1
|
Bisharat G, Kaganovski E, Sapir H, Temnogorod A, Levy T, Resnik J. Repeated stress gradually impairs auditory processing and perception. PLoS Biol 2025; 23:e3003012. [PMID: 39932893 PMCID: PMC11813133 DOI: 10.1371/journal.pbio.3003012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025] Open
Abstract
Repetitive stress, a common feature of modern life, is a major risk factor for psychiatric and sensory disorders. Despite the prevalence of perceptual abnormalities in these disorders, little is known about how repetitive stress affects sensory processing and perception. Here, we combine repetitive stress in mice, longitudinal measurement of cortical activity, and auditory-guided behaviors to test if sound processing and perception of neutral sounds in adults are modulated by repetitive stress. We found that repetitive stress alters sound processing, increasing spontaneous cortical activity while dampening sound-evoked responses in pyramidal and PV cells and heightening sound-evoked responses in SST cells. These alterations in auditory processing culminated in perceptual shifts, particularly a reduction in loudness perception. Additionally, our work reveals that the impact of stress on perception evolves gradually as the stressor persists over time, emphasizing the dynamic and evolving nature of this mechanism. Our findings provide insight into a possible mechanism by which repetitive stress alters sensory processing and behavior, challenging the idea that stress primarily modulates emotionally charged stimuli.
Collapse
Affiliation(s)
- Ghattas Bisharat
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ekaterina Kaganovski
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Hila Sapir
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Anita Temnogorod
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tal Levy
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jennifer Resnik
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
- Zelman Center for Brian Science Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
2
|
Tikhomirova OV. [Stress and sleep: Neurobiological aspects and modern options of insomnia therapy]. Zh Nevrol Psikhiatr Im S S Korsakova 2025; 125:14-21. [PMID: 40371851 DOI: 10.17116/jnevro202512505214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
The article discusses modern concepts of the relationship between stress and dissomnic disorders. The available literature on the possible neurobiological causes of sleep disorders under stress was reviewed, particularly on the role of changes in synthesizing neurotrophic factors in the implementation of such a relationship. Options for treating sleep disorders induced by a stressful situation are considered.
Collapse
Affiliation(s)
- O V Tikhomirova
- Nikiforov Russian Center of Emergency and Radiation Medicine, Moscow, Russia
| |
Collapse
|
3
|
Rodrigues D, Santa C, Manadas B, Monteiro P. Chronic Stress Alters Synaptic Inhibition/Excitation Balance of Pyramidal Neurons But Not PV Interneurons in the Infralimbic and Prelimbic Cortices of C57BL/6J Mice. eNeuro 2024; 11:ENEURO.0053-24.2024. [PMID: 39147579 DOI: 10.1523/eneuro.0053-24.2024] [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: 02/05/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/17/2024] Open
Abstract
The medial prefrontal cortex (mPFC) plays a pivotal role in regulating working memory, executive function, and self-regulatory behaviors. Dysfunction in the mPFC circuits is a characteristic feature of several neuropsychiatric disorders including schizophrenia, depression, and post-traumatic stress disorder. Chronic stress (CS) is widely recognized as a major triggering factor for the onset of these disorders. Although evidence suggests synaptic dysfunction in mPFC circuits following CS exposure, it remains unclear how different neuronal populations in the infralimbic (IL) and prelimbic (PL) cortices are affected in terms of synaptic inhibition/excitation balance (I/E ratio). Here, using neuroproteomic analysis and whole-cell patch-clamp recordings in pyramidal neurons (PNs) and parvalbumin (PV) interneurons within the PL and IL cortices, we examined the synaptic changes after 21 d of chronic unpredictable stress, in male mice. Our results reveal distinct impacts of CS on PL and IL PNs, resulting in an increased I/E ratio in both subregions but through different mechanisms: CS increases inhibitory synaptic drive in the PL while decreasing excitatory synaptic drive in the IL. Notably, the I/E ratio and excitatory and inhibitory synaptic drive of PV interneurons remained unaffected in both PL and IL circuits following CS exposure. These findings offer novel mechanistic insights into the influence of CS on mPFC circuits and support the hypothesis of stress-induced mPFC hypofunction.
Collapse
Affiliation(s)
- Diana Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga 4710-057, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Braga 4710-057, Portugal
- Biomedizinisches Centrum München (BMC), Ludwig-Maximilians-Universität München, Munich 82152, Bayern, Germany
| | - Cátia Santa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra 3004-517, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3004-517, Portugal
| | - Patrícia Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga 4710-057, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimaraes, Braga 4710-057, Portugal
- Department of Biomedicine - Experimental Biology Unit, Faculty of Medicine, University of Porto, Porto 4200-319, Portugal
- RISE-Health, Health Research Network, Porto 4200-319, Portugal
| |
Collapse
|
4
|
Canto-de-Souza L, Baptista-de-Souza D, Nunes-de-Souza RL, Planeta C. Distinct roles of the left and right prelimbic cortices in the modulation of ethanol consumption in male mice under acute and chronic social defeat stress. Psychopharmacology (Berl) 2024; 241:1161-1176. [PMID: 38347153 DOI: 10.1007/s00213-024-06550-8] [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: 11/01/2023] [Accepted: 01/31/2024] [Indexed: 05/21/2024]
Abstract
RATIONALE Chronic stress exposure disrupts the medial prefrontal cortex's (mPFC) ability to regulate impulses, leading to the loss of control over alcohol drinking in rodents, emphasizing the critical role of this forebrain area in regulating alcohol consumption. Moreover, chronic stress exposure causes lateralization of mPFC functions with volumetric and functional changes, resulting in hyperactivity in the right hemisphere and functional decrease in the left. OBJECTIVES This study investigated the inhibitory role of the left prelimbic cortex (LPrL) on ethanol consumption induced by chronic social defeat stress (SDS) in male mice and to examine if inactivation of the LPrL causes disinhibition of the right mPFC, leading to an increase in ethanol consumption. We also investigated the role of lateralization and neurochemical alterations in the mPFC related to ethanol consumption induced by chronic SDS. To this end, we examined the activation patterns of ΔFosB, VGLUT2, and GAD67 in the left and right mPFC. RESULTS Temporarily blocking the LPrL or right PrL (RPrL) cortices during acute SDS did not affect male mice's voluntary ethanol consumption in male mice. When each cortex was blocked in mice previously exposed to chronic SDS, ethanol consumption also remained unaffected. However, male mice with LPrL lesions during chronic SDS showed an increase in voluntary ethanol consumption, which was associated with enhanced ΔFosB/VGLUT2-positive neurons within the RPrL cortex. CONCLUSIONS The results suggest that the LPrL may play a role in inhibiting ethanol consumption induced by chronic SDS, while the RPrL may be involved in the disinhibition of ethanol consumption.
Collapse
Affiliation(s)
- Lucas Canto-de-Souza
- Lab. Pharmacology, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, SP, 14800-903, Brazil
| | - Daniela Baptista-de-Souza
- Lab. Pharmacology, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, SP, 14800-903, Brazil
| | - Ricardo Luiz Nunes-de-Souza
- Lab. Pharmacology, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, SP, 14800-903, Brazil
- Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, SP, 13565-905, Brazil
| | - Cleopatra Planeta
- Lab. Pharmacology, School of Pharmaceutical Sciences, São Paulo State University - UNESP, Araraquara, SP, 14800-903, Brazil.
- Joint Graduate Program in Physiological Sciences UFSCar/UNESP, São Carlos, SP, 13565-905, Brazil.
| |
Collapse
|
5
|
Yin YY, Yan JZ, Lai SX, Wei QQ, Sun SR, Zhang LM, Li YF. Gamma oscillations in the mPFC: A potential predictive biomarker of depression and antidepressant effects. Prog Neuropsychopharmacol Biol Psychiatry 2024; 129:110893. [PMID: 37949392 DOI: 10.1016/j.pnpbp.2023.110893] [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: 08/21/2023] [Revised: 11/06/2023] [Accepted: 11/06/2023] [Indexed: 11/12/2023]
Abstract
Gamma oscillations have attracted much attention in the field of mood disorders, but their role in depression remains poorly understood. This study aimed to investigate whether gamma oscillations in the medial prefrontal cortex (mPFC) could serve as a predictive biomarker of depression. Chronic restraint stress (CRS) or lipopolysaccharide (LPS) were used to induce depression-like behaviors in mice; local field potentials (LFPs) in the mPFC were recorded by electrophysiological techniques; We found that both CRS and LPS induced significant depression-like behaviors in mice, including increasing immobility durations in the forced swimming test (FST) and tail suspension test (TST) and increasing the latency to feed in the novelty-suppressed feeding test (NSFT). Electrophysiological results suggested that CRS and LPS significantly reduced low and high gamma oscillations in the mPFC. Furthermore, a single injection of ketamine or scopolamine for 24 h significantly increased gamma oscillations and elicited rapid-acting antidepressant-like effects. In addition, fluoxetine treatment for 21 days significantly increased gamma oscillations and elicited antidepressant-like effects. Taken together, our findings suggest that gamma oscillations are strongly associated with depression, yielding new insights into investigating the predictive biomarkers of depression and the time course of antidepressant effects.
Collapse
Affiliation(s)
- Yong-Yu Yin
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China.
| | - Jiao-Zhao Yan
- Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Shi-Xin Lai
- School of Medicine, Sun Yat-Sen University, Shenzhen campus, Shenzhen, China
| | - Qian-Qian Wei
- School of Medicine, Nantong University, Nantong, China
| | - Si-Rui Sun
- Beijing Ditan Hospital Capital Medical University, Beijing, China
| | - Li-Ming Zhang
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China
| | - Yun-Feng Li
- Beijing Institute of Pharmacology and Toxicology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Key Laboratory of Neuropsychopharmacology, Beijing, China; Beijing Institute of Basic Medical Sciences, Beijing, China.
| |
Collapse
|
6
|
Goikolea-Vives A, Fernandes C, Thomas MSC, Thornton C, Stolp HB. Sex-Specific Behavioural Deficits in Adulthood following Acute Activation of the GABAA Receptor in the Neonatal Mouse. Dev Neurosci 2024; 46:386-400. [PMID: 38325353 DOI: 10.1159/000536641] [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: 06/23/2023] [Accepted: 01/26/2024] [Indexed: 02/09/2024] Open
Abstract
INTRODUCTION Sex differences exist in the prevalence of neurodevelopmental disorders (NDDs). Part of the aetiology of NDDs has been proposed to be alterations in the balance between excitatory and inhibitory neurotransmission, leading to the question of whether males and females respond differently to altered neurotransmitter balance. We investigated whether pharmacological alteration of GABAA signalling in early development results in sex-dependent changes in adult behaviours associated with NDDs. METHODS Male and female C57BL/6J mice received intraperitoneal injections of 0.5 mg/kg muscimol or saline on postnatal days (P) 3-5 and were subjected to behavioural testing, specifically open field, light/dark box, marble-burying, sucralose preference, social interaction, and olfactory habituation/dishabituation tests between P60 and P90. RESULTS Early postnatal administration of muscimol resulted in reduced anxiety in the light/dark box test in both male and female adult mice. Muscimol reduced sucralose preference in males, but not females, whereas female mice showed reduced social behaviours. Regional alterations in cortical thickness were observed in the weeks following GABAA receptor activation, pointing to an evolving structural difference in the brain underlying adult behaviour. CONCLUSIONS We conclude that activation of the GABAA receptor in the first week of life resulted in long-lasting changes in a range of behaviours in adulthood following altered neurodevelopment. Sex of the individual affected the nature and severity of these abnormalities, explaining part of the varied pathophysiology and neurodevelopmental diagnosis that derive from excitatory/inhibitory imbalance.
Collapse
Affiliation(s)
- Ane Goikolea-Vives
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Cathy Fernandes
- SGDP Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Michael S C Thomas
- Centre for Brain and Cognitive Development, Birkbeck College, University of London, London, UK
| | - Claire Thornton
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Helen B Stolp
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| |
Collapse
|
7
|
Leng L, Zhuang K, Lin H, Ding J, Yang S, Yuan Z, Huang C, Chen G, Chen Z, Wang M, Wang H, Sun H, Li H, Chang H, Chen Z, Xu Q, Yuan T, Zhang J. Menin Reduces Parvalbumin Expression and is Required for the Anti-Depressant Function of Ketamine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305659. [PMID: 38044302 PMCID: PMC10837338 DOI: 10.1002/advs.202305659] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 10/23/2023] [Indexed: 12/05/2023]
Abstract
Dysfunction of parvalbumin (PV) neurons is closely involved in depression, however, the detailed mechanism remains unclear. Based on the previous finding that multiple endocrine neoplasia type 1 (Protein: Menin; Gene: Men1) mutation (G503D) is associated with a higher risk of depression, a Menin-G503D mouse model is generated that exhibits heritable depressive-like phenotypes and increases PV expression in brain. This study generates and screens a serial of neuronal specific Men1 deletion mice, and found that PV interneuron Men1 deletion mice (PcKO) exhibit increased cortical PV levels and depressive-like behaviors. Restoration of Menin, knockdown PV expression or inhibition of PV neuronal activity in PV neurons all can ameliorate the depressive-like behaviors of PcKO mice. This study next found that ketamine stabilizes Menin by inhibiting protein kinase A (PKA) activity, which mediates the anti-depressant function of ketamine. These results demonstrate a critical role for Menin in depression, and prove that Menin is key to the antidepressant function of ketamine.
Collapse
Affiliation(s)
- Lige Leng
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Kai Zhuang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Hui Lin
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Jinjun Ding
- Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghai200030P. R. China
| | - Shangchen Yang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Ziqi Yuan
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Changquan Huang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Guimiao Chen
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Zhenlei Chen
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Mengdan Wang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Han Wang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Hao Sun
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Huifang Li
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - He Chang
- Department of GeriatricsXiang'an Hospital of Xiamen universityXiamenFujian361102P. R. China
| | - Zhenyi Chen
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| | - Qi Xu
- State Key Laboratory of Medical Molecular BiologyInstitute of Basic Medical Sciences Chinese Academy of Medical Sciences and Peking Union Medical CollegeNeuroscience CenterChinese Academy of Medical SciencesBeijing100730P. R. China
| | - Tifei Yuan
- Shanghai Mental Health CenterShanghai Jiaotong University School of MedicineShanghai200030P. R. China
| | - Jie Zhang
- Institute of NeuroscienceDepartment of AnesthesiologyThe First Affiliated Hospital of Xiamen UniversitySchool of MedicineXiamen UniversityXiamenFujian361102P. R. China
| |
Collapse
|
8
|
Tse WS, Pochwat B, Szewczyk B, Misztak P, Bobula B, Tokarski K, Worch R, Czarnota-Bojarska M, Lipton SA, Zaręba-Kozioł M, Bijata M, Wlodarczyk J. Restorative effect of NitroSynapsin on synaptic plasticity in an animal model of depression. Neuropharmacology 2023; 241:109729. [PMID: 37797736 DOI: 10.1016/j.neuropharm.2023.109729] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/21/2023] [Accepted: 09/23/2023] [Indexed: 10/07/2023]
Abstract
In the search for new options for the pharmacological treatment of major depressive disorder, compounds with a rapid onset of action and high efficacy but lacking a psychotomimetic effect are of particular interest. In the present study, we evaluated the antidepressant potential of NitroSynapsin (NS) at behavioural, structural, and functional levels. NS is a memantine derivative and a dual allosteric N-methyl-d-aspartate receptors (NMDAR) antagonist using targeted delivery by the aminoadamantane of a warhead nitro group to inhibitory redox sites on the NMDAR. In a chronic restraint stress (CRS) mouse model of depression, five doses of NS administered on three consecutive days evoked antidepressant-like activity in the chronically stressed male C57BL/6J mice, reversing CRS-induced behavioural disturbances in sucrose preference and tail suspension tests. CRS-induced changes in morphology and density of dendritic spines in cerebrocortical neurons in the medial prefrontal cortex (mPFC) were also reversed by NS. Moreover, CRS-induced reduction in long-term potentiation (LTP) in the mPFC was found to be prevented by NS based on the electrophysiological recordings. Our study showed that NS restores structural and functional synaptic plasticity and reduces depressive behaviour to the level found in naïve animals. These results preliminarily revealed an antidepressant-like potency of NS.
Collapse
Affiliation(s)
- Wing Sze Tse
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093 Warsaw, Poland
| | - Bartłomiej Pochwat
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093 Warsaw, Poland; Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland
| | - Bernadeta Szewczyk
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland
| | - Paulina Misztak
- Department of Neurobiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland; Department of Medicine and Surgery, University of Milano-Bicocca, 20-900, Monza, Italy
| | - Bartosz Bobula
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland
| | - Krzysztof Tokarski
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, 31-343 Krakow, Poland
| | - Remigiusz Worch
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093 Warsaw, Poland
| | - Marta Czarnota-Bojarska
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093 Warsaw, Poland
| | - Stuart A Lipton
- Neurodegeneration New Medicines Center and Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA 92037, United States; Department of Neurosciences, University of California, School of Medicine, La Jolla, San Diego, CA 92093, United States
| | - Monika Zaręba-Kozioł
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093 Warsaw, Poland
| | - Monika Bijata
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093 Warsaw, Poland.
| | - Jakub Wlodarczyk
- Laboratory of Cell Biophysics, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteur Str. 3, 02-093 Warsaw, Poland.
| |
Collapse
|
9
|
Ayala-Rodríguez JD, García-Colunga J. Maternal separation modifies spontaneous synaptic activity in the infralimbic cortex of stress-resilient male rats. PLoS One 2023; 18:e0294151. [PMID: 37943747 PMCID: PMC10635473 DOI: 10.1371/journal.pone.0294151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
Glutamate and GABA signaling systems are necessary to maintain proper function of the central nervous system through excitation/inhibition (E/I) balance. Alteration of this balance in the medial prefrontal cortex (mPFC), as an effect of early-life stress, may lead to the development of anxiety and depressive disorders. Few studies exist in the infralimbic division of the mPFC to understand the effect of early-life stress at different ages, which is the purpose of the present work. Newborn Sprague Dawley male rats were subjected to maternal separation (MS) for two weeks. First, tests measuring anxiety- and depression-like behaviors were performed on adolescent and adult rats subjected to MS (MS-rats). Then, to establish a relationship with behavioral results, electrophysiological recordings were performed in neurons of the infralimbic cortex in acute brain slices of infant, adolescent, and adult rats. In the behavioral tests, there were no significant differences in MS-rats compared to control rats at any age. Moreover, MS had no effect on the passive membrane properties nor neuronal excitability in the infralimbic cortex, whereas spontaneous synaptic activity in infralimbic neurons was altered. The frequency of spontaneous glutamatergic synaptic events increased in infant MS-rats, whereas in adolescent MS-rats both the frequency and the amplitude of spontaneous GABAergic events increased without any effect on glutamatergic synaptic responses. In adult MS-rats, these two parameters decreased in spontaneous GABAergic synaptic events, whereas only the frequency of glutamatergic events decreased. These data suggest that rats subjected to MS did not exhibit behavioral changes and presented an age-dependent E/I imbalance in the infralimbic cortex, possibly due to differential changes in neurotransmitter release and/or receptor expression.
Collapse
Affiliation(s)
- Jesús David Ayala-Rodríguez
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| | - Jesús García-Colunga
- Departamento de Neurobiología Celular y Molecular, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Juriquilla, Querétaro, México
| |
Collapse
|
10
|
Toyoda H, Fujinami Y, Saito M, Maeda Y, Kang Y. Increased vertical dimension of occlusion for varying periods differentially impairs learning and memory in guinea pigs. Behav Brain Res 2023; 452:114547. [PMID: 37331607 DOI: 10.1016/j.bbr.2023.114547] [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: 02/24/2023] [Revised: 06/07/2023] [Accepted: 06/15/2023] [Indexed: 06/20/2023]
Abstract
There is an increasing number of studies showing that occlusal dysfunction impairs learning and memory. We previously demonstrated that the brain has a mechanism to calibrate between the activities of spindle afferents and periodontal-mechanoreceptor afferents for controlling the chewing movement, and the accurate calibration can be done only at the proper vertical dimension of occlusion (VDO). Then, the chewing at an inappropriate VDO may induce a severe mental stress due to a mal-calibration. However, it is not clear how the impairment of learning/memory progresses over the period of stress induced by occlusal dysfunction. We investigated by passive avoidance test how the behavior and learning/memory are altered in guinea pigs in which the VDO was raised by 2-3 mm over the period up to 8 weeks. We found that the guinea pigs reared under the raised occlusal-condition (ROC) for 1 week showed a very high sensitivity to electrical stimulation whereas this did not cause the memory consolidation in the 1st-day retention trial, suggesting that such hypersensitivity rather hampered the fear learning. In the guinea pigs reared under the ROC for 2 and 8 weeks, the learning ability was not largely affected and memory consolidation occurred similarly whereas the memory retention deteriorated more severely in the latter guinea pigs than in the former ones. In the guinea pigs reared under the ROC for 3 and 4 weeks, learning was severely impaired, and memory consolidation did not occur. These results suggest that the occlusal dysfunction for varying periods differentially impairs learning and memory.
Collapse
Affiliation(s)
- Hiroki Toyoda
- Department of Neuroscience and Oral Physiology, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan.
| | - Yozo Fujinami
- Department of Neuroscience and Oral Physiology, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan; Department of Prosthodontics, Gerodontology and Oral Rehabilitation, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan
| | - Mitsuru Saito
- Department of Oral Physiology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Yoshinobu Maeda
- Department of Prosthodontics, Gerodontology and Oral Rehabilitation, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan
| | - Youngnam Kang
- Department of Neuroscience and Oral Physiology, Osaka University Graduate School of Dentistry, Suita, Osaka 565-0871, Japan; Department of Behavioral Sciences, Graduate School of Human Sciences, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
11
|
Wallace T, Myers B. Prefrontal representation of affective stimuli: importance of stress, sex, and context. Cereb Cortex 2023; 33:8232-8246. [PMID: 37032618 PMCID: PMC10321111 DOI: 10.1093/cercor/bhad110] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 04/11/2023] Open
Abstract
Stress-related disorders such as depression and anxiety exhibit sex differences in prevalence and negatively impact both mental and physical health. Affective illness is also frequently accompanied by changes in ventromedial prefrontal cortical (vmPFC) function. However, the neurobiology that underlies sex-specific cortical processing of affective stimuli is poorly understood. Although rodent studies have investigated the prefrontal impact of chronic stress, postmortem studies have focused largely on males and yielded mixed results. Therefore, genetically defined population recordings in behaving animals of both sexes were used to test the hypothesis that chronic variable stress (CVS) impairs the neural processing of affective stimuli in the rodent infralimbic region. Here, we targeted expression of a calcium indicator, GCaMP6s, to infralimbic pyramidal cells. In males, CVS reduced infralimbic responses to social interaction and restraint stress but increased responses to novel objects and food reward. In contrast, females did not have CVS-induced changes in infralimbic activity, which was partially dependent on the ovarian status. These results indicate that both male and female vmPFC cells encode social, stress, and reward stimuli. However, chronic stress effects are sex-dependent and behavior-specific. Ultimately, these findings extend the understanding of chronic stress-induced prefrontal dysfunction and indicate that sex is a critical factor for cortical processing of affective stimuli.
Collapse
Affiliation(s)
- Tyler Wallace
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Brent Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
12
|
Cooper MA, Grizzell JA, Whitten CJ, Burghardt GM. Comparing the ontogeny, neurobiology, and function of social play in hamsters and rats. Neurosci Biobehav Rev 2023; 147:105102. [PMID: 36804399 PMCID: PMC10023430 DOI: 10.1016/j.neubiorev.2023.105102] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/26/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Syrian hamsters show complex social play behavior and provide a valuable animal model for delineating the neurobiological mechanisms and functions of social play. In this review, we compare social play behavior of hamsters and rats and underlying neurobiological mechanisms. Juvenile rats play by competing for opportunities to pin one another and attack their partner's neck. A broad set of cortical, limbic, and striatal regions regulate the display of social play in rats. In hamsters, social play is characterized by attacks to the head in early puberty, which gradually transitions to the flanks in late puberty. The transition from juvenile social play to adult hamster aggression corresponds with engagement of neural ensembles controlling aggression. Play deprivation in rats and hamsters alters dendritic morphology in mPFC neurons and impairs flexible, context-dependent behavior in adulthood, which suggests these animals may have converged on a similar function for social play. Overall, dissecting the neurobiology of social play in hamsters and rats can provide a valuable comparative approach for evaluating the function of social play.
Collapse
Affiliation(s)
- Matthew A Cooper
- Department of Psychology, University of Tennessee Knoxville, Knoxville, TN, USA.
| | - J Alex Grizzell
- Neuroscience and Behavioral Biology, Emory University, Atlanta, GA, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Conner J Whitten
- Department of Psychology, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Gordon M Burghardt
- Department of Psychology, University of Tennessee Knoxville, Knoxville, TN, USA; Department of Ecology & Evolutionary Biology, University of Tennessee Knoxville, Knoxville, TN, USA
| |
Collapse
|
13
|
Woodward E, Rangel-Barajas C, Ringland A, Logrip ML, Coutellier L. Sex-Specific Timelines for Adaptations of Prefrontal Parvalbumin Neurons in Response to Stress and Changes in Anxiety- and Depressive-Like Behaviors. eNeuro 2023; 10:ENEURO.0300-22.2023. [PMID: 36808099 PMCID: PMC9997696 DOI: 10.1523/eneuro.0300-22.2023] [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] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 02/22/2023] Open
Abstract
Women are twice as likely as men to experience emotional dysregulation after stress, resulting in substantially higher psychopathology for equivalent lifetime stress exposure, yet the mechanisms underlying this vulnerability remain unknown. Studies suggest changes in medial prefrontal cortex (mPFC) activity as a potential contributor. Whether maladaptive changes in inhibitory interneurons participate in this process, and whether adaptations in response to stress differ between men and women, producing sex-specific changes in emotional behaviors and mPFC activity, remained undetermined. This study examined whether unpredictable chronic mild stress (UCMS) in mice differentially alters behavior and mPFC parvalbumin (PV) interneuron activity by sex, and whether the activity of these neurons drives sex-specific behavioral changes. Four weeks of UCMS increased anxiety-like and depressive-like behaviors associated with FosB activation in mPFC PV neurons, particularly in females. After 8 weeks of UCMS, both sexes displayed these behavioral and neural changes. Chemogenetic activation of PV neurons in UCMS-exposed and nonstressed males induced significant changes in anxiety-like behaviors. Importantly, patch-clamp electrophysiology demonstrated altered excitability and basic neural properties on the same timeline as the emergence of behavioral effects: changes in females after 4 weeks and in males after 8 weeks of UCMS. These findings show, for the first time, that sex-specific changes in the excitability of prefrontal PV neurons parallel the emergence of anxiety-like behavior, revealing a potential novel mechanism underlying the enhanced vulnerability of females to stress-induced psychopathology and supporting further investigation of this neuronal population to identify new therapeutic targets for stress disorders.
Collapse
Affiliation(s)
- Emma Woodward
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
| | - Claudia Rangel-Barajas
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202
| | - Amanda Ringland
- Department of Psychology, The Ohio State University, Columbus, Ohio 43210
| | - Marian L Logrip
- Department of Psychology, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Laurence Coutellier
- Department of Neuroscience, The Ohio State University, Columbus, Ohio 43210
- Department of Psychology, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
14
|
Witztum J, Singh A, Zhang R, Johnson M, Liston C. An automated platform for Assessing Working Memory and prefrontal circuit function. Neurobiol Stress 2023; 24:100518. [PMID: 36970451 PMCID: PMC10033752 DOI: 10.1016/j.ynstr.2023.100518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 01/02/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Working memory is a process for actively maintaining and updating task-relevant information, despite interference from competing inputs, and is supported in part by sustained activity in prefrontal cortical pyramidal neurons and coordinated interactions with inhibitory interneurons, which may serve to regulate interference. Chronic stress has potent effects on working memory performance, possibly by interfering with these interactions or by disrupting long-range inputs from key upstream brain regions. Still, the mechanisms by which chronic stress disrupts working memory are not well understood, due in part to a need for scalable, easy-to-implement behavioral assays that are compatible with two-photon calcium imaging and other tools for recording from large populations of neurons. Here, we describe the development and validation of a platform that was designed specifically for automated, high-throughput assessments of working memory and simultaneous two-photon imaging in chronic stress studies. This platform is relatively inexpensive and easy to build; fully automated and scalable such that one investigator can test relatively large cohorts of animals concurrently; fully compatible with two-photon imaging, yet also designed to mitigate head-fixation stress; and can be easily adapted for other behavioral paradigms. Our validation data confirm that mice could be trained to perform a delayed response working memory task with relatively high-fidelity over the course of ∼15 days. Two-photon imaging data validate the feasibility of recording from large populations of cells during working memory tasks performance and characterizing their functional properties. Activity patterns in >70% of medial prefrontal cortical neurons were modulated by at least one task feature, and a majority of cells were engaged by multiple task features. We conclude with a brief literature review of the circuit mechanisms supporting working memory and their disruption in chronic stress states-highlighting directions for future research enabled by this platform.
Collapse
|
15
|
Rosen JB, Schulkin J. Hyperexcitability: From Normal Fear to Pathological Anxiety and Trauma. Front Syst Neurosci 2022; 16:727054. [PMID: 35993088 PMCID: PMC9387392 DOI: 10.3389/fnsys.2022.727054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 06/20/2022] [Indexed: 11/16/2022] Open
Abstract
Hyperexcitability in fear circuits is suggested to be important for development of pathological anxiety and trauma from adaptive mechanisms of fear. Hyperexcitability is proposed to be due to acquired sensitization in fear circuits that progressively becomes more severe over time causing changing symptoms in early and late pathology. We use the metaphor and mechanisms of kindling to examine gains and losses in function of one excitatory and one inhibitory neuropeptide, corticotrophin releasing factor and somatostatin, respectively, to explore this sensitization hypothesis. We suggest amygdala kindling induced hyperexcitability, hyper-inhibition and loss of inhibition provide clues to mechanisms for hyperexcitability and progressive changes in function initiated by stress and trauma.
Collapse
Affiliation(s)
- Jeffrey B. Rosen
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
- *Correspondence: Jeffrey B. Rosen,
| | - Jay Schulkin
- School of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
16
|
Palagini L, Bianchini C. Pharmacotherapeutic management of insomnia and effects on sleep processes, neural plasticity, and brain systems modulating stress: A narrative review. Front Neurosci 2022; 16:893015. [PMID: 35968380 PMCID: PMC9374363 DOI: 10.3389/fnins.2022.893015] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionInsomnia is a stress-related sleep disorder, may favor a state of allostatic overload impairing brain neuroplasticity, stress immune and endocrine pathways, and may contribute to mental and physical disorders. In this framework, assessing and targeting insomnia is of importance.AimSince maladaptive neuroplasticity and allostatic overload are hypothesized to be related to GABAergic alterations, compounds targeting GABA may play a key role. Accordingly, the aim of this review was to discuss the effect of GABAA receptor agonists, short-medium acting hypnotic benzodiazepines and the so called Z-drugs, at a molecular level.MethodLiterature searches were done according to PRISMA guidelines. Several combinations of terms were used such as “hypnotic benzodiazepines” or “brotizolam,” or “lormetazepam” or “temazepam” or “triazolam” or “zolpidem” or “zopiclone” or “zaleplon” or “eszopiclone” and “insomnia” and “effects on sleep” and “effect on brain plasticity” and “effect on stress system”. Given the complexity and heterogeneity of existing literature, we ended up with a narrative review.ResultsAmong short-medium acting compounds, triazolam has been the most studied and may regulate the stress system at central and peripheral levels. Among Z-drugs eszopiclone may regulate the stress system. Some compounds may produce more “physiological” sleep such as brotizolam, triazolam, and eszopiclone and probably may not impair sleep processes and related neural plasticity. In particular, triazolam, eszopiclone, and zaleplon studied in vivo in animal models did not alter neuroplasticity.ConclusionCurrent models of insomnia may lead us to revise the way in which we use hypnotic compounds in clinical practice. Specifically, compounds should target sleep processes, the stress system, and sustain neural plasticity. In this framework, among the short/medium acting hypnotic benzodiazepines, triazolam has been the most studied compound while among the Z-drugs eszopiclone has demonstrated interesting effects. Both offer potential new insight for treating insomnia.
Collapse
Affiliation(s)
- Laura Palagini
- Psychiatry Division, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- *Correspondence: Laura Palagini,
| | | |
Collapse
|
17
|
Hartwig M, Bhat A, Peters A. How Stress Can Change Our Deepest Preferences: Stress Habituation Explained Using the Free Energy Principle. Front Psychol 2022; 13:865203. [PMID: 35712161 PMCID: PMC9195169 DOI: 10.3389/fpsyg.2022.865203] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/04/2022] [Indexed: 12/28/2022] Open
Abstract
People who habituate to stress show a repetition-induced response attenuation—neuroendocrine, cardiovascular, neuroenergetic, and emotional—when exposed to a threatening environment. But the exact dynamics underlying stress habituation remain obscure. The free energy principle offers a unifying account of self-organising systems such as the human brain. In this paper, we elaborate on how stress habituation can be explained and modelled using the free energy principle. We introduce habituation priors that encode the agent’s tendency for stress habituation and incorporate them in the agent’s decision-making process. Using differently shaped goal priors—that encode the agent’s goal preferences—we illustrate, in two examples, the optimising (and thus habituating) behaviour of agents. We show that habituation minimises free energy by reducing the precision (inverse variance) of goal preferences. Reducing the precision of goal priors means that the agent accepts adverse (previously unconscionable) states (e.g., lower social status and poverty). Acceptance or tolerance of adverse outcomes may explain why habituation causes people to exhibit an attenuation of the stress response. Given that stress habituation occurs in brain regions where goal priors are encoded, i.e., in the ventromedial prefrontal cortex and that these priors are encoded as sufficient statistics of probability distributions, our approach seems plausible from an anatomical-functional and neuro-statistical point of view. The ensuing formal and generalisable account—based on the free energy principle—further motivate our novel treatment of stress habituation. Our analysis suggests that stress habituation has far-reaching consequences, protecting against the harmful effects of toxic stress, but on the other hand making the acceptability of precarious living conditions and the development of the obese type 2 diabetes mellitus phenotype more likely.
Collapse
Affiliation(s)
- Mattis Hartwig
- German Research Center for Artificial Intelligence (DFKI), Lübeck, Germany
- singularIT GmbH, Leipzig, Germany
| | - Anjali Bhat
- Wellcome Centre for Human Neuroimaging, University College London, London, United Kingdom
| | - Achim Peters
- Medical Clinic 1, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
- *Correspondence: Achim Peters,
| |
Collapse
|
18
|
Impact of stress on inhibitory neuronal circuits, our tribute to Bruce McEwen. Neurobiol Stress 2022; 19:100460. [PMID: 35734023 PMCID: PMC9207718 DOI: 10.1016/j.ynstr.2022.100460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/22/2022] [Accepted: 05/10/2022] [Indexed: 12/03/2022] Open
Abstract
This manuscript is dedicated to the memory of Bruce S. McEwen, to commemorate the impact he had on how we understand stress and neuronal plasticity, and the profound influence he exerted on our scientific careers. The focus of this review is the impact of stressors on inhibitory circuits, particularly those of the limbic system, but we also consider other regions affected by these adverse experiences. We revise the effects of acute and chronic stress during different stages of development and lifespan, taking into account the influence of the sex of the animals. We review first the influence of stress on the physiology of inhibitory neurons and on the expression of molecules related directly to GABAergic neurotransmission, and then focus on specific interneuron subpopulations, particularly on parvalbumin and somatostatin expressing cells. Then we analyze the effects of stress on molecules and structures related to the plasticity of inhibitory neurons: the polysialylated form of the neural cell adhesion molecule and perineuronal nets. Finally, we review the potential of antidepressants or environmental manipulations to revert the effects of stress on inhibitory circuits.
Collapse
|
19
|
Palamarchuk IS, Vaillancourt T. Integrative Brain Dynamics in Childhood Bullying Victimization: Cognitive and Emotional Convergence Associated With Stress Psychopathology. Front Integr Neurosci 2022; 16:782154. [PMID: 35573445 PMCID: PMC9097078 DOI: 10.3389/fnint.2022.782154] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/15/2022] [Indexed: 12/14/2022] Open
Abstract
Bullying victimization is a form of psychological stress that is associated with poor outcomes in the areas of mental health and learning. Although the emotional maladjustment and memory impairment following interpersonal stress are well documented, the mechanisms of complex cerebral dysfunctions have neither been outlined nor studied in depth in the context of childhood bullying victimization. As a contribution to the cross-disciplinary field of developmental psychology and neuroscience, we review the neuropathophysiology of early life stress, as well as general psychological stress to synthesize the data and clarify the versatile dynamics within neuronal networks linked to bullying victimization. The stress-induced neuropsychological cascade and associated cerebral networks with a focus on cognitive and emotional convergence are described. The main findings are that stress-evoked neuroendocrine reactivity relates to neuromodulation and limbic dysregulation that hinder emotion processing and executive functioning such as semantic cognition, cognitive flexibility, and learning. Developmental aspects and interacting neural mechanisms linked to distressed cognitive and emotional processing are pinpointed and potential theory-of-mind nuances in targets of bullying are presented. The results show that childhood stress psychopathology is associated with a complex interplay where the major role belongs to, but is not limited to, the amygdala, fusiform gyrus, insula, striatum, and prefrontal cortex. This interplay contributes to the sensitivity toward facial expressions, poor cognitive reasoning, and distress that affect behavioral modulation and emotion regulation. We integrate the data on major brain dynamics in stress neuroactivity that can be associated with childhood psychopathology to help inform future studies that are focused on the treatment and prevention of psychiatric disorders and learning problems in bullied children and adolescents.
Collapse
|
20
|
Pesarico AP, Carceller H, Guirado R, Coviello S, Nacher J. Long term effects of 24-h-restraint stress on the connectivity and structure of interneurons in the basolateral amygdala. Prog Neuropsychopharmacol Biol Psychiatry 2022; 115:110512. [PMID: 35066055 DOI: 10.1016/j.pnpbp.2022.110512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/15/2022]
Abstract
The effects of intense stressors can last a long time and may lead to the development of psychiatric disorders, including posttraumatic stress disorder. The basolateral amygdala (BLA) plays a critical role in these diseases and is extremely sensitive to stress. Here, we explored in male and female mice the long-term (35 days) impact of a 24-h restraint stress on BLA circuitry. We used Thy1-YFP mice to discriminate 2 subpopulations of excitatory neurons, which participate in "Fear-On" (Thy1-) and "Fear-Off" (Thy1+) circuits. The stress decreased the density of parvalbumin (PV) + inhibitory neurons in both sexes but did not alter their dendritic complexity. We also analyzed the perisomatic input of basket interneurons on Thy1+ and Thy1- neurons, finding sex dependent effects. In males, we did not find alterations in the density of PV+ puncta or in that of cannabinoid receptor 1 (CB1R) + puncta from cholecystokinin+ basket cells. By contrast, in females we found increased the density of PV+ puncta on Thy1+ neurons and reduced on the Thy1- neurons. This adverse experience also reduced in the long term the density of CB1R+ puncta both on Thy1+ and Thy1- cells in females. The expression of the activity marker FosB was not altered in PV+ interneurons and in Thy1+ neurons of stressed animals. The density of perineuronal nets, a specialized region of the extracellular matrix, which covers particularly PV+ interneurons and regulates their connectivity, was increased by stress in male mice. Our findings indicate that a single stressful event can produce long-term alterations in the inhibitory circuits of the BLA, especially on PV+ neurons and their plasticity, and that there is a differential impact depending on the sex and the fear-related circuits involved.
Collapse
Affiliation(s)
- Ana Paula Pesarico
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Hector Carceller
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain.
| | - Ramón Guirado
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Simona Coviello
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain
| | - Juan Nacher
- Neurobiology Unit, Program in Neurosciences and Institute of Biotechnology and Biomedicine (BIOTECMED), Universitat de València, Burjassot, Spain; Spanish National Network for Research in Mental Health, Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Madrid, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, Valencia, Spain.
| |
Collapse
|
21
|
Georgiou C, Kehayas V, Lee KS, Brandalise F, Sahlender DA, Blanc J, Knott G, Holtmaat A. A subpopulation of cortical VIP-expressing interneurons with highly dynamic spines. Commun Biol 2022; 5:352. [PMID: 35418660 PMCID: PMC9008030 DOI: 10.1038/s42003-022-03278-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 03/10/2022] [Indexed: 11/09/2022] Open
Abstract
Structural synaptic plasticity may underlie experience and learning-dependent changes in cortical circuits. In contrast to excitatory pyramidal neurons, insight into the structural plasticity of inhibitory neurons remains limited. Interneurons are divided into various subclasses, each with specialized functions in cortical circuits. Further knowledge of subclass-specific structural plasticity of interneurons is crucial to gaining a complete mechanistic understanding of their contribution to cortical plasticity overall. Here, we describe a subpopulation of superficial cortical multipolar interneurons expressing vasoactive intestinal peptide (VIP) with high spine densities on their dendrites located in layer (L) 1, and with the electrophysiological characteristics of bursting cells. Using longitudinal imaging in vivo, we found that the majority of the spines are highly dynamic, displaying lifetimes considerably shorter than that of spines on pyramidal neurons. Using correlative light and electron microscopy, we confirmed that these VIP spines are sites of excitatory synaptic contacts, and are morphologically distinct from other spines in L1.
Collapse
Affiliation(s)
- Christina Georgiou
- Department of Basic Neurosciences and the Center for Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,The Lemanic Neuroscience Graduate School, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Vassilis Kehayas
- Department of Basic Neurosciences and the Center for Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Institute of Computer Science, Foundation for Research and Technology - Hellas (FORTH), Heraklion, Crete, Greece
| | - Kok Sin Lee
- Department of Basic Neurosciences and the Center for Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,The Lemanic Neuroscience Graduate School, Universities of Geneva and Lausanne, Geneva, Switzerland
| | - Federico Brandalise
- Department of Basic Neurosciences and the Center for Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Department of Bioscience, University of Milan, Milan, Italy
| | | | - Jerome Blanc
- Ecole Polytechnique Federale Lausanne, Lausanne, Switzerland
| | - Graham Knott
- Ecole Polytechnique Federale Lausanne, Lausanne, Switzerland
| | - Anthony Holtmaat
- Department of Basic Neurosciences and the Center for Neuroscience, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
22
|
Ringwald KG, Pfarr JK, Stein F, Brosch K, Meller T, Thomas-Odenthal F, Meinert S, Waltemate L, Breuer F, Winter A, Lemke H, Grotegerd D, Thiel K, Bauer J, Hahn T, Jansen A, Dannlowski U, Krug A, Nenadić I, Kircher T. Association between stressful life events and grey matter volume in the medial prefrontal cortex: A 2-year longitudinal study. Hum Brain Mapp 2022; 43:3577-3584. [PMID: 35411559 PMCID: PMC9248310 DOI: 10.1002/hbm.25869] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/16/2022] [Accepted: 03/31/2022] [Indexed: 12/16/2022] Open
Abstract
Stressful life events (SLEs) in adulthood are a risk factor for various disorders such as depression, cancer or infections. Part of this risk is mediated through pathways altering brain physiology and structure. There is a lack of longitudinal studies examining associations between SLEs and brain structural changes. High-resolution structural magnetic resonance imaging data of 212 healthy subjects were acquired at baseline and after 2 years. Voxel-based morphometry was used to identify associations between SLEs using the Life Events Questionnaire and grey matter volume (GMV) changes during the 2-year period in an ROI approach. Furthermore, we assessed adverse childhood experiences as a possible moderator of SLEs-GMV change associations. SLEs were negatively associated with GMV changes in the left medial prefrontal cortex. This association was stronger when subjects had experienced adverse childhood experiences. The medial prefrontal cortex has previously been associated with stress-related disorders. The present findings represent a potential neural basis of the diathesis-stress model of various disorders.
Collapse
Affiliation(s)
- Kai G Ringwald
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Julia-Katharina Pfarr
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Frederike Stein
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Katharina Brosch
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Tina Meller
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | | | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany.,Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Lena Waltemate
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Fabian Breuer
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Alexandra Winter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Hannah Lemke
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Thiel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Jochen Bauer
- University Clinic for Radiology, University of Münster, Münster, Germany
| | - Tim Hahn
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Andreas Jansen
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany.,Core-Facility BrainImaging, Faculty of Medicine, Philipps-Universität Marburg, Münster, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Axel Krug
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany.,Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Igor Nenadić
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| | - Tilo Kircher
- Department of Psychiatry and Psychotherapy, University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior (CMBB), University of Marburg and Justus Liebig University Giessen, Marburg, Germany
| |
Collapse
|
23
|
Schaeuble D, Myers B. Cortical–Hypothalamic Integration of Autonomic and Endocrine Stress Responses. Front Physiol 2022; 13:820398. [PMID: 35222086 PMCID: PMC8874315 DOI: 10.3389/fphys.2022.820398] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 01/19/2022] [Indexed: 12/18/2022] Open
Abstract
The prevalence and severity of cardiovascular disease (CVD) are exacerbated by chronic stress exposure. While stress-induced sympathetic activity and elevated glucocorticoid secretion impair cardiovascular health, the mechanisms by which stress-responsive brain regions integrate autonomic and endocrine stress responses remain unclear. This review covers emerging literature on how specific cortical and hypothalamic nuclei regulate cardiovascular and neuroendocrine stress responses. We will also discuss the current understanding of the cellular and circuit mechanisms mediating physiological stress responses. Altogether, the reviewed literature highlights the current state of stress integration research, as well unanswered questions about the brain basis of CVD risk.
Collapse
|
24
|
Laine M, Shansky R. Rodent models of stress and dendritic plasticity – Implications for psychopathology. Neurobiol Stress 2022; 17:100438. [PMID: 35257016 PMCID: PMC8897597 DOI: 10.1016/j.ynstr.2022.100438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 11/28/2022] Open
Abstract
Stress, as commonplace as it is, is a major environmental risk factor for psychopathology. While this association intuitively, anecdotally, and empirically makes sense, we are still very early in the process of understanding what the neurobiological manifestations of this risk truly are. Seminal work from the past few decades has established structural plasticity in the brain as a potential key mechanism. In this review we discuss evidence linking particularly chronic stress exposure in rodent models to plasticity at the dendrites, like remodeling of dendritic branches and spines, in a range of brain regions. A number of candidate mechanisms that seek to explain how stress influences neuroanatomy at this level have been proposed, utilizing in vivo, ex vivo and in vitro methods. However, a large gap still remains in our knowledge of how such dynamic structural changes ultimately relate to downstream effects such as altered affective and cognitive states relevant for psychopathology. We propose that future work expand our understanding of plasticity of specific stress-related brain circuits and cell-types. We also note that the vast majority of the work has been conducted solely on male rodents. The next big strides in our understanding of the neurobiology of psychopathology will require the inclusion of female subjects, as several studies have suggested both sex divergent and convergent features. By understanding plasticity, we can harness it. The growth of this body of knowledge will inform our efforts to improve the therapeutic options for stress-related psychopathology.
Collapse
|
25
|
Wang Z, Cao Q, Bai W, Zheng X, Liu T. Decreased Phase-Amplitude Coupling Between the mPFC and BLA During Exploratory Behaviour in Chronic Unpredictable Mild Stress-Induced Depression Model of Rats. Front Behav Neurosci 2022; 15:799556. [PMID: 34975430 PMCID: PMC8716490 DOI: 10.3389/fnbeh.2021.799556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/16/2021] [Indexed: 11/13/2022] Open
Abstract
Depression is a common neuropsychiatric illness observed worldwide, and reduced interest in exploration is one of its symptoms. The control of dysregulated medial prefrontal cortex (mPFC) over the basolateral amygdala (BLA) is related to depression. However, the oscillation interaction in the mPFC-BLA circuit has remained elusive. Therefore, this study used phase-amplitude coupling (PAC), which provides complicated forms of information transmission by the phase of low-frequency rhythm, modulating the amplitude of high-frequency rhythm, and has a potential application for the treatment of neurological disease. The chronic unpredictable mild stress (CUMS) was used to prepare the rat models of depression. Moreover, multichannel in vivo recording was applied to obtain the local field potentials (LFPs) of the mPFC, the BLA in rats in control, and CUMS groups, while they explored the open field. The results showed prominent coupling between the phase of theta oscillation (4-12 Hz) in the mPFC and the amplitude of high-gamma oscillation (70-120 Hz) in the BLA. Compared to the control group, this theta-gamma PAC was significantly decreased in the CUMS group, which was accompanied by the diminished exploratory behaviour. The results indicate that the coupling between the phase of theta in the mPFC and the amplitude of gamma in the BLA is involved in exploratory behaviour, and this decreased coupling may inhibit exploratory behaviour of rats exposed to CUMS.
Collapse
Affiliation(s)
- Zihe Wang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Qingying Cao
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Wenwen Bai
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Xuyuan Zheng
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Tiaotiao Liu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
26
|
Serradas ML, Stein V, Gellner AK. Long-term changes of parvalbumin- and somatostatin-positive interneurons of the primary motor cortex after chronic social defeat stress depend on individual stress-vulnerability. Front Psychiatry 2022; 13:946719. [PMID: 35966477 PMCID: PMC9366473 DOI: 10.3389/fpsyt.2022.946719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic stress is a major risk factor for developing mental illnesses and cognitive deficiencies although stress-susceptibility varies individually. In a recent study, we established the connection between chronic social defeat stress (CSDS) and impaired motor learning abilities accompanied by chronically disturbed structural neuroplasticity in the primary motor cortex (M1) of mice. In this study, we further investigated the long-term effects of CSDS exposure on M1, focusing on the interneuronal cell population. We used repeated CSDS to elicit effects across behavioral, endocrinological, and metabolic parameters in mice. Susceptible and resilient phenotypes were discriminated by symptom load and motor learning abilities were assessed on the rotarod. Structural changes in interneuronal circuits of M1 were studied by immunohistochemistry using parvalbumin (PV+) and somatostatin (SST+) markers. Stress-susceptible mice had a blunted stress hormone response and impaired motor learning skills. These mice presented reduced numbers of both interneuron populations in M1 with layer-dependent distribution, while alterations in cell size and immunoreactivity were found in both susceptible and resilient individuals. These results, together with our previous data, suggest that stress-induced cell loss and degeneration of the GABAergic interneuronal network of M1 could underlay impaired motor learning, due to their role in controlling the excitatory output and spine dynamics of principal neurons required for this task. Our study further highlights the importance of long-term outcomes of chronically stressed individuals which are translationally important due to the long timecourses of stress-induced neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maria L Serradas
- Institute of Physiology II, Medical Faculty, University of Bonn, Bonn, Germany
| | - Valentin Stein
- Institute of Physiology II, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anne-Kathrin Gellner
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
27
|
Zhu Y, Qu Y, Zhang J, Hou J, Fang J, Shen J, Xu C, Huang M, Qiao H, An S. Phencynonate hydrochloride exerts antidepressant effects by regulating the dendritic spine density and altering glutamate receptor expression. Behav Pharmacol 2021; 32:660-672. [PMID: 34751176 DOI: 10.1097/fbp.0000000000000660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Phencynonate hydrochloride (PCH) is a drug that crosses the blood-brain barrier. Cellular experiments confirmed that PCH protects against glutamate toxicity and causes only weak central inhibition and limited side effects. As shown in our previous studies, PCH alleviates depression-like behaviours induced by chronic unpredictable mild stress (CUMS). Here we administered PCH at three different doses (4, 8 and 16 mg/kg) to male rats for two continuous days after CUMS and conducted behavioural tests to assess the dose-dependent antidepressant effects of PCH and its effects on the neuroplasticity in the hippocampus and medial prefrontal cortex (mPFC). Meanwhile, we measured the spine density and expression of related proteins to illustrate the mechanism of PCH. PCH treatment (8 mg/kg) significantly alleviated depression-like behaviours induced by CUMS. All doses of PCH treatment reversed the spine loss in prelimbic and CA3 regions induced by CUMS. Kalirin-7 expression was decreased in the hippocampus and mPFC of the CUMS group. The expression of the NR1 and NR2B subunits in the hippocampus, and NR2B in mPFC are increased by CUMS. PCH treatment (8 and 16 mg/kg) reversed all of these changes of Kalirin-7 in PFC and hippocampus, as well as NR1 and NR2B expression in the hippocampus. PCH is expected to be developed as a new type of rapid antidepressant. Its antidepressant effect may be closely related to the modulation of dendritic spine density in the prelimbic and CA3 regions and the regulation of Kalilin-7 and N-methyl-D-aspartic acid receptor levels in the hippocampus.
Collapse
Affiliation(s)
- Yingqi Zhu
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Yishan Qu
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Jing Zhang
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Jun Hou
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Jie Fang
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Jingxuan Shen
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Chang Xu
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Minyi Huang
- College of Agriculture and Biotechnology, Hunan University of Humanities, Science and Technology, Loudi, Hunan, China
| | - Hui Qiao
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| | - Shucheng An
- Institute of Brain and Behavioural Sciences, College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi
| |
Collapse
|
28
|
Wallace T, Myers B. Effects of Biological Sex and Stress Exposure on Ventromedial Prefrontal Regulation of Mood-Related Behaviors. Front Behav Neurosci 2021; 15:737960. [PMID: 34512290 PMCID: PMC8426926 DOI: 10.3389/fnbeh.2021.737960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/06/2021] [Indexed: 11/13/2022] Open
Abstract
The ventral portion of the medial prefrontal cortex (vmPFC) regulates mood, sociability, and context-dependent behaviors. Consequently, altered vmPFC activity has been implicated in the biological basis of emotional disorders. Recent methodological advances have greatly enhanced the ability to investigate how specific prefrontal cell populations regulate mood-related behaviors, as well as the impact of long-term stress on vmPFC function. However, emerging preclinical data identify prominent sexual divergence in vmPFC behavioral regulation and stress responsivity. Notably, the rodent infralimbic cortex (IL), a vmPFC subregion critical for anti-depressant action, shows marked functional divergence between males and females. Accordingly, this review examines IL encoding and modulation of mood-related behaviors, including coping style, reward, and sociability, with a focus on sex-based outcomes. We also review how these processes are impacted by prolonged stress exposure. Collectively, the data suggest that chronic stress has sex-specific effects on IL excitatory/inhibitory balance that may account for sex differences in the prevalence and course of mood disorders.
Collapse
Affiliation(s)
- Tyler Wallace
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Brent Myers
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
29
|
Santos-Costa N, Baptista-de-Souza D, Canto-de-Souza L, Fresca da Costa V, Nunes-de-Souza RL. Glutamatergic Neurotransmission Controls the Functional Lateralization of the mPFC in the Modulation of Anxiety Induced by Social Defeat Stress in Male Mice. Front Behav Neurosci 2021; 15:695735. [PMID: 34497496 PMCID: PMC8419264 DOI: 10.3389/fnbeh.2021.695735] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/26/2021] [Indexed: 12/01/2022] Open
Abstract
The rodent medial prefrontal cortex (mPFC) is anatomically divided into cingulate (Cg1), prelimbic (PrL), and infralimbic (IL) subareas. The left and right mPFC (L and RmPFC) process emotional responses induced by stress-related stimuli, and LmPFC and RmPFC inhibition elicit anxiogenesis and anxiolysis, respectively. Here we sought to investigate (i) the mPFC functional laterality on social avoidance/anxiogenic-like behaviors in male mice subjected to chronic social defeat stress (SDS), (ii) the effects of left prelimbic (PrL) inhibition (with local injection of CoCl2) on the RmPFC glutamatergic neuronal activation pattern (immunofluorescence assay), and (iii) the effects of the dorsal right mPFC (Cg1 + PrL) NMDA receptor blockade (with local injection of AP7) on the anxiety induced by left dorsal mPFC inhibition in mice exposed to the elevated plus maze (EPM). Results showed that chronic SDS induced anxiogenic-like behaviors followed by the rise of ΔFosB labeling and by ΔFosB + CaMKII double-labeling bilaterally in the Cg1 and IL subareas of the mPFC. Chronic SDS also increased ΔFosB and by ΔFosB + CaMKII labeling only on the right PrL. Also, the left PrL inhibition increased cFos + CaMKII labeling in the contralateral PrL and IL. Moreover, anxiogenesis induced by the left PrL inhibition was blocked by NMDA receptor antagonist AP7 injected into the right PrL. These findings suggest the lateralized control of the glutamatergic neurotransmission in the modulation of emotional-like responses in mice subjected to chronic SDS.
Collapse
Affiliation(s)
- Nathália Santos-Costa
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Universidade Estadual Paulista, Araraquara, Brazil.,Joint Graduate Program in Physiological Sciences (PIPGCF) UFSCar- Universidade Estadual Paulista, São Carlos, Brazil
| | - Daniela Baptista-de-Souza
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Universidade Estadual Paulista, Araraquara, Brazil
| | - Lucas Canto-de-Souza
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Universidade Estadual Paulista, Araraquara, Brazil
| | - Vinícius Fresca da Costa
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Universidade Estadual Paulista, Araraquara, Brazil.,Joint Graduate Program in Physiological Sciences (PIPGCF) UFSCar- Universidade Estadual Paulista, São Carlos, Brazil
| | - Ricardo Luiz Nunes-de-Souza
- Laboratory of Pharmacology, School of Pharmaceutical Sciences, Universidade Estadual Paulista, Araraquara, Brazil.,Joint Graduate Program in Physiological Sciences (PIPGCF) UFSCar- Universidade Estadual Paulista, São Carlos, Brazil
| |
Collapse
|
30
|
Chronic Stress Induces Sex-Specific Functional and Morphological Alterations in Corticoaccumbal and Corticotegmental Pathways. Biol Psychiatry 2021; 90:194-205. [PMID: 33867113 DOI: 10.1016/j.biopsych.2021.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 02/08/2021] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND The medial prefrontal cortex (mPFC) is part of a complex circuit controlling stress responses by sending projections to different limbic structures including the nucleus accumbens (NAc) and ventral tegmental area (VTA). However, the impact of chronic stress on NAc- and VTA-projecting mPFC neurons is still unknown, and the distinct contribution of these pathways to stress responses in males and females is unclear. METHODS Behavioral stress responses were induced by 21 days of chronic variable stress in male and female C57BL/6NCrl mice. An intersectional viral approach was used to label both pathways and assess the functional, morphological, and transcriptional adaptations in NAc- and VTA-projecting mPFC neurons in stressed males and females. Using chemogenetic approaches, we modified neuronal activity of NAc-projecting mPFC neurons to decipher their contribution to stress phenotypes. RESULTS Chronic variable stress induced depressive-like behaviors in males and females. NAc- and VTA-projecting mPFC neurons exhibited sex-specific functional, morphological, and transcriptional alterations. The functional changes were more severe in females in NAc-projecting mPFC neurons, while males exhibited more drastic reductions in dendritic complexity in VTA-projecting mPFC neurons after chronic variable stress. Finally, chemogenetic overactivation of the corticoaccumbal pathway triggered anxiety and behavioral despair in both sexes, while its inhibition rescued the phenotype only in females. CONCLUSIONS Our results suggest that stress responses in males and females result from pathway-specific changes in the activity of transcriptional programs controlling the morphological and synaptic properties of corticoaccumbal and corticotegmental pathways in a sex-specific fashion.
Collapse
|
31
|
Hossain R, Al-Khafaji K, Khan RA, Sarkar C, Islam MS, Dey D, Jain D, Faria F, Akbor R, Atolani O, Oliveira SMR, Siyadatpanah A, Pereira MDL, Islam MT. Quercetin and/or Ascorbic Acid Modulatory Effect on Phenobarbital-Induced Sleeping Mice Possibly through GABA A and GABA B Receptor Interaction Pathway. Pharmaceuticals (Basel) 2021; 14:ph14080721. [PMID: 34451819 PMCID: PMC8398796 DOI: 10.3390/ph14080721] [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: 06/28/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/28/2022] Open
Abstract
Depressive disorder is a recurrent illness that affects large numbers of the general population worldwide. In recent years, the goal of depression treatment has moved from symptomatic response to that of full remission. However, treatment-resistant depression is a major challenge in the treatment of depression or depression-related disorders. Consensus opinion, therefore, suggests that effective combined aggressive initial treatment is the most appropriate strategy. This study aimed to evaluate the effects of quercetin (QUR) and/or ascorbic acid (AA) on Phenobarbital-induced sleeping mice. QUR (50 mg/kg) and/or AA (25 mg/kg) with or without intraperitoneally pre-treated with GABA receptor agonist (diazepam: 2 mg/kg, i.p.) or antagonist (Flumazenil: 2.5 mg/kg, i.p.) to underscore the effects, as well as the possible involvement of the GABA receptor in the modulatory action of QUR and AA in sleeping mice. Additionally, an in silico study was undertaken to predict the involvement of GABA receptors in the sleep mechanism. Findings suggest that the pretreatment of QUR and AA modulated the onset and duration of action of the standard drugs in experimental animals. The acute administration of QUR and/or AA significantly (p < 0.05) reversed the DZP-mediated onset of action and slightly reversed the duration of sleep time in comparison to the vehicle (control) group. A further combination of QUR or AA with the FLU resulted in an enhancement of the onset of action while reducing the duration of action, suggesting a FLU-like effect on the test animals. In in silico studies, AA and QUR showed good to moderate binding affinities with GABAA and GABAB receptors. Both QUR and AA produced a stimulatory-like effect on mice, possibly through the GABAA and GABAB receptor interaction pathways. Further studies are necessary to verify this activity and clarify the exact mechanism of action(s) involved.
Collapse
Affiliation(s)
- Rajib Hossain
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (R.H.); (C.S.); (M.S.I.); (F.F.); (R.A.)
| | - Khattab Al-Khafaji
- Department of Chemistry, Faculty of Arts and Sciences, Gaziantep University, 27310 Gaziantep, Turkey;
| | - Rasel Ahmed Khan
- Pharmacy Discipline, Life Science School, Khulna University, Khulna 9280, Bangladesh;
| | - Chandan Sarkar
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (R.H.); (C.S.); (M.S.I.); (F.F.); (R.A.)
| | - Md. Shahazul Islam
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (R.H.); (C.S.); (M.S.I.); (F.F.); (R.A.)
| | - Dipta Dey
- Department of Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalgonj 8100, Bangladesh;
| | - Divya Jain
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Vanasthali 304022, Rajasthan, India;
| | - Farhana Faria
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (R.H.); (C.S.); (M.S.I.); (F.F.); (R.A.)
| | - Rukaya Akbor
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (R.H.); (C.S.); (M.S.I.); (F.F.); (R.A.)
| | - Olubunmi Atolani
- Department of Chemistry, University of Ilorin, Ilorin P.M.B. 1515, Nigeria;
| | - Sónia M. R. Oliveira
- CICECO-Aveiro Institute of Materials & Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
- Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
| | - Abolghasem Siyadatpanah
- Ferdows School of Paramedical and Health, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials & Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence: (M.d.L.P.); (M.T.I.)
| | - Muhammad Torequl Islam
- Department of Pharmacy, Life Science Faculty, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (R.H.); (C.S.); (M.S.I.); (F.F.); (R.A.)
- Correspondence: (M.d.L.P.); (M.T.I.)
| |
Collapse
|
32
|
Bittar TP, Labonté B. Functional Contribution of the Medial Prefrontal Circuitry in Major Depressive Disorder and Stress-Induced Depressive-Like Behaviors. Front Behav Neurosci 2021; 15:699592. [PMID: 34234655 PMCID: PMC8257081 DOI: 10.3389/fnbeh.2021.699592] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 05/20/2021] [Indexed: 12/13/2022] Open
Abstract
Despite decades of research on the neurobiology of major depressive disorder (MDD), the mechanisms underlying its expression remain unknown. The medial prefrontal cortex (mPFC), a hub region involved in emotional processing and stress response elaboration, is highly impacted in MDD patients and animal models of chronic stress. Recent advances showed alterations in the morphology and activity of mPFC neurons along with profound changes in their transcriptional programs. Studies at the circuitry level highlighted the relevance of deciphering the contributions of the distinct prefrontal circuits in the elaboration of adapted and maladapted behavioral responses in the context of chronic stress. Interestingly, MDD presents a sexual dimorphism, a feature recognized in the molecular field but understudied on the circuit level. This review examines the recent literature and summarizes the contribution of the mPFC circuitry in the expression of MDD in males and females along with the morphological and functional alterations that change the activity of these neuronal circuits in human MDD and animal models of depressive-like behaviors.
Collapse
Affiliation(s)
- Thibault P. Bittar
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Benoit Labonté
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
33
|
Chen S, Gao L, Li X, Ye Y. Allopregnanolone in mood disorders: Mechanism and therapeutic development. Pharmacol Res 2021; 169:105682. [PMID: 34019980 DOI: 10.1016/j.phrs.2021.105682] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 01/23/2023]
Abstract
The neuroactive steroid allopregnanolone (ALLO) is an endogenous positive allosteric modulator of GABA type A receptor (GABAAR), and the down-regulation of its biosynthesis have been attributed to the development of mood disorders, such as depression, anxiety and post-traumatic stress disorder (PTSD). ALLO mediated depression/anxiety involves GABAergic mechanisms and appears to be related to brain-derived neurotrophic factor (BDNF), dopamine receptor, glutamate neurotransmission, and Ca2+ channel. In the clinical, brexanolone, as a newly developed intravenous ALLO preparation, has been approved for the treatment of postpartum depression (PPD). In addition, traditional antidepressants such as selective serotonin reuptake inhibitor (SSRI) could reverse ALLO decline. Recently, the translocation protein (TSPO, 18 kDa), which involves in the speed-limiting step of ALLO synthesis, and ALLO derivatization have been identified as new directions for antidepressant therapy. This review provides an overview of ALLO researches in animal model and patients, discusses its role in the development and treatment of depression/anxiety, and directs its therapeutic potential in future.
Collapse
Affiliation(s)
- Shiyi Chen
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Lijuan Gao
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Xiaoyu Li
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| | - Yiping Ye
- School of Pharmacy, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, Zhejiang, People's Republic of China; Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, Zhejiang, People's Republic of China.
| |
Collapse
|
34
|
Bueno-Fernandez C, Perez-Rando M, Alcaide J, Coviello S, Sandi C, Castillo-Gómez E, Nacher J. Long term effects of peripubertal stress on excitatory and inhibitory circuits in the prefrontal cortex of male and female mice. Neurobiol Stress 2021; 14:100322. [PMID: 33869684 PMCID: PMC8045050 DOI: 10.1016/j.ynstr.2021.100322] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/22/2021] [Accepted: 03/24/2021] [Indexed: 12/01/2022] Open
Abstract
The impact of stressful events is especially important during early life, because certain cortical regions, especially the prefrontal cortex (PFC), are still developing. Consequently, aversive experiences that occur during the peripubertal period can cause long-term alterations in neural connectivity, physiology and related behaviors. Although sex influences the stress response and women are more likely to develop stress-related psychiatric disorders, knowledge about the effects of stress on females is still limited. In order to analyze the long-term effects of peripubertal stress (PPS) on the excitatory and inhibitory circuitry of the adult PFC, and whether these effects are sex-dependent, we applied an unpredictable chronic PPS protocol based on psychogenic stressors. Using two strains of transgenic mice with specific fluorescent cell reporters, we studied male and diestrus females to know how PPS affects the structure and connectivity of parvalbumin expressing (PV+) interneurons and pyramidal neurons. We also studied the expression of molecules related to excitatory and inhibitory neurotransmission, as well as alterations in the expression of plasticity-related molecules. The structure of pyramidal neurons was differentially affected by PPS in male and female mice: while the former had a decreased dendritic spine density, the latter displayed an increase in this parameter. PPS affected the density of puncta expressing excitatory and inhibitory synaptic markers exclusively in the female mPFC. Similarly, only in female mice we observed an increased complexity of the dendritic tree of PV+ neurons. Regarding the perisomatic innervation on pyramidal and PV + neurons by basket cells, we found a significant increase in the density of puncta in stressed animals, with interesting differences between the sexes and the type of basket cell analyzed. Finally, the PPS protocol also altered the total number of somata expressing the polysialylated form of the neural cell adhesion molecule (PSA-NCAM) when we analyzed both sexes together. These results highlight the strong programming effects of aversive experiences during early life for the establishment of cortical circuitry and the special impact of these stressful events on females.
Collapse
Affiliation(s)
- Clara Bueno-Fernandez
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Simona Coviello
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Carmen Sandi
- Department of Life Sciences, Laboratory of Behavioral Genetics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Esther Castillo-Gómez
- Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain.,Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain.,Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain.,Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010, Valencia, Spain
| |
Collapse
|
35
|
Czéh B, Simon M. Benefits of animal models to understand the pathophysiology of depressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110049. [PMID: 32735913 DOI: 10.1016/j.pnpbp.2020.110049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/13/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022]
Abstract
Major depressive disorder (MDD) is a potentially life-threatening mental disorder imposing severe social and economic burden worldwide. Despite the existence of effective antidepressant treatment strategies the exact pathophysiology of the disease is still unknown. Large number of animal models of MDD have been developed over the years, but all of them suffer from significant shortcomings. Despite their limitations these models have been extensively used in academic research and drug development. The aim of this review is to highlight the benefits of animal models of MDD. We focus here on recent experimental data where animal models were used to examine current theories of this complex disease. We argue, that despite their evident imperfections, these models provide invaluable help to understand cellular and molecular mechanisms contributing to the development of MDD. Furthermore, animal models are utilized in research to find clinically useful biomarkers. We discuss recent neuroimaging and microRNA studies since these investigations yielded promising candidates for biomarkers. Finally, we briefly summarize recent progresses in drug development, i.e. the FDA approval of two novel antidepressant drugs: S-ketamine and brexanolone (allopregnanolone). Deeper understanding of the exact molecular and cellular mechanisms of action responsible for the antidepressant efficacy of these rapid acting drugs could aid us to design further compounds with similar effectiveness, but less side effects. Animal studies are likely to provide valuable help in this endeavor.
Collapse
Affiliation(s)
- Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary; Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary.
| | - Maria Simon
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary; Department of Psychiatry and Psychotherapy, Medical School, University of Pécs, Hungary
| |
Collapse
|
36
|
Philip V, Newton DF, Oh H, Collins SM, Bercik P, Sibille E. Transcriptional markers of excitation-inhibition balance in germ-free mice show region-specific dysregulation and rescue after bacterial colonization. J Psychiatr Res 2021; 135:248-255. [PMID: 33508544 PMCID: PMC7914199 DOI: 10.1016/j.jpsychires.2021.01.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/17/2020] [Accepted: 01/15/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Studies of germ-free (GF) mice demonstrate that gut microbiota can influence behaviour by modulating neurochemical pathways in the brain, and that bacterial colonization normalizes behavioural deficits in GF-mice. Since disrupted GABAergic and glutamatergic signaling are reported in mood disorders, this study investigated the effect of gut microbiota manipulations on EIB-relevant gene expression in the brain. METHODS GF Swiss-Webster mice were colonized with E. coli JM83, complex microbiota (specific-pathogen-free; SPF), or no microbiota, and compared with controls (n = 6/group). 21 synaptic genes representing GABAergic, glutamatergic, BDNF, and astrocytic functions were measured in the hippocampus, amygdala, and prefrontal cortex using quantitative PCR. Gene co-expression analysis was used to identify gene modules related to colonization status, and compared by permutation analysis. Gene expression profiles were compared to existing post-mortem cohorts of depressed subjects (n = 28 cases vs 28 controls). RESULTS Region-specific alterations in gene expression were observed in GF-mice compared to controls. 58% of all genes (14/24) altered in GF-mice were normalized following SPF-colonization. GF-mice displayed disorganization of gene co-expression networks in all three brain regions (hippocampus, p = 0.0003; amygdala, p = 0.0012; mPFC, p = 0.0069), which was restored by SPF colonization in hippocampus (p v.s. GF = 0.0003, p v.s. control = 0.60). The hippocampal gene expression profile in GF-mice was significantly correlated with that in human depression (ρ = 0.51, p = 0.027), and this correlation was not observed after colonization. CONCLUSION Together, we show that the absence of gut microbiota disrupts the expression of EIB-relevant genes in mice, and colonization restores EIB-relevant expression, in ways that are relevant to human depression.
Collapse
Affiliation(s)
- Vivek Philip
- Campbell Family Mental Health Research Institute, the Centre for Addiction and Mental Health, Toronto, Canada,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Dwight F. Newton
- Campbell Family Mental Health Research Institute, the Centre for Addiction and Mental Health, Toronto, Canada,Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Hyunjung Oh
- Campbell Family Mental Health Research Institute, the Centre for Addiction and Mental Health, Toronto, Canada,Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Stephen M Collins
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Premysl Bercik
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Etienne Sibille
- Campbell Family Mental Health Research Institute, The Centre for Addiction and Mental Health, Toronto, Canada; Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada; Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
37
|
Era V, Carnevali L, Thayer JF, Candidi M, Ottaviani C. Dissociating cognitive, behavioral and physiological stress-related responses through dorsolateral prefrontal cortex inhibition. Psychoneuroendocrinology 2021; 124:105070. [PMID: 33310375 DOI: 10.1016/j.psyneuen.2020.105070] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/29/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022]
Abstract
The left dorsolateral prefrontal cortex (dlPFC) has been implicated in the regulation of stress-related cognitive processes and physiological responses and is the principal target of noninvasive brain stimulation techniques applied to psychiatric conditions. However, existing studies are mostly correlational and causal evidence on the role of this region in mediating specific psychophysiological mechanisms underpinning stress-related responses are needed to make the application of such techniques more efficient. To fill this gap, this study used inhibitory continuous theta burst stimulation (cTBS) in healthy individuals to examine the extent to which activity of the left dlPFC is associated with cognitive (subjective focus on a tracking task), behavioral (reaction times and variability), and physiological responses (heart rate and its variability and cortisol level) following induction of perseverative cognition. Compared to sham and left ventral PreMotor area stimulation (as active control area), inhibition of left dlPFC determined sustained autonomic and neuroendocrine activation and increased the subjective perception of being task-focused, while not changing the behavioral and self-reported stress-related responses. Adopting a causative approach, we describe a role of left dlPFC in inhibitory control of the physiological stress-response associated to perseverative thinking.
Collapse
Affiliation(s)
- Vanessa Era
- Department of Psychology, Sapienza University of Rome, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Luca Carnevali
- Stress Physiology Lab, Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Italy
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, CA, USA
| | - Matteo Candidi
- Department of Psychology, Sapienza University of Rome, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy
| | - Cristina Ottaviani
- Department of Psychology, Sapienza University of Rome, Rome, Italy; IRCCS Santa Lucia Foundation, Rome, Italy.
| |
Collapse
|
38
|
Pace SA, Christensen C, Schackmuth MK, Wallace T, McKlveen JM, Beischel W, Morano R, Scheimann JR, Wilson SP, Herman JP, Myers B. Infralimbic cortical glutamate output is necessary for the neural and behavioral consequences of chronic stress. Neurobiol Stress 2020; 13:100274. [PMID: 33344727 PMCID: PMC7739189 DOI: 10.1016/j.ynstr.2020.100274] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/08/2020] [Accepted: 11/17/2020] [Indexed: 01/03/2023] Open
Abstract
Exposure to prolonged stress is a major risk-factor for psychiatric disorders such as generalized anxiety and major depressive disorder. Human imaging studies have identified structural and functional abnormalities in the prefrontal cortex of subjects with depression and anxiety disorders, particularly Brodmann's area 25 (BA25). Further, deep brain stimulation of BA25 reduces symptoms of treatment-resistant depression. The rat homolog of BA25 is the infralimbic cortex (IL), which is critical for cognitive appraisal, executive function, and physiological stress reactivity. Previous studies indicate that the IL undergoes stress-induced changes in excitatory/inhibitory balance culminating in reduced activity of glutamate output neurons. However, the regulatory role of IL glutamate output in mood-related behaviors after chronic variable stress (CVS) is unknown. Here, we utilized a lentiviral-packaged small-interfering RNA to reduce translation of vesicular glutamate transporter 1 (vGluT1 siRNA), thereby constraining IL glutamate output. This viral-mediated gene transfer was used in conjunction with a quantitative anatomical analysis of cells expressing the stable immediate-early gene product FosB/ΔFosB, which accumulates in response to repeated neural activation. Through assessment of FosB/ΔFosB-expressing neurons across the frontal lobe in adult male rats, we mapped regions altered by chronic stress and determined the coordinating role of the IL in frontal cortical plasticity. Specifically, CVS-exposed rats had increased density of FosB/ΔFosB-expressing cells in the IL and decreased density in the insula. The latter effect was dependent on IL glutamate output. Next, we examined the interaction of CVS and reduced IL glutamate output in behavioral assays examining coping, anxiety-like behavior, associative learning, and nociception. IL glutamate knockdown decreased immobility during the forced swim test compared to GFP controls, both in rats exposed to CVS as well as rats without previous stress exposure. Further, vGluT1 siRNA prevented CVS-induced avoidance behaviors, while also reducing risk aversion and passive coping. Ultimately, this study identifies the necessity of IL glutamatergic output for regulating frontal cortical neural activity and behavior following chronic stress. These findings also highlight how disruption of excitatory/inhibitory balance within specific frontal cortical cell populations may impact neurobehavioral adaptation and lead to stress-related disorders.
Collapse
Affiliation(s)
- Sebastian A. Pace
- Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | | | | | - Tyler Wallace
- Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Jessica M. McKlveen
- National Institutes of Health, National Center for Complementary and Integrative Health, Bethesda, MD, USA
| | - Will Beischel
- Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Rachel Morano
- Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Jessie R. Scheimann
- Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Steven P. Wilson
- Pharmacology, Physiology, and Neuroscience, University of South Carolina, Columbia, SC, USA
| | - James P. Herman
- Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, USA
| | - Brent Myers
- Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
39
|
Weger M, Alpern D, Cherix A, Ghosal S, Grosse J, Russeil J, Gruetter R, de Kloet ER, Deplancke B, Sandi C. Mitochondrial gene signature in the prefrontal cortex for differential susceptibility to chronic stress. Sci Rep 2020; 10:18308. [PMID: 33110158 PMCID: PMC7591539 DOI: 10.1038/s41598-020-75326-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction was highlighted as a crucial vulnerability factor for the development of depression. However, systemic studies assessing stress-induced changes in mitochondria-associated genes in brain regions relevant to depression symptomatology remain scarce. Here, we performed a genome-wide transcriptomic study to examine mitochondrial gene expression in the prefrontal cortex (PFC) and nucleus accumbens (NAc) of mice exposed to multimodal chronic restraint stress. We identified mitochondria-associated gene pathways as most prominently affected in the PFC and with lesser significance in the NAc. A more detailed mitochondrial gene expression analysis revealed that in particular mitochondrial DNA-encoded subunits of the oxidative phosphorylation complexes were altered in the PFC. The comparison of our data with a reanalyzed transcriptome data set of chronic variable stress mice and major depression disorder subjects showed that the changes in mitochondrial DNA-encoded genes are a feature generalizing to other chronic stress-protocols as well and might have translational relevance. Finally, we provide evidence for changes in mitochondrial outputs in the PFC following chronic stress that are indicative of mitochondrial dysfunction. Collectively, our work reinforces the idea that changes in mitochondrial gene expression are key players in the prefrontal adaptations observed in individuals with high behavioral susceptibility and resilience to chronic stress.
Collapse
Affiliation(s)
- Meltem Weger
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.,Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD, 4072, Australia
| | - Daniel Alpern
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Antoine Cherix
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.,Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, England, UK
| | - Sriparna Ghosal
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Julie Russeil
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - Rolf Gruetter
- Laboratory for Functional and Metabolic Imaging, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland
| | - E Ronald de Kloet
- Departement of Endocrinology and Metabolic Disease, Leiden University Medical Center, Leiden, The Netherlands
| | - Bart Deplancke
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.,Swiss Institute of Bioinformatics, 1015, Lausanne, Switzerland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne, 1015, Lausanne, Switzerland.
| |
Collapse
|
40
|
Sanson A, Riva MA. Anti-Stress Properties of Atypical Antipsychotics. Pharmaceuticals (Basel) 2020; 13:E322. [PMID: 33092112 PMCID: PMC7589119 DOI: 10.3390/ph13100322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022] Open
Abstract
Stress exposure represents a major environmental risk factor for schizophrenia and other psychiatric disorders, as it plays a pivotal role in the etiology as well as in the manifestation of disease symptomatology. It may be inferred that pharmacological treatments must be able to modulate the behavioral, functional, and molecular alterations produced by stress exposure to achieve significant clinical outcomes. This review aims at examining existing clinical and preclinical evidence that supports the ability of atypical antipsychotic drugs (AAPDs) to modulate stress-related alterations. Indeed, while the pharmacodynamic differences between AAPDs have been extensively characterized, less is known on their ability to regulate downstream mechanisms that are critical for functional recovery and patient stabilization. We will discuss stress-related mechanisms, spanning from neuroendocrine function to inflammation and neuronal plasticity, which are relevant for the manifestation of schizophrenic symptomatology, and we will discuss if and how AAPDs may interfere with such mechanisms. Considering the impact of stress in everyday life, we believe that a better understanding of the potential effects of AAPDs on stress-related mechanisms may provide novel and important insights for improving therapeutic strategies aimed at promoting coping mechanisms and enhancing the quality of life of patients affected by psychiatric disorders.
Collapse
Affiliation(s)
| | - Marco A. Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Via Giuseppe Balzaretti 9, 20133 Milan, Italy;
| |
Collapse
|
41
|
Pulopulos MM, Schmausser M, De Smet S, Vanderhasselt MA, Baliyan S, Venero C, Baeken C, De Raedt R. The effect of HF-rTMS over the left DLPFC on stress regulation as measured by cortisol and heart rate variability. Horm Behav 2020; 124:104803. [PMID: 32526225 DOI: 10.1016/j.yhbeh.2020.104803] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022]
Abstract
The prefrontal cortex, and especially the Dorsolateral Prefrontal Cortex (DLPFC), plays an inhibitory role in the regulation of the Hypothalamic-Pituitary-Adrenal (HPA) axis under stressful situations. Moreover, recent evidence suggests that a sustained DLPFC activation is associated with adaptive stress regulation in anticipation of a stressful event, leading to a reduced stress-induced amygdala response, and facilitating the confrontation with the stressor. However, studies using experimental manipulation of the activity of the DLPFC before a stressor are scarce, and more research is needed to understand the specific role of this brain area in the stress-induced physiological response. This pre-registered study investigated the effect on stress regulation of a single excitatory high frequency (versus sham) repetitive transcranial magnetic stimulation (HF-rTMS) session over the left DLPFC applied before the Trier Social Stress Test in 75 healthy young women (M = 21.05, SD = 2.60). Heart rate variability (HRV) and salivary cortisol were assessed throughout the experimental protocol. The active HF-rTMS and the sham group showed a similar cognitive appraisal of the stress task. No differences in HRV were observed during both the anticipation and the actual confrontation with the stress task and therefore, our results did not reflect DLPFC-related adaptive anticipatory adjustments. Importantly, participants in the active HF-rTMS group showed a lower cortisol response to stress. The effect of left prefrontal HF-rTMS on the stress system provides further critical experimental evidence for the inhibitory role played by the DLPFC in the regulation of the HPA axis.
Collapse
Affiliation(s)
- Matias M Pulopulos
- Department of Experimental Clinical and Health Psychology, Ghent University, Belgium.
| | - Maximilian Schmausser
- Department of Experimental Clinical and Health Psychology, Ghent University, Belgium
| | - Stefanie De Smet
- Department of Head and Skin, Ghent University, Belgium; Ghent Experimental Psychiatry (GHEP) Lab, Belgium
| | - Marie-Anne Vanderhasselt
- Department of Experimental Clinical and Health Psychology, Ghent University, Belgium; Department of Head and Skin, Ghent University, Belgium; Ghent Experimental Psychiatry (GHEP) Lab, Belgium
| | - Shishir Baliyan
- Department of Psychobiology, Universidad Nacional de Educación a Distancia (UNED), Spain
| | - César Venero
- Department of Psychobiology, Universidad Nacional de Educación a Distancia (UNED), Spain
| | - Chris Baeken
- Department of Head and Skin, Ghent University, Belgium; Ghent Experimental Psychiatry (GHEP) Lab, Belgium; Department of Psychiatry, University Hospital Brussels (UZBrussel), Belgium; Department of Electrical Engineering, Eindhoven University of Technology, the Netherlands
| | - Rudi De Raedt
- Department of Experimental Clinical and Health Psychology, Ghent University, Belgium
| |
Collapse
|
42
|
Bruno A, Dolcetti E, Rizzo FR, Fresegna D, Musella A, Gentile A, De Vito F, Caioli S, Guadalupi L, Bullitta S, Vanni V, Balletta S, Sanna K, Buttari F, Stampanoni Bassi M, Centonze D, Mandolesi G. Inflammation-Associated Synaptic Alterations as Shared Threads in Depression and Multiple Sclerosis. Front Cell Neurosci 2020; 14:169. [PMID: 32655374 PMCID: PMC7324636 DOI: 10.3389/fncel.2020.00169] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
In the past years, several theories have been advanced to explain the pathogenesis of Major Depressive Disorder (MDD), a neuropsychiatric disease that causes disability in general population. Several theories have been proposed to define the MDD pathophysiology such as the classic "monoamine-theory" or the "glutamate hypothesis." All these theories have been recently integrated by evidence highlighting inflammation as a pivotal player in developing depressive symptoms. Proinflammatory cytokines have been indeed claimed to contribute to stress-induced mood disturbances and to major depression, indicating a widespread role of classical mediators of inflammation in emotional control. Moreover, during systemic inflammatory diseases, peripherally released cytokines circulate in the blood, reach the brain and cause anxiety, anhedonia, social withdrawal, fatigue, and sleep disturbances. Accordingly, chronic inflammatory disorders, such as the inflammatory autoimmune disease multiple sclerosis (MS), have been associated to higher risk of MDD, in comparison with overall population. Importantly, in both MS patients and in its experimental mouse model, Experimental Autoimmune Encephalomyelitis (EAE), the notion that depressive symptoms are reactive epiphenomenon to the MS pathology has been recently challenged by the evidence of their early manifestation, even before the onset of the disease. Furthermore, in association to such mood disturbance, inflammatory-dependent synaptic dysfunctions in several areas of MS/EAE brain have been observed independently of brain lesions and demyelination. This evidence suggests that a fine interplay between the immune and nervous systems can have a huge impact on several neurological functions, including depressive symptoms, in different pathological conditions. The aim of the present review is to shed light on common traits between MDD and MS, by looking at inflammatory-dependent synaptic alterations associated with depression in both diseases.
Collapse
Affiliation(s)
- Antonio Bruno
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Ettore Dolcetti
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Francesca Romana Rizzo
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | | | - Francesca De Vito
- Unit of Neurology, Mediterranean Neurological Institute IRCCS Neuromed, Pozzilli, Italy
| | - Silvia Caioli
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Silvia Bullitta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
| | - Sara Balletta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Krizia Sanna
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
| | - Fabio Buttari
- Unit of Neurology, Mediterranean Neurological Institute IRCCS Neuromed, Pozzilli, Italy
| | | | - Diego Centonze
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University of Rome, Rome, Italy
- Unit of Neurology, Mediterranean Neurological Institute IRCCS Neuromed, Pozzilli, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| |
Collapse
|
43
|
Chronic Restraint Stress Affects Network Oscillations in the Anterior Cingulate Cortex in Mice. Neuroscience 2020; 437:172-183. [PMID: 32335214 DOI: 10.1016/j.neuroscience.2020.04.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/21/2022]
Abstract
The anterior cingulate cortex (ACC) is vulnerable to stress. Its dysfunction is observed in psychiatric disorders manifested as alterations in network oscillations. Mechanisms linking stress load to disturbed emotional-cognitive behaviors are of essential importance to further elucidate therapeutic strategies for psychiatric diseases. Here, we analyzed the effects of chronic restraint stress (CRS) load in juvenile mice on kainic acid (KA)-induced network oscillations in ACC slice preparations and on the forced swim test (FST). The immobility time (IT) was shortened at the beginning of the FST in CRS mice. Power spectral density (PSD) obtained from KA-induced oscillations in field potentials in the superficial layers of the ACC were altered in slices from the CRS mice. The PSD was decreased in CRS mice at the alpha (8-12 Hz), beta (13-30 Hz), low gamma (30-50 Hz), and high gamma (50-80 Hz) components. Noradrenaline increased the PSD of the theta (3-8 Hz) components in both the control and CRS groups, and also in alpha components only in the CRS group. Dopamine did not modulate the PSD of any frequency components in the control mice, whereas it enhanced the PSD of theta and alpha components in CRS mice. It was suggested that chronic stress load affects the dynamics of the network oscillations in the ACC with enhanced cathecolaminergic modulation.
Collapse
|
44
|
Carceller H, Guirado R, Nacher J. Dark exposure affects plasticity‐related molecules and interneurons throughout the visual system during adulthood. J Comp Neurol 2020; 528:1349-1366. [DOI: 10.1002/cne.24832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 11/16/2019] [Accepted: 11/22/2019] [Indexed: 11/08/2022]
Affiliation(s)
- Hector Carceller
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED)Universitat de Valencia Valencia Spain
| | - Ramon Guirado
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED)Universitat de Valencia Valencia Spain
| | - Juan Nacher
- Neurobiology Unit, Department of Cell Biology, Interdisciplinary Research Structure for Biotechnology and Biomedicine (BIOTECMED)Universitat de Valencia Valencia Spain
- CIBERSAM: Spanish National Network for Research in Mental Health Madrid Spain
- Fundación Investigación Hospital Clínico de Valencia, INCLIVA Valencia Spain
| |
Collapse
|
45
|
Δ-9-Tetrahydrocannabinol treatment during adolescence and alterations in the inhibitory networks of the adult prefrontal cortex in mice subjected to perinatal NMDA receptor antagonist injection and to postweaning social isolation. Transl Psychiatry 2020; 10:177. [PMID: 32488050 PMCID: PMC7266818 DOI: 10.1038/s41398-020-0853-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 05/11/2020] [Accepted: 05/19/2020] [Indexed: 12/18/2022] Open
Abstract
The prefrontal cortex (PFC) continues its development during adolescence and alterations in its structure and function, particularly of inhibitory networks, have been detected in schizophrenic patients. Since cannabis use during adolescence is a risk factor for this disease, our main objective was to investigate whether THC administration during this period might exacerbate alterations in prefrontocortical inhibitory networks in mice subjected to a perinatal injection of MK801 and postweaning social isolation. This double-hit model (DHM) combines a neurodevelopmental manipulation and the exposure to an aversive experience during early life; previous work has shown that DHM mice have important alterations in the structure and connectivity of PFC interneurons. In the present study we found that DHM had reductions in prepulse inhibition of the startle reflex (PPI), GAD67 expression and cingulate 1 cortex volume. Interestingly, THC by itself induced increases in PPI and decreases in the dendritic complexity of somatostatin expressing interneurons. Both THC and DHM reduced the density of parvalbumin expressing cells surrounded by perineuronal nets and, when combined, they disrupted the ratio between the density of puncta expressing excitatory and inhibitory markers. Our results support previous work showing alterations in parameters involving interneurons in similar animal models and schizophrenic patients. THC treatment does not modify further these parameters, but changes some others related also to interneurons and their plasticity, in some cases in the opposite direction to those induced by the DHM, suggesting a protective effect.
Collapse
|
46
|
Khan AR, Geiger L, Wiborg O, Czéh B. Stress-Induced Morphological, Cellular and Molecular Changes in the Brain-Lessons Learned from the Chronic Mild Stress Model of Depression. Cells 2020; 9:cells9041026. [PMID: 32326205 PMCID: PMC7226496 DOI: 10.3390/cells9041026] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 04/14/2020] [Accepted: 04/19/2020] [Indexed: 02/07/2023] Open
Abstract
Major depressive disorder (MDD) is a severe illness imposing an increasing social and economic burden worldwide. Numerous rodent models have been developed to investigate the pathophysiology of MDD. One of the best characterized and most widely used models is the chronic mild stress (CMS) model which was developed more than 30 years ago by Paul Willner. More than 2000 published studies used this model, mainly to assess novel compounds with potential antidepressant efficacy. Most of these studies examined the behavioral consequences of stress and concomitant drug intervention. Much fewer studies focused on the CMS-induced neurobiological changes. However, the stress-induced cellular and molecular changes are important as they may serve as potential translational biomarkers and increase our understanding of the pathophysiology of MDD. Here, we summarize current knowledge on the structural and molecular alterations in the brain that have been described using the CMS model. We discuss the latest neuroimaging and postmortem histopathological data as well as molecular changes including recent findings on microRNA levels. Different chronic stress paradigms occasionally deliver dissimilar findings, but the available experimental data provide convincing evidence that the CMS model has a high translational value. Future studies examining the neurobiological changes in the CMS model in combination with clinically effective antidepressant drug intervention will likely deliver further valuable information on the pathophysiology of MDD.
Collapse
Affiliation(s)
- Ahmad Raza Khan
- Centre of Biomedical Research, Sanjay Gandhi Post Graduate Institute (SGPGI) Campus, Lucknow-226017, U.P, India;
| | - Lili Geiger
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary;
- Department of Laboratory Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Ove Wiborg
- Department of Health Science and Technology, Aalborg University, 9220 Aalborg, Denmark;
| | - Boldizsár Czéh
- Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, 7624 Pécs, Hungary;
- Department of Laboratory Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
- Correspondence:
| |
Collapse
|
47
|
Bidirectional Optogenetically-Induced Plasticity of Evoked Responses in the Rat Medial Prefrontal Cortex Can Impair or Enhance Cognitive Set-Shifting. eNeuro 2020; 7:ENEURO.0363-19.2019. [PMID: 31852759 PMCID: PMC6946542 DOI: 10.1523/eneuro.0363-19.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 11/21/2019] [Accepted: 12/10/2019] [Indexed: 12/20/2022] Open
Abstract
Chronic stress compromises cognition, including executive function mediated in the medial prefrontal cortex (mPFC). To investigate mechanisms underlying these processes, we use chronic unpredictable stress (CUS), which reduces activity in the mPFC and impairs cognitive set-shifting, a measure of cognitive flexibility in laboratory rats. It has been shown that CUS attenuates the local electrical field potential response evoked in the mPFC by stimulation of the ascending excitatory afferent from the mediodorsal thalamus (MDT). Thus, in this study, to investigate the role that such changes in afferent-evoked responsivity of the mPFC might play in the cognitive deficits induced by CUS, we used optogenetics to directly induce plastic changes in the thalamic-mPFC afferent pathway. Glutamatergic neurons in the MDT were virally-induced to express the ChETA variant of channelrhodopsin. Then, to first validate the optogenetic induction of plasticity, long-term depression (LTD) or long-term potentiation (LTP) were induced by laser stimulation of ChETA-expressing terminals in the mPFC of anesthetized rats. In subsequent experiments, induction of opto-LTD in awake animals produced set-shifting deficits similar to those induced by CUS. By contrast, inducing opto-LTP in rats that had received prior CUS treatment corrected the stress-induced deficit in set-shifting. These results suggest that stress-induced plasticity in the thalamic-mPFC pathway is sufficient to produce stress-induced cognitive deficits, and may represent a novel target for effective therapeutic intervention to correct cognitive impairment in stress-related psychiatric disorders.
Collapse
|
48
|
Xu S, Liu Y, Pu J, Gui S, Zhong X, Tian L, Song X, Qi X, Wang H, Xie P. Chronic Stress in a Rat Model of Depression Disturbs the Glutamine-Glutamate-GABA Cycle in the Striatum, Hippocampus, and Cerebellum. Neuropsychiatr Dis Treat 2020; 16:557-570. [PMID: 32158215 PMCID: PMC7047974 DOI: 10.2147/ndt.s245282] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/17/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a complex psychiatric illness involving multiple brain regions. Increasing evidence indicates that the striatum is involved in depression, but the molecular mechanisms remain unclear. METHODS In this study, we performed a gas chromatography-mass spectrometer (GC/MS)-based metabolomic analysis in the striatum of depressed rats induced by chronic unpredictable mild stress (CUMS). We then compared striatal data with our previous data from the hippocampus and cerebellum to systematically investigate the potential pathogenesis of depression. RESULTS We identified 22 differential metabolites in the striatum between the CUMS and control groups; these altered metabolites were mainly involved in amino acid, carbohydrate, and nucleotide metabolism. Pathway analysis revealed that the shared metabolic pathways of the striatum, hippocampus, and cerebellum were mainly involved in the glutamine-glutamate metabolic system. Four genes in the striatum (GS, GLS2, GLT1, and SSADH), six genes in the hippocampus (GS, SNAT1, GAD1, SSADH, VGAT, and ABAT), and five genes in the cerebellum (GS, ABAT, SNAT1, VGAT, and GDH) were found to be significantly altered using RT-qPCR. Correlation analysis indicated that these differential genes were strongly correlated. CONCLUSION These results suggest that chronic stress might induce depressive behaviors by disturbing the glutamine-glutamate-GABA cycle in the striatum, hippocampus, and cerebellum, and that the glutamine-glutamate-GABA cycle among these three brain regions might generate cooperative action in response to chronic stress.
Collapse
Affiliation(s)
- Shaohua Xu
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, People's Republic of China.,NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China
| | - Yiyun Liu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China
| | - Juncai Pu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China
| | - Siwen Gui
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China.,College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xiaogang Zhong
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China
| | - Lu Tian
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China
| | - Xuemian Song
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China.,College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xunzhong Qi
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China
| | - Haiyang Wang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, People's Republic of China.,NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, People's Republic of China.,Chongqing Key Laboratory of Neurobiology, Chongqing 400016, People's Republic of China.,Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
49
|
Carnevali L, Pattini E, Sgoifo A, Ottaviani C. Effects of prefrontal transcranial direct current stimulation on autonomic and neuroendocrine responses to psychosocial stress in healthy humans. Stress 2020; 23:26-36. [PMID: 31177885 DOI: 10.1080/10253890.2019.1625884] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Prolonged or repeated activation of the stress response can have negative psychological and physical consequences. The prefrontal cortex (PFC) is thought to exert an inhibitory influence on the activity of autonomic and neuroendocrine stress response systems. In this study, we further investigated this hypothesis by increasing PFC excitability using transcranial direct current stimulation (tDCS). Healthy male participants were randomized to receive either anodal (excitatory) tDCS (n = 15) or sham stimulation (n = 15) over the left dorsolateral prefrontal cortex (DLPFC) immediately before and during the exposure to a psychosocial stress test. Autonomic (heart rate (HR) and its variability) and neuroendocrine (salivary cortisol) parameters were assessed. One single session of excitatory tDCS over the left DLPFC (i) reduced HR and favored a larger vagal prevalence prior to stress exposure, (ii) moderated stress-induced HR acceleration and sympathetic activation/vagal withdrawal, but (iii) had no effect on stress-induced cortisol release. However, anodal tDCS over the left DLPFC prevented stress-induced changes in the cortisol awakening response. Finally, participants receiving excitatory tDCS reported a reduction in their levels of state anxiety upon completion of the psychosocial stress test. In conclusion, this study provides first insights into the efficacy of one single session of excitatory tDCS over the left DLPFC in attenuating autonomic and neuroendocrine effects of psychosocial stress exposure. These findings might be indicative of the important role of the left DLPFC, which is a cortical target for noninvasive brain stimulation treatment of depression, for successful coping with stressful stimuli.
Collapse
Affiliation(s)
- Luca Carnevali
- Department of Chemistry, Life Sciences and Environmental Sustainability, Stress Physiology Lab, University of Parma, Parma, Italy
| | - Elena Pattini
- Centro per la Cura, la Diagnosi e lo Studio dei Disturbi della Comunicazione e della Socializzazione, Ausl Parma, Parma, Italy
| | - Andrea Sgoifo
- Department of Chemistry, Life Sciences and Environmental Sustainability, Stress Physiology Lab, University of Parma, Parma, Italy
| | - Cristina Ottaviani
- Department of Psychology, Sapienza University of Rome, Rome, Italy
- Neuroimaging Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
50
|
Prefrontal parvalbumin cells are sensitive to stress and mediate anxiety-related behaviors in female mice. Sci Rep 2019; 9:19772. [PMID: 31875035 PMCID: PMC6930291 DOI: 10.1038/s41598-019-56424-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 12/10/2019] [Indexed: 11/08/2022] Open
Abstract
Reduced activity of the prefrontal cortex (PFC) is seen in mood disorders including depression and anxiety. The mechanisms of this hypofrontality remain unclear. Because of their specific physiological properties, parvalbumin-expressing (PV+) inhibitory interneurons contribute to the overall activity of the PFC. Our recent work using a chronic stress mouse model showed that stress-induced increases in prefrontal PV expression correlates with increased anxiety-like behaviors in female mice. Our goal is now to provide a causal relationship between changes in prefrontal PV+ cells and changes in emotional behaviors in mice. We first show that, in addition to increasing overall level of PV expression, chronic stress increases the activity of prefrontal PV+ cells. We then used a chemogenetic approach to mimic the effects of chronic stress and specifically increase the activity of prefrontal PV+ cells. We observed that chemogenetic activation of PV+ cells caused an overall reduction in prefrontal activity, and that chronic activation of PV+ cells lead to increased anxiety-related behaviors in female mice only. These results demonstrate that activity of prefrontal PV+ cells could represent a novel sex-specific modulator of anxiety-related behaviors, potentially through changes in overall prefrontal activity. The findings also support the idea that prefrontal PV+ cells are worth further investigation to better understand mood disorders that are more prevalent in female populations.
Collapse
|