111/psyg.13080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/16/2023] [Accepted: 01/06/2024] [Indexed: 01/21/2024]
Abstract
BACKGROUND Spinocerebellar ataxia type 2 (SCA2) exhibits mainly cerebellar and oculomotor dysfunctions but also, frequently, cognitive impairment and neuropsychological symptoms. The mechanism of the progression of SCA2 remains unclear. This study aimed to evaluate longitudinal structural changes in the brains of SCA2 patients based on atrophy rate. METHODS The OpenNeuro Dataset ds001378 was used. It comprises the demographic data and two magnetic resonance images each of nine SCA2 patients and 16 healthy controls. All structural images were preprocessed using FreeSurfer software, and each region's bilateral volume was summed. Atrophy rates were calculated based on the concept of symmetrised percent change and compared between SCA2 patients and healthy controls using non-parametric statistics. As post hoc analysis, correlation analysis was performed between infratentorial volume ratio and the accumbens area atrophy rates in SCA2 patients. RESULTS There were no significant differences between groups for age, gender, and the time between scans. Statistical analysis indicated a significantly larger atrophy rate of the accumbens area in SCA2 patients than in controls. Additionally, the infratentorial volume ratio and accumbens area atrophy rates showed moderate negative correlation. CONCLUSIONS This study found that nucleus accumbens (NAc) atrophy was significantly accelerated in SCA2 patients. Anatomically, the NAc is densely connected with infratentorial brain regions, so it is reasonable to posit that degeneration propagates from the cerebellum and brainstem to the NAc and other supratentorial areas. Functionally, the NAc is essential for appropriate behaviour, so NAc degeneration might contribute to neuropsychological symptoms in SCA2 patients.
Collapse
Affiliation(s)
- Kenjiro Nakayama
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, Japan
| | - Kiyotaka Nemoto
- Department of Psychiatry, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tetsuaki Arai
- Department of Psychiatry, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
11
|
Asir B, Boscutti A, Fenoy AJ, Quevedo J. Deep Brain Stimulation (DBS) in Treatment-Resistant Depression (TRD): Hope and Concern. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1456:161-186. [PMID: 39261429 DOI: 10.1007/978-981-97-4402-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
In this chapter, we explore the historical evolution, current applications, and future directions of Deep Brain Stimulation (DBS) for Treatment-Resistant Depression (TRD). We begin by highlighting the early efforts of neurologists and neurosurgeons who laid the foundations for today's DBS techniques, moving from controversial lobotomies to the precision of stereotactic surgery. We focus on the advent of DBS, emphasizing its emergence as a significant breakthrough for movement disorders and its extension to psychiatric conditions, including TRD. We provide an overview of the neural networks implicated in depression, detailing the rationale for the choice of common DBS targets. We also cover the technical aspects of DBS, from electrode placement to programming and parameter selection. We then critically review the evidence from clinical trials and open-label studies, acknowledging the mixed outcomes and the challenges posed by placebo effects and trial design. Safety and ethical considerations are also discussed. Finally, we explore innovative directions for DBS research, including the potential of closed-loop systems, dual stimulation strategies, and noninvasive alternatives like ultrasound neuromodulation. In the last section, we outline recommendations for future DBS studies, including the use of alternative designs for placebo control, the collection of neural and behavioral recordings, and the application of machine-learning approaches.
Collapse
Affiliation(s)
- Bashar Asir
- Department of Psychiatry and Behavioral Sciences at McGovern Medical School, UTHealth Houston, Houston, TX, USA.
| | - Andrea Boscutti
- Department of Psychiatry and Behavioral Sciences at McGovern Medical School, UTHealth Houston, Houston, TX, USA
| | - Albert J Fenoy
- Department of Neurosurgery and Psychiatry, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Joao Quevedo
- Department of Psychiatry and Behavioral Sciences at McGovern Medical School, UTHealth Houston, Houston, TX, USA
| |
Collapse
|
12
|
Mulc D, Smilović D, Krsnik Ž, Junaković-Munjas A, Kopić J, Kostović I, Šimić G, Vukšić M. Fetal development of the human amygdala. J Comp Neurol 2024; 532:e25580. [PMID: 38289194 DOI: 10.1002/cne.25580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 11/03/2023] [Accepted: 12/31/2023] [Indexed: 02/01/2024]
Abstract
The intricate development of the human amygdala involves a complex interplay of diverse processes, varying in speed and duration. In humans, transient cytoarchitectural structures deliquesce, leading to the formation of functionally distinct nuclei as a result of multiple interdependent developmental events. This study compares the amygdala's cytoarchitectural development in conjunction with specific antibody reactivity for neuronal, glial, neuropil, and radial glial fibers, synaptic, extracellular matrix, and myelin components in 39 fetal human brains. We recognized that the early fetal period, as a continuation of the embryonic period, is still dominated by relatively uniform histogenetic processes. The typical appearance of ovoid cell clusters in the lateral nucleus during midfetal period is most likely associated with the cell migration and axonal growth processes in the developing human brain. Notably, synaptic markers are firstly detected in the corticomedial group of nuclei, while immunoreactivity for the panaxonal neurofilament marker SMI 312 is found dorsally. The late fetal period is characterized by a protracted migration process evidenced by the presence of doublecortin and SOX-2 immunoreactivity ventrally, in the prospective paralaminar nucleus, reinforced by vimentin immunoreactivity in the last remaining radial glial fibers. Nearing the term period, SMI 99 immunoreactivity indicates that perinatal myelination becomes prominent primarily along major axonal pathways, laying the foundation for more pronounced functional maturation. This study comprehensively elucidates the rate and sequence of maturational events in the amygdala, highlighting the key role of prenatal development in its behavioral, autonomic, and endocrine regulation, with subsequent implications for both normal functioning and psychiatric disorders.
Collapse
Affiliation(s)
- Damir Mulc
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
- Psychiatric Hospital Vrapče, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Dinko Smilović
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Željka Krsnik
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Alisa Junaković-Munjas
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Janja Kopić
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Ivica Kostović
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Goran Šimić
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| | - Mario Vukšić
- Croatian Institute for Brain Research, School of Medicine, Scientific Centre of Excellence for Basic, Clinical and Translational Neuroscience, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
13
|
Selçuk B, Aksu T, Dereli O, Adebali O. Downregulated NPAS4 in multiple brain regions is associated with major depressive disorder. Sci Rep 2023; 13:21596. [PMID: 38062059 PMCID: PMC10703936 DOI: 10.1038/s41598-023-48646-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
Major Depressive Disorder (MDD) is a commonly observed psychiatric disorder that affects more than 2% of the world population with a rising trend. However, disease-associated pathways and biomarkers are yet to be fully comprehended. In this study, we analyzed previously generated RNA-seq data across seven different brain regions from three distinct studies to identify differentially and co-expressed genes for patients with MDD. Differential gene expression (DGE) analysis revealed that NPAS4 is the only gene downregulated in three different brain regions. Furthermore, co-expressing gene modules responsible for glutamatergic signaling are negatively enriched in these regions. We used the results of both DGE and co-expression analyses to construct a novel MDD-associated pathway. In our model, we propose that disruption in glutamatergic signaling-related pathways might be associated with the downregulation of NPAS4 and many other immediate-early genes (IEGs) that control synaptic plasticity. In addition to DGE analysis, we identified the relative importance of KEGG pathways in discriminating MDD phenotype using a machine learning-based approach. We anticipate that our study will open doors to developing better therapeutic approaches targeting glutamatergic receptors in the treatment of MDD.
Collapse
Affiliation(s)
- Berkay Selçuk
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Istanbul, Turkey
| | - Tuana Aksu
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Istanbul, Turkey
| | - Onur Dereli
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Istanbul, Turkey
| | - Ogün Adebali
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, 34956, Istanbul, Turkey.
- TÜBİTAK Research Institute for Fundamental Sciences, 41470, Gebze, Turkey.
| |
Collapse
|
14
|
Taraku B, Loureiro JR, Sahib AK, Zavaliangos-Petropulu A, Al-Sharif N, Leaver A, Wade B, Joshi S, Woods RP, Espinoza R, Narr KL. Ketamine treatment modulates habenular and nucleus accumbens static and dynamic functional connectivity in major depression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.01.23299282. [PMID: 38106178 PMCID: PMC10723506 DOI: 10.1101/2023.12.01.23299282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Dysfunctional reward processing in major depressive disorder (MDD) involves functional circuitry of the habenula (Hb) and nucleus accumbens (NAc). Ketamine elicits rapid antidepressant and alleviates anhedonia in MDD. To clarify how ketamine perturbs reward circuitry in MDD, we examined how serial ketamine infusions (SKI) modulate static and dynamic functional connectivity (FC) in Hb and NAc networks. MDD participants (n=58, mean age=40.7 years, female=28) received four ketamine infusions (0.5mg/kg) 2-3 times weekly. Resting-state fMRI scans and clinical assessments were collected at baseline and 24 hours post-SKI completion. Static FC (sFC) and dynamic FC variability (dFCv) were calculated from left and right Hb and NAc seeds to all other brain regions. Paired t-tests examined changes in FC pre-to-post SKI, and correlations were used to determine relationships between FC changes with mood and anhedonia. Following SKI, significant increases in left Hb-bilateral visual cortex FC, decreases in left Hb-left inferior parietal cortex FC, and decreases in left NAc-right cerebellum FC occurred. Decreased dFCv between left Hb and right precuneus and visual cortex, and decreased dFCv between right NAc and right visual cortex both significantly correlated with improvements in Hamilton Depression Rating Scale. Decreased FC between left Hb and bilateral visual/parietal cortices as well as increased FC between left NAc and right visual/parietal cortices both significantly correlated with improvements in anhedonia. Subanesthetic ketamine modulates functional pathways linking the Hb and NAc with visual, parietal, and cerebellar regions. Overlapping effects between Hb and NAc functional systems were associated with ketamine's therapeutic response.
Collapse
Affiliation(s)
- Brandon Taraku
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Joana R Loureiro
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Ashish K Sahib
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Artemis Zavaliangos-Petropulu
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Noor Al-Sharif
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Benjamin Wade
- Division of Neuropsychiatry and Neuromodulation, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shantanu Joshi
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Roger P Woods
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Randall Espinoza
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Katherine L Narr
- Ahmanson-Lovelace Brain Mapping Center, Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Psychiatry and Biobehavioral Sciences, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
15
|
Zhang J, Lu Y, Jia M, Bai Y, Sun L, Dong Z, Tian W, Yin F, Wei S, Wang Y. Kappa opioid receptor in nucleus accumbens regulates depressive-like behaviors following prolonged morphine withdrawal in mice. iScience 2023; 26:107536. [PMID: 37636073 PMCID: PMC10448166 DOI: 10.1016/j.isci.2023.107536] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/12/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
Prolonged withdrawal from opioids leads to negative emotions. Kappa opioid receptor (KOR) plays an important role in opioid addiction and affective disorders. However, the underlying mechanism of KOR in withdrawal-related depression is still lacking. We found that escitalopram treatment had a limited effect in improving depression symptoms in heroin-dependent patients. In mice, we demonstrated prolonged (4 weeks) but not acute (24 h) withdrawal from morphine induced depressive-like behaviors. The number of c-Fos positive cells and the expression of KOR in the nucleus accumbens (NAc), were significantly increased in the prolonged morphine withdrawal mice. Conditional KOR knockdown in NAc significantly improved depressive-like behaviors. Repeated but not acute treatment with the KOR antagonist norBNI improved depressive-like behaviors and reversed PSD95, synaptophysin, p-ERK, p-CREB, and BDNF in NAc. This study demonstrated the important role of striatal KOR in morphine withdrawal-related depressive-like behaviors and offered therapeutic potential for the treatment of withdrawal-related depression.
Collapse
Affiliation(s)
- Jinyu Zhang
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Ye Lu
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Min Jia
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Yuying Bai
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Lulu Sun
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Ziqing Dong
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
- Shaanxi Belt and Road Joint Laboratory of Precision Medicine in Psychiatry, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Wenrong Tian
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Fangyuan Yin
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Shuguang Wei
- College of Forensic Science, School of Medicine, Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| | - Yunpeng Wang
- Department of Psychiatry and Center for Brain Science, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061, China
| |
Collapse
|
16
|
Rivera-Irizarry JK, Hámor PU, Rowson SA, Asfouri J, Liu D, Zallar LJ, Garcia AF, Skelly MJ, Pleil KE. Valence and salience encoding by parallel circuits from the paraventricular thalamus to the nucleus accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547570. [PMID: 37461604 PMCID: PMC10349961 DOI: 10.1101/2023.07.03.547570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
The anterior and posterior subregions of the paraventricular thalamus (aPVT and pPVT, respectively) play unique roles in learned behaviors, from fear conditioning to alcohol/drug intake, potentially through differentially organized projections to limbic brain regions including the nucleus accumbens medial shell (mNAcSh). Here, we found that the aPVT projects broadly to the mNAcSh and that the aPVT-mNAcSh circuit encodes positive valence, such that in vivo manipulations of the circuit modulated both innately programmed and learned behavioral responses to positively and negatively valenced stimuli, particularly in females. Further, the endogenous activity of aPVT presynaptic terminals in the mNAcSh was greater in response to positively than negatively valenced stimuli, and the probability of synaptic glutamate release from aPVT neurons in the mNAcSh was higher in females than males. In contrast, we found that the pPVT-mNAcSh circuit encodes stimulus salience regardless of valence. While pPVT-mNAcSh circuit inhibition suppressed behavioral responses in both sexes, circuit activation increased behavioral responses to stimuli only in males. Our results point to circuit-specific stimulus feature encoding by parallel PVT-mNAcSh circuits that have sex-dependent biases in organization and function.
Collapse
|
17
|
Varastehmoradi B, Smith KL, Müller HK, Elfving B, Sanchez C, Wegener G. Kappa opioid activation changes protein profiles in different regions of the brain relevant to depression. Eur Neuropsychopharmacol 2023; 72:9-17. [PMID: 37040689 DOI: 10.1016/j.euroneuro.2023.03.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 03/18/2023] [Accepted: 03/23/2023] [Indexed: 04/13/2023]
Abstract
Depression is a widespread disorder with a significant burden on individuals and society. There are various available treatments for patients with depression. However, not all patients respond adequately to their treatment. Recently, the opioid system has regained interest in depression studies. Research in animals and humans suggest that blocking the kappa opioid receptor (KOR) may potentially alleviate the symptoms of depression. The mechanism behind this effect is not fully understood. Stress and alterations in hypothalamic-pituitary-adrenal axis (HPA-axis) activity are thought to play a crucial role in depression. This study aimed to characterize stress hormones and stress-related protein expression following activation of KOR using a selective agonist. The longitudinal effect was investigated 24 h after KOR activation using the selective agonist U50,488 in Sprague Dawley rats. Stress-related hormones and protein expression patterns were explored using multiplex bead-based assays and western blotting. We found that KOR activation caused an increase in both adrenocorticotropic hormone (ACTH) and corticosterone (CORT) in serum. Regarding protein assays in different brain regions, phosphorylated glucocorticoid receptors also increased significantly in thalamus (THL), hypothalamus (HTH), and striatum (STR). C-Fos increased time-dependently in THL following KOR activation, extracellular signal-regulated kinases 1/2 (ERK1/2) increased significantly in STR and amygdala (AMG), while phosphorylated ERK1/2 decreased during the first 2 h and then increased again in AMG and prefrontal cortex (PFC). This study shows that KOR activation alters the HPA axis and ERK signaling which may cause to develop mood disorders.
Collapse
Affiliation(s)
- Bardia Varastehmoradi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Karen L Smith
- Alkermes, Inc., Biology, Waltham, MA, United States of America
| | - Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Connie Sanchez
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Alkermes, Inc., Biology, Waltham, MA, United States of America
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
18
|
Francis TC, Porcu A. Emotionally clocked out: cell-type specific regulation of mood and anxiety by the circadian clock system in the brain. Front Mol Neurosci 2023; 16:1188184. [PMID: 37441675 PMCID: PMC10333695 DOI: 10.3389/fnmol.2023.1188184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/29/2023] [Indexed: 07/15/2023] Open
Abstract
Circadian rhythms are self-sustained oscillations of biological systems that allow an organism to anticipate periodic changes in the environment and optimally align feeding, sleep, wakefulness, and the physiological and biochemical processes that support them within the 24 h cycle. These rhythms are generated at a cellular level by a set of genes, known as clock genes, which code for proteins that inhibit their own transcription in a negative feedback loop and can be perturbed by stress, a risk factor for the development of mood and anxiety disorders. A role for circadian clocks in mood and anxiety has been suggested for decades on the basis of clinical observations, and the dysregulation of circadian rhythms is a prominent clinical feature of stress-related disorders. Despite our understanding of central clock structure and function, the effect of circadian dysregulation in different neuronal subtypes in the suprachiasmatic nucleus (SCN), the master pacemaker region, as well as other brain systems regulating mood, including mesolimbic and limbic circuits, is just beginning to be elucidated. In the brain, circadian clocks regulate neuronal physiological functions, including neuronal activity, synaptic plasticity, protein expression, and neurotransmitter release which in turn affect mood-related behaviors via cell-type specific mechanisms. Both animal and human studies have revealed an association between circadian misalignment and mood disorders and suggest that internal temporal desynchrony might be part of the etiology of psychiatric disorders. To date, little work has been conducted associating mood-related phenotypes to cell-specific effects of the circadian clock disruptions. In this review, we discuss existing literature on how clock-driven changes in specific neuronal cell types might disrupt phase relationships among cellular communication, leading to neuronal circuit dysfunction and changes in mood-related behavior. In addition, we examine cell-type specific circuitry underlying mood dysfunction and discuss how this circuitry could affect circadian clock. We provide a focus for future research in this area and a perspective on chronotherapies for mood and anxiety disorders.
Collapse
Affiliation(s)
- T. Chase Francis
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| | - Alessandra Porcu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
19
|
Papp M, Gruca P, Litwa E, Lason M, Willner P. Optogenetic stimulation of transmission from prelimbic cortex to nucleus accumbens core overcomes resistance to venlafaxine in an animal model of treatment-resistant depression. Prog Neuropsychopharmacol Biol Psychiatry 2023; 123:110715. [PMID: 36610613 DOI: 10.1016/j.pnpbp.2023.110715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
BACKGROUND Our earlier study demonstrated that repeated optogenetic stimulation of afferents from ventral hippocampus (vHIP) to the prelimbic region of medial prefrontal cortex (mPFC) overcame resistance to antidepressant treatment in Wistar-Kyoto (WKY) rats. These results suggested that antidepressant resistance may result from an insufficiency of transmission from vHIP to mPFC. Here we examined whether similar effects can be elicited from major output of mPFC; the pathway from to nucleus accumbens core (NAc). METHOD WKY rats were subjected to Chronic Mild Stress and were used in two sets of experiments: 1) they were treated acutely with optogenetic stimulation of afferents to NAc core originating from the mPFC, and 2) they were treated with chronic (5 weeks) venlafaxine (10 mg/kg) and/or repeated (once weekly) optogenetic stimulation of afferents to NAc originating from either mPFC or vHIP. RESULTS Chronic mild stress procedure decreased sucrose intake, open arm entries on elevated plus maze, and novel object recognition test. Acute optogenetic stimulation of the mPFC-NAc and vHIP-NAc pathways had no effect in sucrose or plus maze tests, but increased object recognition. Neither venlafaxine nor mPFC-NAc optogenetic stimulation alone was effective in reversing the effects of CMS, but the combination of chronic antidepressant and repeated optogenetic stimulation improved behaviour on all three measures. CONCLUSIONS The synergism between venlafaxine and mPFC-NAc optogenetic stimulation supports the hypothesis that the mechanisms of non-responsiveness of WKY rats involves a failure of antidepressant treatment to restore transmission in the mPFC-NAc pathway. Together with earlier results, this implicates insufficiency in a vHIP-mPFC-NAc circuit in non-responsiveness to antidepressant drugs.
Collapse
Affiliation(s)
- Mariusz Papp
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland.
| | - Piotr Gruca
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Ewa Litwa
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Magdalena Lason
- Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Paul Willner
- Department of Psychology, Swansea University, Swansea, UK
| |
Collapse
|
20
|
Hicks EM, Seah C, Cote A, Marchese S, Brennand KJ, Nestler EJ, Girgenti MJ, Huckins LM. Integrating genetics and transcriptomics to study major depressive disorder: a conceptual framework, bioinformatic approaches, and recent findings. Transl Psychiatry 2023; 13:129. [PMID: 37076454 PMCID: PMC10115809 DOI: 10.1038/s41398-023-02412-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 04/21/2023] Open
Abstract
Major depressive disorder (MDD) is a complex and heterogeneous psychiatric syndrome with genetic and environmental influences. In addition to neuroanatomical and circuit-level disturbances, dysregulation of the brain transcriptome is a key phenotypic signature of MDD. Postmortem brain gene expression data are uniquely valuable resources for identifying this signature and key genomic drivers in human depression; however, the scarcity of brain tissue limits our capacity to observe the dynamic transcriptional landscape of MDD. It is therefore crucial to explore and integrate depression and stress transcriptomic data from numerous, complementary perspectives to construct a richer understanding of the pathophysiology of depression. In this review, we discuss multiple approaches for exploring the brain transcriptome reflecting dynamic stages of MDD: predisposition, onset, and illness. We next highlight bioinformatic approaches for hypothesis-free, genome-wide analyses of genomic and transcriptomic data and their integration. Last, we summarize the findings of recent genetic and transcriptomic studies within this conceptual framework.
Collapse
Affiliation(s)
- Emily M Hicks
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Carina Seah
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Alanna Cote
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Shelby Marchese
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Kristen J Brennand
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06511, USA
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA
| | - Matthew J Girgenti
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA.
| | - Laura M Huckins
- Pamela Sklar Division of Psychiatric Genomics, Departments of Psychiatry and of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, 10029, USA.
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, 06511, USA.
| |
Collapse
|
21
|
Jiang Y, Zou M, Wang Y, Wang Y. Nucleus accumbens in the pathogenesis of major depressive disorder: A brief review. Brain Res Bull 2023; 196:68-75. [PMID: 36889362 DOI: 10.1016/j.brainresbull.2023.03.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/16/2023] [Accepted: 03/05/2023] [Indexed: 03/08/2023]
Abstract
Major depressive disorder (MDD) is the most prevalent mental disorder characterized by anhedonia, loss of motivation, avolition, behavioral despair and cognitive abnormalities. Despite substantial advancements in the pathophysiology of MDD in recent years, the pathogenesis of this disorder is not fully understood. Meanwhile,the treatment of MDD with currently available antidepressants is inadequate, highlighting the urgent need for clarifying the pathophysiology of MDD and developing novel therapeutics. Extensive studies have demonstrated the involvement of nuclei such as the prefrontal cortex (PFC), hippocampus (HIP), nucleus accumbens (NAc), hypothalamus, etc., in MDD. NAc,a region critical for reward and motivation,dysregulation of its activity seems to be a hallmark of this mood disorder. In this paper, we present a review of NAc related circuits, cellular and molecular mechanisms underlying MDD and share an analysis of the gaps in current research and possible future research directions.
Collapse
Affiliation(s)
- Yajie Jiang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China
| | - Manshu Zou
- Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China
| | - Yeqing Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Yuhong Wang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China; Hunan Key Laboratory of Traditional Chinese Medicine Prevention & Treatment of Depressive Diseases, Changsha, China.
| |
Collapse
|
22
|
Ryan KM, Smyth P, Blackshields G, Kranaster L, Sartorius A, Sheils O, McLoughlin DM. Electroconvulsive Stimulation in Rats Induces Alterations in the Hippocampal miRNome: Translational Implications for Depression. Mol Neurobiol 2023; 60:1150-1163. [PMID: 36414911 DOI: 10.1007/s12035-022-03131-8] [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: 08/18/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022]
Abstract
MicroRNAs (miRNAs) may contribute to the development of depression and its treatment. Here, we used the hypothesis-neutral approach of next-generation sequencing (NGS) to gain comprehensive understanding of the effects of a course of electroconvulsive stimulation (ECS), the animal model equivalent of electroconvulsive therapy (ECT), on rat hippocampal miRNAs. Significant differential expression (p < 0.001) of six hippocampal miRNAs was noted following NGS, after correcting for multiple comparisons. Three of these miRNAs were upregulated (miR-132, miR-212, miR-331) and three downregulated (miR-204, miR-483, miR-301a). qRT-PCR confirmed significant changes in four of the six miRNAs (miR-132, miR-212, miR-204, miR-483). miR-483 was also significantly reduced in frontal cortex, though no other significant alterations were noted in frontal cortex, cerebellum, or whole blood. Assessing the translatability of the results, miR-132 and miR-483 were significantly reduced in whole blood samples from medicated patients with depression (n = 50) compared to healthy controls (n = 45), though ECT had no impact on miRNA levels. Notably, pre-ECT miR-204 levels moderately positively correlated with depression severity at baseline and moderately negatively correlated with mood score reduction post-ECT. miRNAs were also examined in cerebrospinal fluid and serum from a separate cohort of patients (n = 8) treated with ECT; no significant changes were noted post-treatment. However, there was a large positive correlation between changes in miR-212 and mood score post-ECT in serum. Though replication studies using larger sample sizes are required, alterations in miRNA expression may be informative about the mechanism of action of ECS/ECT and in turn might give insight into the neurobiology of depression.
Collapse
Affiliation(s)
- Karen M Ryan
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland. .,Department of Psychiatry, Trinity College Dublin, St Patrick's University Hospital, Dublin 8, Ireland.
| | - Paul Smyth
- Department of Histopathology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Gordon Blackshields
- Department of Histopathology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Laura Kranaster
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty, Mannheim/Heidelberg University, Mannheim, Germany
| | - Alexander Sartorius
- Department of Psychiatry and Psychotherapy, Central Institute of Mental Health, Medical Faculty, Mannheim/Heidelberg University, Mannheim, Germany
| | - Orla Sheils
- Department of Histopathology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Declan M McLoughlin
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.,Department of Psychiatry, Trinity College Dublin, St Patrick's University Hospital, Dublin 8, Ireland
| |
Collapse
|
23
|
Peng S, Zhou Y, Xiong L, Wang Q. Identification of novel targets and pathways to distinguish suicide dependent or independent on depression diagnosis. Sci Rep 2023; 13:2488. [PMID: 36781900 PMCID: PMC9925752 DOI: 10.1038/s41598-023-29101-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
In recent years, postmortem brain studies have revealed that some molecular, cellular, and circuit changes associated with suicide, have an independent or additive effect on depression. The aim of the present study is to identify potential phenotypic, tissue, and sex-specific novel targets and pathways to distinguish depression or suicide from major depressive disorder (MDD) comorbid with suicide. The mRNA expression profiling datasets from two previous independent postmortem brain studies of suicide and depression (GSE102556 and GSE101521) were retrieved from the GEO database. Machine learning analysis was used to differentiate three regrouped gene expression profiles, i.e., MDD with suicide, MDD without suicide, and suicide without depression. Weighted correlation network analysis (WGCNA) was further conducted to identify the key modules and hub genes significantly associated with each of these three sub-phenotypes. TissueEnrich approaches were used to find the essential brain tissues and the difference of tissue enriched genes between depression with or without suicide. Dysregulated gene expression cross two variables, including phenotypes and tissues, were determined by global analysis with Vegan. RRHO analysis was applied to examine the difference in global expression pattern between male and female groups. Using the optimized machine learning model, several ncRNAs and mRNAs with higher AUC and MeanDecreaseGini, including GCNT1P1 and AC092745.1, etc., were identified as potential molecular targets to distinguish suicide with, or without MDD and depression without suicide. WGCNA analysis identified some key modules significantly associated with these three phenotypes, and the gene biological functions of the key modules mainly relate to ncRNA and miRNA processing, as well as oxidoreductase and dehydrogenase activity. Hub genes such as RP11-349A22.5, C20orf196, MAPK8IP3 and RP11-697N18.2 were found in these key modules. TissueEnrich analysis showed that nucleus accumbens and subiculum were significantly changed among the 6 brain regions studied. Global analysis with Vegan and RRHO identified PRS26, ARNT and SYN3 as the most significantly differentially expressed genes across phenotype and tissues, and there was little overlap between the male and female groups. In this study, we have identified novel gene targets, as well as annotated functions of co-expression patterns and hub genes that are significantly distinctive between depression with suicide, depression without suicide, and suicide without depression. Moreover, global analysis across three phenotypes and tissues confirmed the evidence of sex difference in mood disorders.
Collapse
Affiliation(s)
- Siqi Peng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yalan Zhou
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lan Xiong
- Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada.
| | - Qingzhong Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
24
|
Hu C, Dewey BE, Mowry EM, Fitzgerald KC. Deep gray matter substructure volumes and depressive symptoms in a large multiple sclerosis cohort. Mult Scler 2023:13524585221148144. [PMID: 36691798 DOI: 10.1177/13524585221148144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Consistent findings on underlying brain features or specific structural atrophy patterns contributing to depression in multiple sclerosis (MS) are limited. OBJECTIVE To investigate how deep gray matter (DGM) features predict depressive symptom trajectories in MS patients. METHODS We used data from the MS Partners Advancing Technology and Health Solutions (MS PATHS) network in which standardized patient information and outcomes are collected. We performed whole-brain segmentation using SLANT-CRUISE. We assessed if DGM structures were associated with elevated depressive symptoms over follow-up and with depressive symptom phenotypes. RESULTS We included 3844 participants (average age: 46.05 ± 11.83 years; 72.7% female) of whom 1905 (49.5%) experienced ⩾1 periods of elevated depressive symptoms over 2.6 ± 0.9 years mean follow-up. Higher caudate, putamen, accumbens, ventral diencephalon, thalamus, and amygdala volumes were associated with lower odds of elevated depressive symptoms over follow-up (odds ratio (OR) range per 1 SD (standard deviation) increase in volume: 0.88-0.94). For example, a 1 SD increase in accumbens or caudate volume was associated with 12% or 10% respective lower odds of having a period of elevated depressive symptoms over follow-up (for accumbens: OR: 0.88; 95% confidence interval (CI): 0.83-0.93; p < 0.001; for caudate: OR: 0.90; 95% CI: 0.85-0.96; p = 0.003). CONCLUSION Lower DGM volumes were associated with depressive symptom trajectories in MS.
Collapse
Affiliation(s)
- Chen Hu
- Division of Neuroimmunology and Neurological Infections, Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Blake E Dewey
- Division of Neuroimmunology and Neurological Infections, Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ellen M Mowry
- Division of Neuroimmunology and Neurological Infections, Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kathryn C Fitzgerald
- Division of Neuroimmunology and Neurological Infections, Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA/Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
25
|
Shi P, Hu L, Ren H, Dai Q. Reward enhances resilience to chronic social defeat stress in mice: Neural ECs and mGluR5 mechanism via neuroprotection in VTA and DRN. Front Psychiatry 2023; 14:1084367. [PMID: 36873216 PMCID: PMC9978385 DOI: 10.3389/fpsyt.2023.1084367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/13/2023] [Indexed: 02/18/2023] Open
Abstract
INTRODUCTION Stress often leads to emotional disorders such as depression. The reward might render this effect through the enhancement of stress resilience. However, the effect of reward on stress resilience under different intensities of stress needs more evidence, and its potential neural mechanism has been poorly revealed. It has been reported that the endogenous cannabinoid system (ECs) and downstream metabolic glutamate receptor 5 (mGluR5) are closely related to stress and reward, which might be the potential cerebral mechanism between reward and stress resilience, but there is a lack of direct evidence. This study aims to observe the effect of reward on stress resilience under different intensities of stress and further explore potential cerebral mechanisms underlying this effect. METHODS Using the chronic social defeat stress model, we applied reward (accompanied by a female mouse) under different intensities of stress in mice during the modeling process. The impact of reward on stress resilience and the potential cerebral mechanism were observed after modeling through behavioral tests and biomolecules. RESULTS The results showed that stronger stress led to higher degrees of depression-like behavior. Reward reduced depression-like behavior and enhanced stress resilience (all p-value <0.05) (more social interaction in the social test, less immobility time in the forced swimming test, etc.), with a stronger effect under the large stress. Furthermore, the mRNA expression levels of CB1 and mGluR5, the protein expression level of mGluR5, and the expression level of 2-AG (2-arachidonoylglycerol) in both ventral tegmental area (VTA) and dorsal raphe nucleus (DRN) were significantly upregulated by reward after modeling (all p-value <0.05). However, the protein expression of CB1 in VTA and DRN and the expression of AEA (anandamide) in VTA did not differ significantly between groups. Intraperitoneal injection of a CB1 agonist (URB-597) during social defeat stress significantly reduced depression-like behavior compared with a CB1 inhibitor (AM251) (all p-value <0.05). Interestingly, in DRN, the expression of AEA in the stress group was lower than that of the control group, with or without reward (all p-value <0.05). DISCUSSION These findings demonstrate that combined social and sexual reward has a positive effect on stress resilience during chronic social defeat stress, potentially by influencing the ECs and mGluR5 in VTA and DRN.
Collapse
Affiliation(s)
- Peixia Shi
- Department of Medical Psychology, Army Medical University, Chongqing, China.,Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Linlin Hu
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hui Ren
- Department of Nursing Psychology, Army Medical University, Chongqing, China
| | - Qin Dai
- Department of Medical Psychology, Army Medical University, Chongqing, China.,Department of Nursing Psychology, Army Medical University, Chongqing, China
| |
Collapse
|
26
|
Thompson SM. Plasticity of synapses and reward circuit function in the genesis and treatment of depression. Neuropsychopharmacology 2023; 48:90-103. [PMID: 36057649 PMCID: PMC9700729 DOI: 10.1038/s41386-022-01422-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/18/2022] [Accepted: 08/01/2022] [Indexed: 11/08/2022]
Abstract
What changes in brain function cause the debilitating symptoms of depression? Can we use the answers to this question to invent more effective, faster acting antidepressant drug therapies? This review provides an overview and update of the converging human and preclinical evidence supporting the hypothesis that changes in the function of excitatory synapses impair the function of the circuits they are embedded in to give rise to the pathological changes in mood, hedonic state, and thought processes that characterize depression. The review also highlights complementary human and preclinical findings that classical and novel antidepressant drugs relieve the symptoms of depression by restoring the functions of these same synapses and circuits. These findings offer a useful path forward for designing better antidepressant compounds.
Collapse
Affiliation(s)
- Scott M Thompson
- Department of Psychiatry, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, 80045, CO, USA.
| |
Collapse
|
27
|
Szczurowska E, Szánti-Pintér E, Chetverikov N, Randáková A, Kudová E, Jakubík J. Modulation of Muscarinic Signalling in the Central Nervous System by Steroid Hormones and Neurosteroids. Int J Mol Sci 2022; 24:ijms24010507. [PMID: 36613951 PMCID: PMC9820491 DOI: 10.3390/ijms24010507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
Muscarinic acetylcholine receptors expressed in the central nervous system mediate various functions, including cognition, memory, or reward. Therefore, muscarinic receptors represent potential pharmacological targets for various diseases and conditions, such as Alzheimer's disease, schizophrenia, addiction, epilepsy, or depression. Muscarinic receptors are allosterically modulated by neurosteroids and steroid hormones at physiologically relevant concentrations. In this review, we focus on the modulation of muscarinic receptors by neurosteroids and steroid hormones in the context of diseases and disorders of the central nervous system. Further, we propose the potential use of neuroactive steroids in the development of pharmacotherapeutics for these diseases and conditions.
Collapse
Affiliation(s)
- Ewa Szczurowska
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Náměstí 2, Prague 6, 166 10 Prague, Czech Republic
| | - Eszter Szánti-Pintér
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Náměstí 2, Prague 6, 166 10 Prague, Czech Republic
| | - Nikolai Chetverikov
- Institute of Physiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Alena Randáková
- Institute of Physiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Eva Kudová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo Náměstí 2, Prague 6, 166 10 Prague, Czech Republic
- Correspondence: (E.K.); (J.J.)
| | - Jan Jakubík
- Institute of Physiology, Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
- Correspondence: (E.K.); (J.J.)
| |
Collapse
|
28
|
Enhancing Endocannabinoid Signaling via β-Catenin in the Nucleus Accumbens Attenuates PTSD- and Depression-like Behavior of Male Rats. Biomedicines 2022; 10:biomedicines10081789. [PMID: 35892688 PMCID: PMC9394396 DOI: 10.3390/biomedicines10081789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 12/15/2022] Open
Abstract
Inhibition of fatty acid amide hydrolase (FAAH), which increases anandamide levels, has been suggested as a potential treatment for stress-related conditions. We examined whether the stress-preventing effects of the FAAH inhibitor URB597 on behavior are mediated via β-catenin in the nucleus accumbens (NAc). Male rats were exposed to the shock and reminders model of PTSD and then treated with URB597 (0.4 mg/kg; i.p.). They were tested for anxiety- (freezing, startle response), depression-like behaviors (despair, social preference, anhedonia), and memory function (T-maze, social recognition). We also tested the involvement of the CB1 receptor (CB1r), β-catenin, and metabotropic glutamate receptor subtype 5 (mGluR5) proteins. URB597 prevented the shock- and reminders-induced increase in anxiety- and depressive-like behaviors, as well as the impaired memory via the CB1r-dependent mechanism. In the NAc, viral-mediated β-catenin overexpression restored the behavior of rats exposed to stress and normalized the alterations in protein levels in the NAc and the prefrontal cortex. Importantly, when NAc β-catenin levels were downregulated by viral-mediated gene transfer, the therapeutic-like effects of URB597 were blocked. We suggest a potentially novel mechanism for the therapeutic-like effects of FAAH inhibition that is dependent on β-catenin activation in the NAc in a PTSD rat model.
Collapse
|
29
|
Kim J, Kang S, Choi TY, Chang KA, Koo JW. Metabotropic Glutamate Receptor 5 in Amygdala Target Neurons Regulates Susceptibility to Chronic Social Stress. Biol Psychiatry 2022; 92:104-115. [PMID: 35314057 DOI: 10.1016/j.biopsych.2022.01.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 01/10/2022] [Accepted: 01/13/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND Metabotropic glutamate receptor 5 (mGluR5) has been implicated in stress-related psychiatric disorders, particularly major depressive disorder. Although growing evidence supports the proresilient role of mGluR5 in corticolimbic circuitry in the depressive-like behaviors following chronic stress exposure, the underlying neural mechanisms, including circuits and molecules, remain unknown. METHODS We measured the c-Fos expression and probability of neurotransmitter release in and from basolateral amygdala (BLA) neurons projecting to the medial prefrontal cortex (mPFC) and to the ventral hippocampus (vHPC) after chronic social defeat stress. The role of BLA projections in depressive-like behaviors was assessed using optogenetic manipulations, and the underlying molecular mechanisms of mGluR5 and downstream signaling were investigated by Western blotting, viral-mediated gene transfer, and pharmacological manipulations. RESULTS Chronic social defeat stress disrupted neural activity and glutamatergic transmission in both BLA projections. Optogenetic activation of BLA projections reversed the detrimental effects of chronic social defeat stress on depressive-like behaviors and mGluR5 expression in the mPFC and vHPC. Conversely, inhibition of BLA projections of mice undergoing subthreshold social defeat stress induced a susceptible phenotype and mGluR5 reduction. These two BLA circuits appeared to act in an independent way. We demonstrate that mGluR5 overexpression in the mPFC or vHPC was proresilient while the mGluR5 knockdown was prosusceptible and that the proresilient effects of mGluR5 are mediated through distinctive downstream signaling pathways in the mPFC and vHPC. CONCLUSIONS These findings identify mGluR5 in the mPFC and vHPC that receive BLA inputs as a critical mediator of stress resilience, highlighting circuit-specific signaling for depressive-like behaviors.
Collapse
Affiliation(s)
- Jeongseop Kim
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Shinwoo Kang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Tae-Yong Choi
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| | - Keun-A Chang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea; Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea.
| | - Ja Wook Koo
- Emotion, Cognition and Behavior Research Group, Korea Brain Research Institute, Daegu, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea.
| |
Collapse
|
30
|
Figee M, Riva-Posse P, Choi KS, Bederson L, Mayberg HS, Kopell BH. Deep Brain Stimulation for Depression. Neurotherapeutics 2022; 19:1229-1245. [PMID: 35817944 PMCID: PMC9587188 DOI: 10.1007/s13311-022-01270-3] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2022] [Indexed: 11/29/2022] Open
Abstract
Deep brain stimulation has been extensively studied as a therapeutic option for treatment-resistant depression (TRD). DBS across different targets is associated with on average 60% response rates in previously refractory chronically depressed patients. However, response rates vary greatly between patients and between studies and often require extensive trial-and-error optimizations of stimulation parameters. Emerging evidence from tractography imaging suggests that targeting combinations of white matter tracts, rather than specific grey matter regions, is necessary for meaningful antidepressant response to DBS. In this article, we review efficacy of various DBS targets for TRD, which networks are involved in their therapeutic effects, and how we can use this information to improve targeting and programing of DBS for individual patients. We will also highlight how to integrate these DBS network findings into developing adaptive stimulation and optimal trial designs.
Collapse
Affiliation(s)
- Martijn Figee
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Patricio Riva-Posse
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Georgia, GA, USA
| | - Ki Sueng Choi
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lucia Bederson
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Helen S Mayberg
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian H Kopell
- Nash Family Center for Advanced Circuit Therapeutics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
31
|
Liu J. Involvement of PKMζ in Stress Response and Depression. Front Cell Neurosci 2022; 16:907767. [PMID: 35669107 PMCID: PMC9163780 DOI: 10.3389/fncel.2022.907767] [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: 03/30/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022] Open
Abstract
The stress system in the brain plays a pivotal role in keeping humans and animals from harmful stimuli. However, excessive stress will cause maladaptive changes to the stress system and lead to depression. Despite the high prevalence of depression, the treatment remains limited. PKMζ, an atypical PKC isoform, has been demonstrated to play a crucial role in maintaining long-term potentiation and memory. Recent evidence shows that PKMζ is also involved in stress response and depressive-like behavior. In particular, it was demonstrated that stress that resulted in depressive-like behavior could decrease the expression of PKMζ in the prefrontal cortex, which could be reversed by antidepressants. Importantly, modulation of PKMζ expression could regulate depressive-like behaviors and the actions of antidepressants. These data suggested that PKMζ could be a molecular target for developing novel antidepressants. Here, I review the advance on the role of PKMζ in mediating stress response and its involvement in the development of depression.
Collapse
Affiliation(s)
- Jianfeng Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| |
Collapse
|
32
|
O’Brien C, Vemireddy R, Mohammed U, Barker DJ. Stress reveals a specific behavioral phenotype for opioid abuse susceptibility. J Exp Anal Behav 2022; 117:518-531. [PMID: 35119105 PMCID: PMC9090955 DOI: 10.1002/jeab.738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/22/2021] [Accepted: 01/06/2022] [Indexed: 11/07/2022]
Abstract
Susceptibility to stress has long been considered important for the development of substance use disorders. Nonetheless, behavioral and physiological responses to stress are highly variable, making it difficult to identify the individuals who are most likely to abuse drugs. In the present study, we employed a comprehensive battery of tests for negative valence behaviors and nociception to identify individuals predisposed to opioid seeking following oral opioid self-administration. Furthermore, we examined how this profile was affected by a history of stress. We observed that mice receiving foot shock stress failed to exhibit a preference for sucrose, showed increased immobility in the forced swim task, and exhibited mechanical hypersensitivity when compared to controls. When considering these behaviors in light of future fentanyl-seeking responses, we observed that heightened mechanical sensitivity corresponded to higher opioid preference in mice with a history of stress, but not controls. Moreover, we were surprised to discover that paradoxically high sucrose preferences predicted fentanyl preference in shock mice, while signs of anhedonia predicted fentanyl preference in controls. Taken together, these results indicate that stress can act as a physiological modulator, shifting profiles of opioid abuse susceptibility depending on an individual's history.
Collapse
Affiliation(s)
- Chris O’Brien
- Department of Psychology, Rutgers, The State University of New Jersey
| | - Roshni Vemireddy
- Department of Psychology, Rutgers, The State University of New Jersey
| | - Uzma Mohammed
- Department of Psychology, Rutgers, The State University of New Jersey
| | - David J. Barker
- Department of Psychology, Rutgers, The State University of New Jersey
- Brain Health Institute, Rutgers University, Piscataway, NJ
| |
Collapse
|
33
|
Huang X, Hu J, Peng H, Cheng HW. Embryonic Exposure to Tryptophan Yields Bullying Victimization via Reprogramming the Microbiota-Gut-Brain Axis in a Chicken Model. Nutrients 2022; 14:nu14030661. [PMID: 35277020 PMCID: PMC8839409 DOI: 10.3390/nu14030661] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/26/2022] [Accepted: 01/30/2022] [Indexed: 02/04/2023] Open
Abstract
Maternal metabolic disorder during early pregnancy may give rise to emotional and behavioral disorders in the child, vulnerable to bullying. Placental tryptophan fluctuation consequently disrupts offspring gut microbiome and brain neurogenesis with long-lasting physiological and social behavioral impacts. The aim of this study was to examine the hypothesis that the excess gestational tryptophan may affect children’s mental and physical development via modifying the microbiota-gut-brain axis, which lays the foundation of their mental status. Chicken embryo was employed due to its robust microbiota and independence of maternal influences during embryogenesis. The results indicated that embryonic tryptophan exposure reduced body weight and aggressiveness in the male offspring before and during adolescence. Additionally, the relative gut length and crypt depth were increased, while the villus/crypt ratio was decreased in tryptophan treated roosters, which was corresponding to the changes in the cecal microbiota composition. Furthermore, the catecholamine concentrations were increased in tryptophan group, which may be associated with the alterations in the gut microbiome and the gut-brain axis’s function. These changes may underlie the sociometric status of bullying; clarify how gestational tryptophan fluctuation compromises bullying and provide a strategy to prevent bullying by controlling dietary tryptophan and medication therapy during pregnancy.
Collapse
Affiliation(s)
- Xiaohong Huang
- Institute of Neuroregeneration & Neurorehabilitation, Department of Pathophysiology, Qingdao University, Qingdao 266071, China
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.H.); (H.-w.C.)
- Correspondence: ; Tel.: +86-15908942478
| | - Jiaying Hu
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.H.); (H.-w.C.)
| | - Haining Peng
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266071, China;
| | - Heng-wei Cheng
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; (J.H.); (H.-w.C.)
- Livestock Behavior Research Unit, USDA-ARS, West Lafayette, IN 47907, USA
| |
Collapse
|
34
|
Fulton S, Décarie-Spain L, Fioramonti X, Guiard B, Nakajima S. The menace of obesity to depression and anxiety prevalence. Trends Endocrinol Metab 2022; 33:18-35. [PMID: 34750064 DOI: 10.1016/j.tem.2021.10.005] [Citation(s) in RCA: 160] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/14/2021] [Accepted: 10/16/2021] [Indexed: 02/07/2023]
Abstract
The incidence of depression and anxiety is amplified by obesity. Mounting evidence reveals that the psychiatric consequences of obesity stem from poor diet, inactivity, and visceral adipose accumulation. Resulting metabolic and vascular dysfunction, including inflammation, insulin and leptin resistance, and hypertension, have emerged as key risks to depression and anxiety development. Recent research advancements are exposing the important contribution of these different corollaries of obesity and their impact on neuroimmune status and the neural circuits controlling mood and emotional states. Along these lines, this review connects the clinical manifestations of depression and anxiety in obesity to our current understanding of the origins and biology of immunometabolic threats to central nervous system function and behavior.
Collapse
Affiliation(s)
- Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Nutrition, Université de Montréal, Montréal, QC H3T1J4, Canada.
| | - Léa Décarie-Spain
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Université de Montréal, Montréal, QC H3T1J4, Canada
| | - Xavier Fioramonti
- NutriNeuro, UMR 1286 INRAE, Bordeaux INP, Bordeaux University, Bordeaux, France
| | - Bruno Guiard
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), CNRS UMR5169, UPS, Université de Toulouse, Toulouse, France
| | - Shingo Nakajima
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Nutrition, Université de Montréal, Montréal, QC H3T1J4, Canada
| |
Collapse
|
35
|
Mantas I, Saarinen M, Xu ZQD, Svenningsson P. Update on GPCR-based targets for the development of novel antidepressants. Mol Psychiatry 2022; 27:534-558. [PMID: 33589739 PMCID: PMC8960420 DOI: 10.1038/s41380-021-01040-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 01/31/2023]
Abstract
Traditional antidepressants largely interfere with monoaminergic transport or degradation systems, taking several weeks to have their therapeutic actions. Moreover, a large proportion of depressed patients are resistant to these therapies. Several atypical antidepressants have been developed which interact with G protein coupled receptors (GPCRs) instead, as direct targeting of receptors may achieve more efficacious and faster antidepressant actions. The focus of this review is to provide an update on how distinct GPCRs mediate antidepressant actions and discuss recent insights into how GPCRs regulate the pathophysiology of Major Depressive Disorder (MDD). We also discuss the therapeutic potential of novel GPCR targets, which are appealing due to their ligand selectivity, expression pattern, or pharmacological profiles. Finally, we highlight recent advances in understanding GPCR pharmacology and structure, and how they may provide new avenues for drug development.
Collapse
Affiliation(s)
- Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Marcus Saarinen
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Zhi-Qing David Xu
- Department of Neurobiology, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
36
|
Eren-Koçak E, Dalkara T. Ion Channel Dysfunction and Neuroinflammation in Migraine and Depression. Front Pharmacol 2021; 12:777607. [PMID: 34858192 PMCID: PMC8631474 DOI: 10.3389/fphar.2021.777607] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/22/2021] [Indexed: 01/15/2023] Open
Abstract
Migraine and major depression are debilitating disorders with high lifetime prevalence rates. Interestingly these disorders are highly comorbid and show significant heritability, suggesting shared pathophysiological mechanisms. Non-homeostatic function of ion channels and neuroinflammation may be common mechanisms underlying both disorders: The excitation-inhibition balance of microcircuits and their modulation by monoaminergic systems, which depend on the expression and function of membrane located K+, Na+, and Ca+2 channels, have been reported to be disturbed in both depression and migraine. Ion channels and energy supply to synapses not only change excitability of neurons but can also mediate the induction and maintenance of inflammatory signaling implicated in the pathophysiology of both disorders. In this respect, Pannexin-1 and P2X7 large-pore ion channel receptors can induce inflammasome formation that triggers release of pro-inflammatory mediators from the cell. Here, the role of ion channels involved in the regulation of excitation-inhibition balance, synaptic energy homeostasis as well as inflammatory signaling in migraine and depression will be reviewed.
Collapse
Affiliation(s)
- Emine Eren-Koçak
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey.,Department of Psychiatry, Medical Faculty, Hacettepe University, Ankara, Turkey
| | - Turgay Dalkara
- Institute of Neurological Sciences and Psychiatry, Hacettepe University, Ankara, Turkey
| |
Collapse
|
37
|
Movassaghi CS, Perrotta KA, Yang H, Iyer R, Cheng X, Dagher M, Fillol MA, Andrews AM. Simultaneous serotonin and dopamine monitoring across timescales by rapid pulse voltammetry with partial least squares regression. Anal Bioanal Chem 2021; 413:6747-6767. [PMID: 34686897 PMCID: PMC8551120 DOI: 10.1007/s00216-021-03665-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/11/2021] [Accepted: 09/14/2021] [Indexed: 11/12/2022]
Abstract
Many voltammetry methods have been developed to monitor brain extracellular dopamine levels. Fewer approaches have been successful in detecting serotonin in vivo. No voltammetric techniques are currently available to monitor both neurotransmitters simultaneously across timescales, even though they play integrated roles in modulating behavior. We provide proof-of-concept for rapid pulse voltammetry coupled with partial least squares regression (RPV-PLSR), an approach adapted from multi-electrode systems (i.e., electronic tongues) used to identify multiple components in complex environments. We exploited small differences in analyte redox profiles to select pulse steps for RPV waveforms. Using an intentionally designed pulse strategy combined with custom instrumentation and analysis software, we monitored basal and stimulated levels of dopamine and serotonin. In addition to faradaic currents, capacitive currents were important factors in analyte identification arguing against background subtraction. Compared to fast-scan cyclic voltammetry-principal components regression (FSCV-PCR), RPV-PLSR better differentiated and quantified basal and stimulated dopamine and serotonin associated with striatal recording electrode position, optical stimulation frequency, and serotonin reuptake inhibition. The RPV-PLSR approach can be generalized to other electrochemically active neurotransmitters and provides a feedback pipeline for future optimization of multi-analyte, fit-for-purpose waveforms and machine learning approaches to data analysis.
Collapse
Affiliation(s)
- Cameron S Movassaghi
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Katie A Perrotta
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hongyan Yang
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Rahul Iyer
- Department of Electrical Engineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinyi Cheng
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Merel Dagher
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Miguel Alcañiz Fillol
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Universitat Politècnica de València - Universitat de València, Camino de Vera s/n, 46022, Valencia, Spain.
| | - Anne M Andrews
- Department of Chemistry & Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
38
|
A Role of BDNF in the Depression Pathogenesis and a Potential Target as Antidepressant: The Modulator of Stress Sensitivity "Shati/Nat8l-BDNF System" in the Dorsal Striatum. Pharmaceuticals (Basel) 2021; 14:ph14090889. [PMID: 34577589 PMCID: PMC8469819 DOI: 10.3390/ph14090889] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Depression is one of the most common mental diseases, with increasing numbers of patients globally each year. In addition, approximately 30% of patients with depression are resistant to any treatment and do not show an expected response to first-line antidepressant drugs. Therefore, novel antidepressant agents and strategies are required. Although depression is triggered by post-birth stress, while some individuals show the pathology of depression, others remain resilient. The molecular mechanisms underlying stress sensitivity remain unknown. Brain-derived neurotrophic factor (BDNF) has both pro- and anti-depressant effects, dependent on brain region. Considering the strong region-specific contribution of BDNF to depression pathogenesis, the regulation of BDNF in the whole brain is not a beneficial strategy for the treatment of depression. We reviewed a novel finding of BDNF function in the dorsal striatum, which induces vulnerability to social stress, in addition to recent research progress regarding the brain regional functions of BDNF, including the prefrontal cortex, hippocampus, and nucleus accumbens. Striatal BDNF is regulated by Shati/Nat8l, an N-acetyltransferase through epigenetic regulation. Targeting of Shati/Nat8l would allow BDNF to be striatum-specifically regulated, and the striatal Shati/Nat8l-BDNF pathway could be a promising novel therapeutic agent for the treatment of depression by modulating sensitivity to stress.
Collapse
|
39
|
Arasappan D, Eickhoff SB, Nemeroff CB, Hofmann HA, Jabbi M. Transcription Factor Motifs Associated with Anterior Insula Gene Expression Underlying Mood Disorder Phenotypes. Mol Neurobiol 2021; 58:1978-1989. [PMID: 33411239 DOI: 10.1007/s12035-020-02195-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/30/2020] [Indexed: 10/22/2022]
Abstract
Mood disorders represent a major cause of morbidity and mortality worldwide but the brain-related molecular pathophysiology in mood disorders remains largely undefined. Because the anterior insula is reduced in volume in patients with mood disorders, RNA was extracted from the anterior insula postmortem anterior insula of mood disorder samples and compared with unaffected controls for RNA-sequencing identification of differentially expressed genes (DEGs) in (a) bipolar disorder (BD; n = 37) versus (vs.) controls (n = 33), and (b) major depressive disorder (MDD n = 30) vs. controls, and (c) low vs. high axis I comorbidity (a measure of cumulative psychiatric disease burden). Given the regulatory role of transcription factors (TFs) in gene expression via specific-DNA-binding domains (motifs), we used JASPAR TF binding database to identify TF-motifs. We found that DEGs in BD vs. controls, MDD vs. controls, and high vs. low axis I comorbidity were associated with TF-motifs that are known to regulate expression of toll-like receptor genes, cellular homeostatic-control genes, and genes involved in embryonic, cellular/organ, and brain development. Robust imaging-guided transcriptomics by using meta-analytic imaging results to guide independent postmortem dissection for RNA-sequencing was applied by targeting the gray matter volume reduction in the anterior insula in mood disorders, to guide independent postmortem identification of TF motifs regulating DEG. Our findings of TF-motifs that regulate the expression of immune, cellular homeostatic-control, and developmental genes provide novel information about the hierarchical relationship between gene regulatory networks, the TFs that control them, and proximate underlying neuroanatomical phenotypes in mood disorders.
Collapse
Affiliation(s)
- Dhivya Arasappan
- Center for Biomedical Research Support, University of Texas at Austin, Austin, TX, USA
| | - Simon B Eickhoff
- Institute of Systems Neuroscience, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Institute of Neuroscience and Medicine (INM-7), Research Centre Jülich, Jülich, Germany
| | - Charles B Nemeroff
- Department of Psychiatry, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- The Mulva Clinic for Neurosciences, Dell Medical School, University of Texas at Austin, Austin, TX, USA
- Institute of Early Life Adversity Research, Austin, TX, USA
| | - Hans A Hofmann
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - Mbemba Jabbi
- Department of Psychiatry, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
- The Mulva Clinic for Neurosciences, Dell Medical School, University of Texas at Austin, Austin, TX, USA.
- Institute for Neuroscience, University of Texas at Austin, Austin, TX, USA.
- Department of Psychology, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
40
|
Meng Y, Li H, Wang J, Xu Y, Wang B. Cognitive behavioral therapy for patients with mild to moderate depression: Treatment effects and neural mechanisms. J Psychiatr Res 2021; 136:288-295. [PMID: 33631654 DOI: 10.1016/j.jpsychires.2021.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 01/29/2021] [Accepted: 02/08/2021] [Indexed: 11/18/2022]
Abstract
In this study, we combined clinical assessment and magnetic resonance imaging (MRI) techniques to investigate the brain mechanisms in mild to moderate depression (MMD) patients following cognitive behavioral therapy (CBT). Data were collected from 30 MMD patients and 18 healthy controls, and we divided patients into two treatment periods (4 weeks, 8 weeks). Clinical assessment indicated that depression characteristics, as quantified by Hamilton Depression Rating Scale (HAMD), were significantly higher in MMD patients than in healthy controls. At the baseline, MRI data revealed abnormalities in the hippocampus and nucleus accumbens (NAc) of patients with MMD, e.g., smaller gray matter volumes of the hippocampus and nucleus accumbens (NAc), as well as weaker functional connectivity between NAc and the posterior cingulate cortex/precuneus. Moreover, the hippocampus and NAc volumes were negatively correlated with the HAMD scores in MMD patients. After CBT intervention, the HAMD scores decreased, and the structural and functional characteristics of NAc in MMD patients obtained at 8-week were improved; e.g., no significant differences in NAc volume or NAc-based functional connectivity between the two groups. Taken together, our results provided evidence suggesting that CBT is an effective treatment for MMD patients. Alterations of gray matter volume and resting-state functional connectivity after 8 weeks of CBT indicated a potential modulation mechanism in brain structural modifications and functional connectivity plasticity within the NAc in MMD patients.
Collapse
Affiliation(s)
- Yanjun Meng
- Nursing College, Shanxi Medical University, Taiyuan, China; Nursing College, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Hong Li
- CAS Key Laboratory of Behavioral Science, Institute of Psychology, Beijing, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing, China; Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Junjie Wang
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Yong Xu
- Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Binquan Wang
- Nursing College, Shanxi Medical University, Taiyuan, China; Department of Otolaryngology, Head and Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
41
|
Castro‐Zavala A, Martín‐Sánchez A, Luján MÁ, Valverde O. Maternal separation increases cocaine intake through a mechanism involving plasticity in glutamate signalling. Addict Biol 2021; 26:e12911. [PMID: 32329565 DOI: 10.1111/adb.12911] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 04/05/2020] [Accepted: 04/08/2020] [Indexed: 12/16/2022]
Abstract
Early-life stress (ELS) is associated with negative consequences, including maladaptive long-lasting brain effects. These alterations seem to increase the likelihood of developing substance use disorders. However, the molecular consequences of ELS are poorly understood. In the present study, we tested the impact of ELS induced by maternal separation with early weaning (MSEW) in CD1 male mice at different phases of cocaine self-administration (SA). We also investigated the subsequent alterations on GluR2, GluR1, cAMP response element-binding (CREB), and CREB-phosphorylation (pCREB) in ventral tegmental area (VTA) and nucleus accumbens (NAc) induced by both MSEW and cocaine SA. Our results show that MSEW animals expressed a higher cocaine intake, an increased vulnerability to the acquisition of cocaine SA, and incapacity to extinguish cocaine SA behaviour. MSEW mice showed decreased GluR2 and increased GluR1 and pCREB in NAc. Also, results displayed reduction of basal levels of GluR1 and CREB and an elevation of GluR1/GluR2 ratio in the VTA. Such results hint at an enhanced glutamatergic function in NAc and increased excitability of VTA DA neurons in maternally separated mice. Altogether, our results suggest that MSEW induces molecular alterations in the brain areas related to reward processing, increasing the vulnerability to depression and cocaine-seeking behaviour.
Collapse
Affiliation(s)
- Adriana Castro‐Zavala
- Neurobiology of Behaviour Research Group (GReNeC‐NeuroBio), Department of Experimental and Health Sciences Universitat Pompeu Fabra Barcelona Spain
| | - Ana Martín‐Sánchez
- Neurobiology of Behaviour Research Group (GReNeC‐NeuroBio), Department of Experimental and Health Sciences Universitat Pompeu Fabra Barcelona Spain
- Neuroscience Research Programme IMIM‐Hospital del Mar Research Institute Barcelona Spain
| | - Miguel Ángel Luján
- Neurobiology of Behaviour Research Group (GReNeC‐NeuroBio), Department of Experimental and Health Sciences Universitat Pompeu Fabra Barcelona Spain
| | - Olga Valverde
- Neurobiology of Behaviour Research Group (GReNeC‐NeuroBio), Department of Experimental and Health Sciences Universitat Pompeu Fabra Barcelona Spain
- Neuroscience Research Programme IMIM‐Hospital del Mar Research Institute Barcelona Spain
| |
Collapse
|
42
|
Clinton SM, Shupe EA, Glover ME, Unroe KA, McCoy CR, Cohen JL, Kerman IA. Modeling heritability of temperamental differences, stress reactivity, and risk for anxiety and depression: Relevance to research domain criteria (RDoC). Eur J Neurosci 2021; 55:2076-2107. [PMID: 33629390 PMCID: PMC8382785 DOI: 10.1111/ejn.15158] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/29/2021] [Accepted: 02/20/2021] [Indexed: 01/04/2023]
Abstract
Animal models provide important tools to study biological and environmental factors that shape brain function and behavior. These models can be effectively leveraged by drawing on concepts from the National Institute of Mental Health Research Domain Criteria (RDoC) Initiative, which aims to delineate molecular pathways and neural circuits that underpin behavioral anomalies that transcend psychiatric conditions. To study factors that contribute to individual differences in emotionality and stress reactivity, our laboratory utilized Sprague-Dawley rats that were selectively bred for differences in novelty exploration. Selective breeding for low versus high locomotor response to novelty produced rat lines that differ in behavioral domains relevant to anxiety and depression, particularly the RDoC Negative Valence domains, including acute threat, potential threat, and loss. Bred Low Novelty Responder (LR) rats, relative to their High Responder (HR) counterparts, display high levels of behavioral inhibition, conditioned and unconditioned fear, avoidance, passive stress coping, anhedonia, and psychomotor retardation. The HR/LR traits are heritable, emerge in the first weeks of life, and appear to be driven by alterations in the developing amygdala and hippocampus. Epigenomic and transcriptomic profiling in the developing and adult HR/LR brain suggest that DNA methylation and microRNAs, as well as differences in monoaminergic transmission (dopamine and serotonin in particular), contribute to their distinct behavioral phenotypes. This work exemplifies ways that animal models such as the HR/LR rats can be effectively used to study neural and molecular factors driving emotional behavior, which may pave the way toward improved understanding the neurobiological mechanisms involved in emotional disorders.
Collapse
Affiliation(s)
- Sarah M Clinton
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Elizabeth A Shupe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Matthew E Glover
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Keaton A Unroe
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Chelsea R McCoy
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Joshua L Cohen
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Ilan A Kerman
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.,Behavioral Health Service Line, Veterans Affairs Pittsburgh Health System, Pittsburgh, PA, USA
| |
Collapse
|
43
|
Antibiotic-induced microbiome depletion in adult mice disrupts blood-brain barrier and facilitates brain infiltration of monocytes after bone-marrow transplantation. Brain Behav Immun 2021; 92:102-114. [PMID: 33242652 DOI: 10.1016/j.bbi.2020.11.032] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/16/2020] [Accepted: 11/22/2020] [Indexed: 02/08/2023] Open
Abstract
The crosstalk between intestinal bacteria and the central nervous system, so called "the gut-brain axis", is critically important for maintaining brain homeostasis and function. This study aimed to investigate the integrity of the blood-brain barrier (BBB) and migration of bone marrow (BM)-derived cells to the brain parenchyma after intestinal microbiota depletion in adult mice. Gut microbiota dysbiosis was induced with 5 non-absorbable antibiotics in drinking water in mice that had received bone marrow transplantation (BMT) from green fluorescent protein (GFP) transgenic mice. Antibiotic-induced microbiome depletion reduced expression of tight-junction proteins of the brain blood vessels and increased BBB permeability. Fecal microbiota transplantation of antibiotics treated mice with pathogen-free gut microbiota decreased BBB permeability and up-regulated the expression of tight junction proteins. The BM-derived GFP+ cells were observed to infiltrate specific brain regions, including the nucleus accumbens (NAc), the septal nucleus (SPT) and the hippocampus (CA3). The infiltrated cells acquired a ramified microglia-like morphology and Iba1, a microglia marker, was expressed in all GFP+ cells, whereas they were negative for the astrocyte marker GFAP. Furthermore, treatment with CCR2 antagonist (RS102895) suppressed the recruitment of BM-derived monocytes to the brain. We report for the first time the migration of BM-derived monocytes to the brain regions involved in regulating emotional behaviors after depletion of intestinal microbiota in BMT background mice. However, mechanisms responsible for the migration and functions of the microglia-like infiltrated cells in the brain need further investigation. These findings indicate that monocyte recruitment to the brain in response to gut microbiota dysbiosis may represent a novel cellular mechanism that contributes to the development of brain disorders.
Collapse
|
44
|
Brown RW, Bhide PG, Gill WD, Peeters LD. The adenosine A(2A) receptor agonist CGS 21680 alleviates auditory sensorimotor gating deficits and increases in accumbal CREB in rats neonatally treated with quinpirole. Psychopharmacology (Berl) 2020; 237:3519-3527. [PMID: 32772144 PMCID: PMC7686116 DOI: 10.1007/s00213-020-05631-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022]
Abstract
RATIONALE AND OBJECTIVE The adenosine A(2A) receptor forms a mutually inhibitory heteromer with the dopamine D2 receptor, and A(2A) agonists decrease D2 signaling. This study analyzed whether an adenosine A(2A) agonist would alleviate deficits in sensorimotor gating and increases in cyclic-AMP response element binding protein (CREB) in the nucleus accumbens (NAc) in the neonatal quinpirole model of schizophrenia (SZ). METHODS Male and female Sprague-Dawley rats were neonatally treated with saline (NS) or quinpirole HCl (NQ; 1 mg/kg) from postnatal days (P) 1-21. Animals were raised to P44 and behaviorally tested on auditory sensorimotor gating as measured through prepulse inhibition (PPI) from P44 to P48. Approximately 15 min before each session, animals were given an ip administration of saline or the adenosine A(2A) agonist CGS 21680 (0.03 or 0.09 mg/kg). One day after PPI was complete on P49, animals were administered a locomotor activity test in the open field after saline or CGS 21680 treatment, respectively. On P50, the nucleus accumbens (NAc) was evaluated for CREB protein. RESULTS NQ-treated rats demonstrated a deficit in PPI that was alleviated to control levels by either dose of CGS 21680. The 0.03 mg/kg dose of CGS 21680 increased startle amplitude in males. The 0.09 mg/kg dose of CGS 21680 resulted in an overall decrease in locomotor activity. NQ treatment significantly increased NAc CREB that was attenuated to control levels by either dose of CGS 21680. CONCLUSIONS This study revealed that an adenosine A(2A) receptor agonist was effective to alleviate PPI deficits in the NQ model of SZ in both male and female rats.
Collapse
Affiliation(s)
- Russell W. Brown
- Department of Biomedical Sciences, James H. Quillen College of Medicine East Tennessee State University Johnson City, TN 37614
| | - Pradeep G. Bhide
- Department of Biomedical Sciences and Neuroscience, Florida State University College of Medicine, Tallahassee, FL 32306
| | - W. Drew Gill
- Department of Biomedical Sciences, James H. Quillen College of Medicine East Tennessee State University Johnson City, TN 37614
| | - Loren D. Peeters
- Department of Biomedical Sciences, James H. Quillen College of Medicine East Tennessee State University Johnson City, TN 37614
| |
Collapse
|
45
|
Si Y, Xue X, Liu S, Feng C, Zhang H, Zhang S, Ren Y, Ma H, Dong Y, Li H, Xie L, Zhu Z. CRTC1 signaling involvement in depression-like behavior of prenatally stressed offspring rat. Behav Brain Res 2020; 399:113000. [PMID: 33161032 DOI: 10.1016/j.bbr.2020.113000] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/08/2020] [Accepted: 11/02/2020] [Indexed: 11/30/2022]
Abstract
A large body of literature has demonstrated that prenatal stress (PS) can induce depression-like behavior in the offspring. However, the underlying mechanism remains largely unknown. CREB-regulated transcriptional coactivator 1(CRTC1) has recently been shown to involve in mood regulation. This research aims to investigate whether CRTC1 signaling was involved in the depression-like behavior of prenatally stressed offspring rats. Sucrose preference test (SPT), forced swimming test (FST) and open field test (OFT) were adopted to test the depression-like behavior in the male offspring rats, and CRTC1 signaling was measured. The results showed that there were significantly reduced sucrose intake in SPT and prolonged immobility time in FST in PS-exposure offspring rats. It was also found decreased levels of total CRTC1, nuclear CRTC1, calcineurin, brain-derived neurotrophic factor (BDNF) and c-fos, but increased cytoplasmic p-CRTC1 in the hippocampus (HIP) and prefrontal cortex (PFC) of the offspring rats. Furthermore, the mRNA level of CRTC1, calcineurin, BDNF, c-fos were down-regulated. Abnormal expression of CRTC1 signaling could be alleviated by fluoxetine treatment. In conclusion, our research indicated that the aberration of CRTC1 expression and/or phosphorylation activity might play a vital role in PS-induced depression-like behavior of offspring rats.
Collapse
Affiliation(s)
- Yufang Si
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China
| | - Xing Xue
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China
| | - Si Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China
| | - Caixia Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China
| | - Huiping Zhang
- Department of Neonatology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Sisi Zhang
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China
| | - Yating Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China
| | - Hengyu Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China
| | - Yankai Dong
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China
| | - Hui Li
- Department of Neonatology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Longshan Xie
- Department of Functional Neuroscience, The First People's Hospital of Foshan (The Affiliated Foshan Hospital of Sun Yat -sen University), Foshan 528000, Guangdong, China.
| | - Zhongliang Zhu
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education. Institute of Maternal and Infant Health, Northwest University, Xi'an 710069, Shaanxi, China.
| |
Collapse
|
46
|
Abstract
An intranasal formulation of esketamine, the S enantiomer of ketamine, in conjunction with an oral antidepressant, has been approved by the FDA for treating treatment-resistant major depressive disorder (TRD) in 2019, almost 50 years after it was approved as an intravenous anesthetic. In contrast to traditional antidepressants, ketamine shows a rapid (within 2 h) and sustained (∼7 days) antidepressant effect and has significant positive effects on antisuicidal ideation. Ketamine's antidepressant mechanism is predominantly mediated by the N-methyl-d-aspartate receptor (NMDA) receptor, although NMDA-independent mechanisms are not ruled out. At the neurocircuitry level, ketamine affects the brain's reward and mood circuitry located in the corticomesolimbic structures involving the hippocampus, nucleus accumbens, and prefrontal cortex. Repurposing of ketamine for treating TRD provided a new understanding of the pathophysiology of depression, a paradigm shift from monoamine to glutamatergic neurotransmission, thus making it a unique tool to investigate the brain and its complex neurocircuitries.
Collapse
Affiliation(s)
- Joydip Das
- Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
47
|
Sex differences in adult mood and in stress-induced transcriptional coherence across mesocorticolimbic circuitry. Transl Psychiatry 2020; 10:59. [PMID: 32066699 PMCID: PMC7026087 DOI: 10.1038/s41398-020-0742-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 12/26/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
Women are approximately two times as likely to be diagnosed with major depressive disorder (MDD) compared to men. While sex differences in MDD might be driven by circulating gonadal hormones, we hypothesized that developmental hormone exposure and/or genetic sex might play a role. Mice were gonadectomized in adulthood to isolate the role of developmental hormones. We examined the effects of developmental gonadal and genetic sex on anhedonia-/depressive-like behaviors under non-stress and chronic stress conditions and performed RNA-sequencing in three mood-relevant brain regions. We used an integrative network approach to identify transcriptional modules and stress-specific hub genes regulating stress susceptibility, with a focus on whether these differed by sex. After identifying sex differences in anhedonia-/depressive-like behaviors (female > male), we show that both developmental hormone exposure (gonadal female > gonadal male) and genetic sex (XX > XY) contribute to the sex difference. The top biological pathways represented by differentially expressed genes were related to immune function; we identify which differentially expressed genes are driven by developmental gonadal or genetic sex. There was very little overlap in genes affected by chronic stress in males and females. We also identified highly co-expressed gene modules affected by stress, some of which were affected in opposite directions in males and females. Since all mice had equivalent hormone exposure in adulthood, these results suggest that sex differences in gonadal hormone exposure during sensitive developmental periods program adult sex differences in mood, and that these sex differences are independent of adult circulating gonadal hormones.
Collapse
|
48
|
Cathomas F, Murrough JW, Nestler EJ, Han MH, Russo SJ. Neurobiology of Resilience: Interface Between Mind and Body. Biol Psychiatry 2019; 86:410-420. [PMID: 31178098 PMCID: PMC6717018 DOI: 10.1016/j.biopsych.2019.04.011] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/26/2019] [Accepted: 04/05/2019] [Indexed: 12/12/2022]
Abstract
Stress-related neuropsychiatric disorders, such as major depressive disorder and posttraumatic stress disorder, exact enormous socioeconomic and individual consequences. Resilience, the process of adaptation in the face of adversity, is an important concept that is enabling the field to understand individual differences in stress responses, with the hope of harnessing this information for the development of novel therapeutics that mimic the body's natural resilience mechanisms. This review provides an update on the current state of research of the neurobiological mechanisms of stress resilience. We focus on physiological and transcriptional adaptations of specific brain circuits, the role of cellular and humoral factors of the immune system, the gut microbiota, and changes at the interface between the brain and the periphery, the blood-brain barrier. We propose viewing resilience as a process that requires the integration of multiple central and peripheral systems and that elucidating the underlying neurobiological mechanisms will ultimately lead to novel therapeutic options.
Collapse
Affiliation(s)
- Flurin Cathomas
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James W Murrough
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Mood and Anxiety Disorders Program, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eric J Nestler
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ming-Hu Han
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Scott J Russo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Center for Affective Neuroscience of the Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
49
|
Winokur SB, Lopes KL, Moparthi Y, Pereira M. Depression-related disturbances in rat maternal behaviour are associated with altered monoamine levels within mesocorticolimbic structures. J Neuroendocrinol 2019; 31:e12766. [PMID: 31265182 DOI: 10.1111/jne.12766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 06/27/2019] [Accepted: 06/27/2019] [Indexed: 12/25/2022]
Abstract
The ability of mothers to sensitively attune their maternal responses to the needs of their developing young is fundamental to a healthy mother-young relationship. The biological mechanisms that govern how mothers adjust caregiving to the dynamic changes in the demands of the young remain an open question. In the present study, we examined whether changes in monoamine levels, within discrete mesocorticolimbic structures involved in cognitive and motivational processes key to parenting, modulate this flexibility in caregiving across the postpartum period. The present study used a Wistar-Kyoto (WKY) animal model of depression and control Sprague-Dawley (SD) rats, which differ dramatically in their cognitive, motivational, and parenting performance. Levels of the monoamine neurotransmitters, dopamine, noradrenaline and serotonin, as well as their major metabolites, were measured within the medial prefrontal cortex, striatum, nucleus accumbens and medial preoptic area of SD and WKY mothers at early (postpartum day [PPD]7-8), late (PPD15-16) and weaning (PPD25) postpartum stages using high-performance liquid chromatography with electrochemical detection. Consistent with our prior work, we find that caregiving of SD mothers declined as the postpartum period progressed. Relative to nulliparous females, early postpartum mothers had lower intracellular concentrations of monoamines, as well as lower noradrenaline turnover, and an elevated serotonin turnover within most structures. Postpartum behavioural trajectories subsequently corresponded to a progressive increase in all three monoamine levels within multiple structures. Compared to SD mothers, WKY mothers were inconsistent and disorganised in caring for their offspring and exhibit profound deficits in maternal behaviour. Additionally, WKY mothers had generally lower levels of all three monoamines, as well as different patterns of change across the postpartum period, compared to SD mothers, suggesting dysfunctional central monoamine pathways in WKY mothers as they transition and experience motherhood. Taken together, the results of the present study suggest a role for monoamines at multiple mesocorticolimbic structures with repect to modulating caregiving behaviours attuned to the changing needs of the young.
Collapse
Affiliation(s)
- Sarah B Winokur
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| | - Keianna L Lopes
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| | - Yashaswani Moparthi
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| | - Mariana Pereira
- Department of Psychological and Brain Sciences, University of Massachusetts, Amherst, MA, USA
| |
Collapse
|
50
|
Marín‐Lahoz J, Sampedro F, Martinez‐Horta S, Pagonabarraga J, Kulisevsky J. Depression as a Risk Factor for Impulse Control Disorders in Parkinson Disease. Ann Neurol 2019; 86:762-769. [DOI: 10.1002/ana.25581] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/11/2019] [Accepted: 08/12/2019] [Indexed: 12/26/2022]
Affiliation(s)
- Juan Marín‐Lahoz
- Movement Disorders Unit, Neurology DepartmentHospital of Santa Creu and Sant Pau Barcelona
- Sant Pau Biomedical Research Institute Barcelona
- Neuroscience Institute, Autonomous University of Barcelona Barcelona
- Center for Biomedical Research on Neurodegenerative Diseases Madrid Spain
| | - Frederic Sampedro
- Movement Disorders Unit, Neurology DepartmentHospital of Santa Creu and Sant Pau Barcelona
- Sant Pau Biomedical Research Institute Barcelona
- Center for Biomedical Research on Neurodegenerative Diseases Madrid Spain
| | - Saül Martinez‐Horta
- Movement Disorders Unit, Neurology DepartmentHospital of Santa Creu and Sant Pau Barcelona
- Sant Pau Biomedical Research Institute Barcelona
- Center for Biomedical Research on Neurodegenerative Diseases Madrid Spain
| | - Javier Pagonabarraga
- Movement Disorders Unit, Neurology DepartmentHospital of Santa Creu and Sant Pau Barcelona
- Sant Pau Biomedical Research Institute Barcelona
- Neuroscience Institute, Autonomous University of Barcelona Barcelona
- Center for Biomedical Research on Neurodegenerative Diseases Madrid Spain
- Medicine DepartmentAutonomous University of Barcelona Barcelona
| | - Jaime Kulisevsky
- Movement Disorders Unit, Neurology DepartmentHospital of Santa Creu and Sant Pau Barcelona
- Sant Pau Biomedical Research Institute Barcelona
- Neuroscience Institute, Autonomous University of Barcelona Barcelona
- Center for Biomedical Research on Neurodegenerative Diseases Madrid Spain
- Medicine DepartmentAutonomous University of Barcelona Barcelona
| |
Collapse
|