1
|
Dehghani A, Meftahi GH, Sahraei H. The administration of a phentolamine infusion into the basolateral amygdala enhances long-term memory and diminishes anxiety-like behavior in stressed rats. Behav Pharmacol 2024; 35:419-431. [PMID: 39436284 DOI: 10.1097/fbp.0000000000000796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
The basolateral amygdala (BLA) contains adrenergic receptors, which are known to be involved in stress, anxiety, and memory. The objective of this study was to explore whether inhibition of α-adrenergic receptors (by phentolamine, an α-adrenergic receptor antagonist) in the BLA can reduce foot-shock stress-induced anxiety-like behavior, memory deficits, and long-term potentiation (LTP) deficits within the CA1 region of the rat hippocampus. Forty male Wistar rats were assigned to the intact, control, stress (Str), Phent (phentolamine), and Phent + Str groups. Animals were subjected to six shocks on 4 consecutive days, and phentolamine was injected into BLA 20 min before the animals were placed in the foot-shock stress apparatus. Results from the elevated plus maze test (EPM) revealed a reduction in anxiety-like behaviors (by an increased number of entries into the open arm, percentage of time spent in the open arm, and rearing and freezing) among stressed animals upon receiving injections of phentolamine into the BLA. The open-field test results (increased rearing, grooming, and freezing behaviors) were consistent with the EPM test results. Phentolamine infusion into the BLA enhanced spatial memory, reducing errors in finding the target hole and decreasing latency time in the Barnes maze test for stress and nonstress conditions. Injecting phentolamine into the BLA on both sides effectively prevented LTP impairment in hippocampal CA1 neurons after being subjected to foot-shock stress. It has been suggested that phentolamine in the BLA can effectively improve anxiety-like behaviors and memory deficits induced by foot-shock stress.
Collapse
Affiliation(s)
| | | | - Hedayat Sahraei
- Neuroscience Research Center
- Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Sancho-Balsells A, Borràs-Pernas S, Brito V, Alberch J, Girault JA, Giralt A. Cognitive and Emotional Symptoms Induced by Chronic Stress Are Regulated by EGR1 in a Subpopulation of Hippocampal Pyramidal Neurons. Int J Mol Sci 2023; 24:ijms24043833. [PMID: 36835243 PMCID: PMC9962724 DOI: 10.3390/ijms24043833] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Chronic stress is a core risk factor for developing a myriad of neurological disorders, including major depression. The chronicity of such stress can lead to adaptive responses or, on the contrary, to psychological maladaptation. The hippocampus is one of the most affected brain regions displaying functional changes in chronic stress. Egr1, a transcription factor involved in synaptic plasticity, is a key molecule regulating hippocampal function, but its role in stress-induced sequels has been poorly addressed. Emotional and cognitive symptoms were induced in mice by using the chronic unpredictable mild stress (CUMS) protocol. We used inducible double-mutant Egr1-CreERT2 x R26RCE mice to map the formation of Egr1-dependent activated cells. Results show that short- (2 days) or long-term (28 days) stress protocols in mice induce activation or deactivation, respectively, of hippocampal CA1 neural ensembles in an Egr1-activity-dependent fashion, together with an associated dendritic spine pathology. In-depth characterization of these neural ensembles revealed a deep-to-superficial switch in terms of Egr1-dependent activation of CA1 pyramidal neurons. To specifically manipulate deep and superficial pyramidal neurons of the hippocampus, we then used Chrna7-Cre (to express Cre in deep neurons) and Calb1-Cre mice (to express Cre in superficial neurons). We found that specific manipulation of superficial but not deep pyramidal neurons of the CA1 resulted in the amelioration of depressive-like behaviors and the restoration of cognitive impairments induced by chronic stress. In summary, Egr1 might be a core molecule driving the activation/deactivation of hippocampal neuronal subpopulations underlying stress-induced alterations involving emotional and cognitive sequels.
Collapse
Affiliation(s)
- Anna Sancho-Balsells
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Casanova 143, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 08036 Barcelona, Spain
| | - Sara Borràs-Pernas
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Casanova 143, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 08036 Barcelona, Spain
| | - Verónica Brito
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Casanova 143, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 08036 Barcelona, Spain
| | - Jordi Alberch
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Casanova 143, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 08036 Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036 Barcelona, Spain
| | - Jean-Antoine Girault
- Inserm UMR-S 1270, 75005 Paris, France
- Science and Engineering Faculty, Sorbonne Université, 75005 Paris, France
- Institut du Fer à Moulin, 75005 Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina, Institut de Neurociències, Universitat de Barcelona, Casanova 143, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 08036 Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, 08036 Barcelona, Spain
- Correspondence: ; Tel.: +34-934037980
| |
Collapse
|
3
|
Thompson SM. Plasticity of synapses and reward circuit function in the genesis and treatment of depression. Neuropsychopharmacology 2023; 48:90-103. [PMID: 36057649 PMCID: PMC9700729 DOI: 10.1038/s41386-022-01422-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/18/2022] [Accepted: 08/01/2022] [Indexed: 11/08/2022]
Abstract
What changes in brain function cause the debilitating symptoms of depression? Can we use the answers to this question to invent more effective, faster acting antidepressant drug therapies? This review provides an overview and update of the converging human and preclinical evidence supporting the hypothesis that changes in the function of excitatory synapses impair the function of the circuits they are embedded in to give rise to the pathological changes in mood, hedonic state, and thought processes that characterize depression. The review also highlights complementary human and preclinical findings that classical and novel antidepressant drugs relieve the symptoms of depression by restoring the functions of these same synapses and circuits. These findings offer a useful path forward for designing better antidepressant compounds.
Collapse
Affiliation(s)
- Scott M Thompson
- Department of Psychiatry, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, 80045, CO, USA.
| |
Collapse
|
4
|
Electroacupuncture Enhances Cognitive Deficits in a Rat Model of Rapid Eye Movement Sleep Deprivation via Targeting MiR-132. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:7044208. [PMID: 36159559 PMCID: PMC9507748 DOI: 10.1155/2022/7044208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/12/2022] [Accepted: 08/13/2022] [Indexed: 11/17/2022]
Abstract
Deprivation of rapid eye movement sleep (REMSD) reduces the potential for learning and memory. The neuronal foundation of cognitive performance is synapse plasticity. MicroRNA-132 (MiR-132) is an important microRNA related to cognitive and synapse plasticity. Acupuncture is effective at improving cognitive impairment caused by sleep deprivation. Furthermore, its underlying principle is still unclear. Herein, whether electroacupuncture (EA) helps alleviate cognitive impairment in REMSD by targeting miR-132 was assessed. A rat model of REMSD was constructed using the developing multiplatform water environment technique, as well as EA therapy in Baihui (GV20) and Dazhui (GV14) was performed for 15 minutes, once daily for 7 days. Agomir or antagomir of MiR-132 was injected into the hippocampal CA1 areas to assess the EA mechanism in rats with REMSD. Then, the learning and memory abilities were detected by behavioral tests; synapse structure was assessed by transmission electron microscope (TCM); and dendrites branches and length were examined by Golgi staining. MiR-132-3p was assessed by real-time quantitative polymerase chain reaction (qRT-PCR). P250GAP, ras-related C3 botulinum toxin substrate 1 (Rac1), and cell division cycle 42 (Cdc42) expression levels in hippocampal tissues were evaluated by immunohistochemistry and Western blot. According to the research, EA therapy enhanced cognitive in REMSD rats, as evidenced by reduced escape latency; upregulated the performance of platform crossings and prolonged duration in the goal region; and improved spontaneous alternation. EA administration restored synaptic and dendritic structural damage in hippocampal neurons, enhanced miR-132 expression, and reduced p250GAP mRNA and protein levels. Additionally, EA boosted the protein level of Rac1 and Cdc42 associated with synaptic plasticity. MiR-132 agomir enhanced this effect, whereas miR-13 antagomir reversed this action. The current data demonstrate that EA at GV20 and GV14 attenuates cognitive impairment and modulates synaptic plasticity in hippocampal neurons via miR-132 in a sleep-deprived rat model.
Collapse
|
5
|
Wang B, Chen D, Jiang R, Ntim M, Lu J, Xia M, Yang X, Wang Y, Kundu S, Guan R, Li S. TIP60 buffers acute stress response and depressive behaviour by controlling PPARγ-mediated transcription. Brain Behav Immun 2022; 101:410-422. [PMID: 35114329 DOI: 10.1016/j.bbi.2022.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/29/2021] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Tat-interacting protein 60 (TIP60) as nuclear receptors (NRs) coregulator, acts as a tumor suppressor and also has promising therapeutic potential to target Alzheimer's disease. Stress has been implicated in many psychiatric disorders, and these disorders are characterized by impairments in cognitive function. Until now, there are no experimental data available on the regulatory effect of TIP60 in acute stress and depression. There is also no definitive explanation on which specific modulation of target gene expression is achieved by TIP60. Here, we identify TIP60 as a novel positive regulator in response to acute restraint stress (ARS) and a potentially effective target of antidepressants. Firstly, we discovered increased hippocampal TIP60 expressions in the ARS model. Furthermore, using the TIP60 inhibitor, MG149, we proved that TIP60 function correlates with behavioral and synaptic activation in the two-hour ARS. Secondly, the lentivirus vector (LV)-TIP60overexpression (OE) was injected into the hippocampus prior to the chronic restraint stress (CRS) experiments and it was found that over-expressed TIP60 compensates for TIP60 decrease and improves depression index in CRS. Thirdly, through the intervention of TIP60 expression in vitro, we established the genetic regulation of TIP60 on synaptic proteins, confirmed the TIP60 function as a specific coactivator for PPARγ and found that the PPARγ-mediated TIP60 function modulates transcriptional activation of synaptic proteins. Finally, the LV-TIP60OE and PPARγ antagonist, GW9662, were both administered in the CRS model and the data indicated that blocking PPARγ significantly weakened the protective effect of TIP60 against the CRS-induced depression. Conclusively, these findings together support TIP60 as a novel positive factor in response to acute stress and interacts with PPARγ to modulate the pathological mechanism of CRS-induced depression.
Collapse
Affiliation(s)
- Bin Wang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Defang Chen
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Rong Jiang
- Department of Physiology, Binzhou Medical University, Yantai Campus, 346 Guanhai Road, Laishan District, Yantai, Shandong, China
| | - Michael Ntim
- Department of Physiology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jincheng Lu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Min Xia
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - XueWei Yang
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Ying Wang
- Department of Cardiology, Institute of Heart and Vessel Diseases of Dalian Medical University, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Supratik Kundu
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Rongxiao Guan
- Department of Cardiology, Institute of Heart and Vessel Diseases of Dalian Medical University, the Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shao Li
- Department of Physiology, College of Basic Medical Sciences, Liaoning Provincial Key Laboratory of Cerebral Diseases, National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
6
|
Rusakov DA, Giese KP, Sandi C, Dommett E, Overton PG. Remembering Mike Stewart. Neuropharmacology 2022; 207:108962. [PMID: 35051447 DOI: 10.1016/j.neuropharm.2022.108962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Dmitri A Rusakov
- UCL Queen Square Institute of Neurology, University College London, Queen Square, London, WC1N 3BG, UK.
| | - Karl Peter Giese
- Institute of Psychiatry, Physiology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Carmen Sandi
- Swiss Federal Institute of Technology in Lausanne (EPFL), Rte Cantonale, 1015, Lausanne, Switzerland
| | - Eleanore Dommett
- Institute of Psychiatry, Physiology and Neuroscience, King's College London, De Crespigny Park, London, SE5 8AF, UK
| | - Paul G Overton
- Department of Psychology, University of Sheffield, Vicar Lane, Sheffield, S1 2LT, UK
| |
Collapse
|
7
|
Rusakov DA. Obituary for Professor Michael G. Stewart: Life in Neurosciences. Brain Res Bull 2022; 180:147-149. [DOI: 10.1016/j.brainresbull.2022.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
8
|
A Novel Estimation Method for the Counting of Dendritic Spines. J Neurosci Methods 2021; 368:109454. [PMID: 34952089 DOI: 10.1016/j.jneumeth.2021.109454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/15/2021] [Accepted: 12/17/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND Since Cajal's visualisations of the synaptic spine, this feature of the neuron has been of interest to neuroscientists and has been investigated usually in reference to degeneration or proliferation of dendrites and their neurons. Synaptic spine measurement often forms a critical element of any study investigating neuronal morphology. However, the way researchers have counted spines hasn't changed for almost a century. Some of the currently used legacy methods fail to accommodate obscured pisnes or factor-in visibility differences between histological stains. NEW METHOD Here we investigate the neuronal dendrite and its synaptic spines, and reveal that using confocal or bright-field technologies may in fact obfuscate spine counts. A mathematical model is developed for the distribution of synaptic spines within the rat, that should, by nature of the formula and the impartiality of probability quotients, be applied to estimate the number of synaptic spines across any length of dendrite that has protrusions within any species. RESULTS Using this estimation method, we show that, depending on the method of image capture, there are in fact more spines present than typically counted on lengths of dendrite, something that may have biased morphological studies in the past. COMPARISON WITH EXISTING METHODS This new estimation method has been collapsed down into an easy-to-use free website. With input of only four fields, we provide the researcher with a more accurate estimation of the amount of spines on a length of dendrite. This was made possible by fluorescing a Golgi stain and comparing two-photon, bright-field and confocal images. CONCLUSIONS An easy web-based resource has been made available to use this new method for spine calculation. Using this method improves the validity of spine measurement and provides a means to review previously published work.
Collapse
|
9
|
Zhvania M, Japaridze N, Tizabi Y, Sharikadze I, Pochkhidze N, Cheishvili L. Anxiety and ultrastructural consequences of chronic mild stress in rats. Neurosci Lett 2021; 771:136390. [PMID: 34896437 DOI: 10.1016/j.neulet.2021.136390] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/05/2021] [Indexed: 10/19/2022]
Abstract
Detrimental consequences following exposure to severe stress, either acute or chronic are well recognized. Chronic mild stress (CMS) is also a leading cause of emotional distress and neuropsychiatric conditions such as anxiety disorders. However, the neurobiological substrates of the latter, particularly at the ultrastructural levels have not been adequately investigated. In this study, adult male Wistar rats were subjected to 4 h daily mild restraint for 20 days and their behavior in open field and elevated plus maze (EPM) were evaluated 24 h after the last restraint. Anxiety-like behavior was evident in CMS exposed rats by increases in rearing and grooming in the open field and the avoidance of open arms in the EPM. Concomitant ultrastructural alterations such as chromatolysis, agglutination of synaptic vesicles or mitochondrial damage were also observed in the central nucleus of amygdala (CNA), an area intimately involved in emotional and fear response, in CMS exposed rats. These results while confirming detrimental consequences of CMS, also suggest that ultrastructural alterations in CNA may be a basis for CMS-induced anxiety.
Collapse
Affiliation(s)
- Mzia Zhvania
- School of Natural Sciences and Medicine, Ilia State University. 3/5 K. Cholokashvili Avenue, Tbilisi 0162, Georgia; Department of Brain Ultrastructure and Nanoarchitecture I. Beritashvili Center of Experimental Biomedicine. 14 Gotua Street, Tbilisi 0160, Georgia.
| | - Nadezhda Japaridze
- Department of Brain Ultrastructure and Nanoarchitecture I. Beritashvili Center of Experimental Biomedicine. 14 Gotua Street, Tbilisi 0160, Georgia; School of Medicine, New Vision University. 1A Evgeni Mikeladze Street, Tbilisi 0159, Georgia
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, USA
| | - Irina Sharikadze
- School of Natural Sciences and Medicine, Ilia State University. 3/5 K. Cholokashvili Avenue, Tbilisi 0162, Georgia
| | - Nino Pochkhidze
- School of Natural Sciences and Medicine, Ilia State University. 3/5 K. Cholokashvili Avenue, Tbilisi 0162, Georgia; Department of Brain Ultrastructure and Nanoarchitecture I. Beritashvili Center of Experimental Biomedicine. 14 Gotua Street, Tbilisi 0160, Georgia
| | - Levan Cheishvili
- School of Natural Sciences and Medicine, Ilia State University. 3/5 K. Cholokashvili Avenue, Tbilisi 0162, Georgia
| |
Collapse
|
10
|
Huzian O, Baka J, Csakvari E, Dobos N, Leranth C, Siklos L, Duman RS, Farkas T, Hajszan T. Stress Resilience is Associated with Hippocampal Synaptoprotection in the Female Rat Learned Helplessness Paradigm. Neuroscience 2021; 459:85-103. [PMID: 33524494 DOI: 10.1016/j.neuroscience.2021.01.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 01/11/2021] [Accepted: 01/21/2021] [Indexed: 12/28/2022]
Abstract
The synaptogenic hypothesis of major depressive disorder implies that preventing the onset of depressive-like behavior also prevents the loss of hippocampal spine synapses. By applying the psychoactive drugs, diazepam and fluoxetine, we investigated whether blocking the development of helpless behavior by promoting stress resilience in the rat learned helplessness paradigm is associated with a synaptoprotective action in the hippocampus. Adult ovariectomized and intact female Sprague-Dawley rats (n = 297) were treated with either diazepam, fluoxetine, or vehicle, exposed to inescapable footshocks or sham stress, and tested in an active escape task to assess helpless behavior. Escape-evoked corticosterone secretion, as well as remodeling of hippocampal spine synapses at a timepoint representing the onset of escape testing were also analyzed. In ovariectomized females, treatment with diazepam prior to stress exposure prevented helpless behavior, blocked the loss of hippocampal spine synapses, and muted the corticosterone surge evoked by escape testing. Although fluoxetine stimulated escape performance and hippocampal synaptogenesis under non-stressed conditions, almost all responses to fluoxetine were abolished following exposure to inescapable stress. Only a much higher dose of fluoxetine was capable of partly reproducing the strong protective actions of diazepam. Importantly, these protective actions were retained in the presence of ovarian hormones. Our findings indicate that stress resilience is associated with the preservation of spine synapses in the hippocampus, raising the possibility that, besides synaptogenesis, hippocampal synaptoprotection is also implicated in antidepressant therapy.
Collapse
Affiliation(s)
- Orsolya Huzian
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Judith Baka
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Eszter Csakvari
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Nikoletta Dobos
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Csaba Leranth
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States; Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States
| | - Laszlo Siklos
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary
| | - Ronald S Duman
- Department of Psychiatry, Yale University School of Medicine, 34 Park Street, New Haven, CT 06508, United States; Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, United States
| | - Tamas Farkas
- Department of Physiology, Anatomy, and Neuroscience, University of Szeged Faculty of Science and Informatics, Kozep Fasor 52, 6726 Szeged, Hungary
| | - Tibor Hajszan
- Institute of Biophysics, Biological Research Center, Temesvari Krt 62, 6726 Szeged, Hungary; Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, United States.
| |
Collapse
|
11
|
Cursano S, Battaglia CR, Urrutia-Ruiz C, Grabrucker S, Schön M, Bockmann J, Braumüller S, Radermacher P, Roselli F, Huber-Lang M, Boeckers TM. A CRHR1 antagonist prevents synaptic loss and memory deficits in a trauma-induced delirium-like syndrome. Mol Psychiatry 2021; 26:3778-3794. [PMID: 32051550 PMCID: PMC8550963 DOI: 10.1038/s41380-020-0659-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/10/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022]
Abstract
Older patients with severe physical trauma are at high risk of developing neuropsychiatric syndromes with global impairment of cognition, attention, and consciousness. We employed a thoracic trauma (TxT) mouse model and thoroughly analyzed age-dependent spatial and temporal posttraumatic alterations in the central nervous system. Up to 5 days after trauma, we observed a transient 50% decrease in the number of excitatory synapses specifically in hippocampal pyramidal neurons accompanied by alterations in attention and motor activity and disruption of contextual memory consolidation. In parallel, hippocampal corticotropin-releasing hormone (CRH) expression was highly upregulated, and brain-derived neurotrophic factor (BDNF) levels were significantly reduced. In vitro experiments revealed that CRH application induced neuronal autophagy with rapid lysosomal degradation of BDNF via the NF-κB pathway. The subsequent synaptic loss was rescued by BDNF as well as by specific NF-κB and CRH receptor 1 (CRHR1) antagonists. In vivo, the chronic application of a CRHR1 antagonist after TxT resulted in reversal of the observed histological, molecular, and behavioral alterations. The data suggest that neuropsychiatric syndromes (i.e., delirium) after peripheral trauma might be at least in part due to the activation of the hippocampal CRH/NF-κB/BDNF pathway, which results in a dramatic loss of synaptic contacts. The successful rescue by stress hormone receptor antagonists should encourage clinical trials focusing on trauma-induced delirium and/or other posttraumatic syndromes.
Collapse
Affiliation(s)
- Silvia Cursano
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany ,International Graduate School in Molecular Medicine, IGradU, 89081 Ulm, Germany
| | - Chiara R. Battaglia
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany ,International Graduate School in Molecular Medicine, IGradU, 89081 Ulm, Germany
| | - Carolina Urrutia-Ruiz
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Stefanie Grabrucker
- grid.10049.3c0000 0004 1936 9692Department of Biological Sciences, University of Limerick, Limerick, V94 PH61 Ireland
| | - Michael Schön
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Jürgen Bockmann
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| | - Sonja Braumüller
- grid.6582.90000 0004 1936 9748Institute for Anesthesiological Pathophysiology, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Peter Radermacher
- grid.6582.90000 0004 1936 9748Institute for Anesthesiological Pathophysiology, Ulm University, Helmholtzstr. 8/1, 89081 Ulm, Germany
| | - Francesco Roselli
- grid.6582.90000 0004 1936 9748Clinic for Neurology, Ulm University, 89081 Ulm, Germany
| | - Markus Huber-Lang
- grid.6582.90000 0004 1936 9748Institute of Clinical and Experimental Trauma-Immunology, Ulm University, 89081 Ulm, Germany
| | - Tobias M. Boeckers
- grid.6582.90000 0004 1936 9748Institute for Anatomy and Cell Biology, Ulm University, Albert-Einstein-Allee 11, 89081 Ulm, Germany
| |
Collapse
|
12
|
Sun DS, Zhong G, Cao HX, Hu Y, Hong XY, Li T, Li X, Liu Q, Wang Q, Ke D, Liu GP, Ma RH, Luo DJ. Repeated Restraint Stress Led to Cognitive Dysfunction by NMDA Receptor-Mediated Hippocampal CA3 Dendritic Spine Impairments in Juvenile Sprague-Dawley Rats. Front Mol Neurosci 2020; 13:552787. [PMID: 33192290 PMCID: PMC7604534 DOI: 10.3389/fnmol.2020.552787] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/19/2020] [Indexed: 11/13/2022] Open
Abstract
Although numerous studies have indicated that chronic stress causes cognitive dysfunction with the impairment of synaptic structures and functions, the relationship between cognitive deficits induced by repeated restraint stress and the level of NMDA receptors in the subregion of the hippocampus has been relatively unknown until now. In this study, 3-week-old male Sprague-Dawley rats were exposed to repeated restraint stress for seven consecutive days, their cognitive functions were evaluated through behavioral tests, and then they were sacrificed for electrophysiological, morphological, and biochemical assays. Chronic repeated restraint stress led to cognitive and electrophysiological impairments, with a reduced density of dendritic spines. We also found that the protein level of NMDA receptors only increased in the hippocampal CA3 region. Nevertheless, repeated restraint stress-induced cognitive and synaptic dysfunction were effectively reversed by Ro25-6981, an inhibitor of the GluN2B receptor. These findings suggest that repeated restraint stress-induced synaptic and cognitive deficits are probably mediated through NMDA receptors.
Collapse
Affiliation(s)
- Dong-Sheng Sun
- Institute of Anesthesiology & Pain (IAP), Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Gang Zhong
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Xia Cao
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Yue Hong
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Li
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qun Wang
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Ke
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Key Laboratory of Ministry of Education of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Rong-Hong Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Ju Luo
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Deonaraine KK, Wang Q, Cheng H, Chan KL, Lin HY, Liu K, Parise LF, Cathomas F, Leclair KB, Flanigan ME, Li L, Aleyasin H, Guevara C, Hao K, Zhang B, Russo SJ, Wang J. Sex-specific peripheral and central responses to stress-induced depression and treatment in a mouse model. J Neurosci Res 2020; 98:2541-2553. [PMID: 32918293 DOI: 10.1002/jnr.24724] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 06/17/2020] [Accepted: 08/15/2020] [Indexed: 11/11/2022]
Abstract
Major depressive disorder affects ~20% of the world population and is characterized by strong sexual dimorphism with females being two to three times more likely to develop this disorder. Previously, we demonstrated that a combination therapy with dihydrocaffeic acid and malvidin-glucoside to synergistically target peripheral inflammation and stress-induced synaptic maladaptation in the brain was effective in alleviating chronic social defeat stress (CSDS)-induced depression-like phenotype in male mice. Here, we test the combination therapy in a female CSDS model for depression and compared sex-specific responses to stress in the periphery and the central nervous system. Similar to male mice, the combination treatment is also effective in promoting resilience against the CSDS-induced depression-like behavior in female mice. However, there are sex-specific differences in peripheral immune responses and differential gene regulation in the prefrontal cortex to chronic stress and to the treatment. These data indicate that while therapeutic approaches to combat stress-related disorders may be effective in both sexes, the mechanisms underlying these effects differ, emphasizing the need for inclusion of both sexes in preclinical studies using animal models.
Collapse
Affiliation(s)
- Kristina K Deonaraine
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qian Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute of Genomics and Multi-Scale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Haoxiang Cheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kenny L Chan
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hsiao-Yun Lin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kalena Liu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lyonna F Parise
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Flurin Cathomas
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Katherine B Leclair
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meghan E Flanigan
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Long Li
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hossein Aleyasin
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher Guevara
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ke Hao
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute of Genomics and Multi-Scale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott J Russo
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jun Wang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Geriatric Research, Education and Clinical Center, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
| |
Collapse
|
14
|
Huang C, Chen JT. Chronic retinoic acid treatment induces affective disorders by impairing the synaptic plasticity of the hippocampus. J Affect Disord 2020; 274:678-689. [PMID: 32664002 DOI: 10.1016/j.jad.2020.05.114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/18/2020] [Accepted: 05/17/2020] [Indexed: 11/26/2022]
Abstract
BACKGROUND More and more people are suffering from depression in modern society. It is believed that the development of depression results from alterations in synaptic transmission, especially in the hippocampus. Animal experiments and clinical studies have demonstrated that retinoids are essential components in hippocampal synaptic plasticity, and they have a close relationship with depression. However, it is still unclear how excessive retinoic acid (RA) causes depression and what synaptic and molecular mechanisms underlie it. METHODS Behavioral, electrophysiological, and molecular approaches were employed to characterize the effects of RA on depression and synaptic plasticity. RA was continuously administered intracerebroventricularly through an osmotic pump. RESULTS RA treatment induced depression-like behaviors, as evidenced by decreased sucrose preference and increased immobile duration in both the forced swim test and the tail suspension test. RA administration also induced anxiety-like behaviors, indicated by decreased duration in the open arms of the elevated plus maze and the central of the open field. RA treatment decreased the neuronal excitability of the hippocampus either by changing the excitatory/inhibitory receptor balance or by promoting the synthesis of inhibitory neurotransmitters. Moreover, long-term potentiation was decreased in both the excitatory postsynaptic potential and the population spike in RA-treated rats, presumably a consequence of the reduced glur1 transcript level. LIMITATIONS The mechanism of how excess RA affects the hippocampal gene expression and synaptic plasticity requires further study. CONCLUSIONS RA treatment can induce depression-like behavior in rats and impair hippocampal plasticity. Thus, improving synaptic plasticity in the hippocampus may ameliorate the affective disorders caused by excessive RA.
Collapse
Affiliation(s)
- Chuan Huang
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| | - Ju-Tao Chen
- Chinese Academy of Science Key Laboratory of Brain Function and Diseases, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| |
Collapse
|
15
|
Zhu D, Sun M, Liu Q, Yue Y, Lu J, Lin X, Shi J. Angiotensin (1-7) through modulation of the NMDAR-nNOS-NO pathway and serotonergic metabolism exerts an anxiolytic-like effect in rats. Behav Brain Res 2020; 390:112671. [PMID: 32437889 DOI: 10.1016/j.bbr.2020.112671] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/05/2020] [Accepted: 04/21/2020] [Indexed: 01/02/2023]
Abstract
Although recent studies have shown that angiotensin (1-7) (Ang [1-7]) exerts anti-stress and anxiolytic-like effects, the underlying mechanisms remain elusive. The ventral hippocampus (VH) is proposed to be a critical brain region for mood and stress management through the N-methyl-d-aspartate receptor (NMDAR) signaling pathway. However, the role of VH NMDAR signaling in the effects of Ang (1-7) remains unclear. In the present study, Ang (1-7) was injected into the bilateral VH of stressed rats, or in combination with a Fyn kinase inhibitor, NMDAR antagonist, neuronal nitric oxide synthase (nNOS) inhibitor, or nitric oxide (NO) scavenger. Anxiety-like behaviors were assessed using the open field test and elevated plus maze test, while alterations in NMDAR-nNOS-NO signaling and serotonergic metabolism were examined in the VH. After 21 days of chronic restraint stress, anxiety-like behaviors were evident. Levels of phosphorylated NR2B (a key NMDAR subunit), its upstream kinase Fyn, as well as activity of nNOS and monoamine oxidase (MAO) were markedly reduced. In contrast, levels of serotonin were increased. Bilateral VH infusion of Ang (1-7) recovered NMDAR-nNOS-NO signaling and MAO-mediated serotonin metabolism, as well as reducing anxiety-like behaviors in stressed rats. These effects were diminished by blockade of MasR (Ang [1-7]-specific receptor), Fyn kinase, NMDAR, nNOS, or NO production. Altogether, these findings indicate that Ang (1-7) exerts anxiolytic effects through modulation of the NMDAR-nNOS-NO pathway and serotonergic metabolism. Future translational research should focus on the relationship between Ang (1-7), glutamatergic neurotransmission, and serotonergic neurotransmission in the VH.
Collapse
Affiliation(s)
- Donglin Zhu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Ming Sun
- Emergency Department, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, PR China
| | - Qinqin Liu
- Department of Neurology, The Affiliated Suqian Hospital of Xuzhou Medical University, Suqian, PR China
| | - Yu Yue
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Jie Lu
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Xingjian Lin
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China
| | - Jingping Shi
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, PR China.
| |
Collapse
|
16
|
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
|
17
|
Sandini C, Chambaz M, Schneider M, Armando M, Zöller D, Schaer M, Sandi C, Van De Ville D, Eliez S. Pituitary dysmaturation affects psychopathology and neurodevelopment in 22q11.2 Deletion Syndrome. Psychoneuroendocrinology 2020; 113:104540. [PMID: 31958652 DOI: 10.1016/j.psyneuen.2019.104540] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 12/03/2019] [Accepted: 12/04/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND 22q11.2 Deletion Syndrome (22q11DS) confers strongly increased genetic risk for multiple psychiatric disorders. Similarly to the general population, rates of psychiatric comorbidity suggest that common disease mechanisms are shared across dimensions of psychopathology. Such pleiotropic disease mechanisms remain however currently unknown. We hypothesized that pituitary dysmaturation, indicative of HPA-axis dysregulation, could correlate to reduced tolerance to daily life stressors and reflect pleiotropic risk factor for psychopathology. Moreover HPA-axis dysregulation could affect atypical cortical and hippocampal development previously described in 22q11DS. METHODS Pituitary volume, hippocampal volume and cortical thickness measures were obtained from T1-weighted MRI images in a large longitudinal cohort of youth with 22q11DS (115 subjects, 260 scans, age-range = 5.4-31.6) and healthy controls (151 subjects, 280 scans, age-range = 5.1-32.3). We explored effects of pituitary dysmaturation on tolerance to stress, psychopathology and neurodevelopment employing mixed-models linear regression. Associations of pituitary and cortical development were correlated with the expression pattern of glucocorticoid receptor gene NR3C1 obtained from the Allen-Human-Brain-Atlas. RESULTS We observed aberrant pituitary developmental trajectories in 22q11DS, with volumetric reductions emerging by young-adulthood (P = 0.0006). Longitudinal pituitary decline was associated with to reduced tolerance to stress (P = 0.04), higher overall psychopathology (P = 0.0003) and increased risk of psychiatric comorbidity (P = 0.02). Moreover, pituitary decline correlated with blunted growth of the right hippocampus (P = 0.03) and to increased cortical thinning of mostly temporal and orbitofrontal regions mediated by NR3C1 gene expression. CONCLUSION Atypical pituitary development could reflect progressive extinction of HPAA due to chronic hyper-activation, in agreement with existing biochemical evidence in 22q11DS. HPAA dysregulation could represent and endophenotype that confers pleiotropic vulnerability to psychopathology and atypical neurodevelopment in 22q11DS.
Collapse
Affiliation(s)
- Corrado Sandini
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland.
| | - Maëlle Chambaz
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Maude Schneider
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Center for Contextual Psychiatry, Research Group Psychiatry, Department of Neuroscience, KU Leuven, Leuven, Belgium
| | - Marco Armando
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Daniela Zöller
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Marie Schaer
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland
| | - Carmen Sandi
- Brain Mind Institute, School of Life Sciences, École Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Dimitri Van De Ville
- Department of Radiology and Medical Informatics, University of Geneva, Switzerland; Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Stephan Eliez
- Developmental Imaging and Psychopathology Laboratory, University of Geneva School of Medicine, Geneva, Switzerland; Department of Genetic Medicine and Development, University of Geneva School of Medicine, Geneva, Switzerland
| |
Collapse
|
18
|
Klebe D, Tibrewal M, Sharma DR, Vanaparthy R, Krishna S, Varghese M, Cheng B, Mouton PR, Velíšková J, Dobrenis K, Hof PR, Ballabh P. Reduced Hippocampal Dendrite Branching, Spine Density and Neurocognitive Function in Premature Rabbits, and Reversal with Estrogen or TrkB Agonist Treatment. Cereb Cortex 2019; 29:4932-4947. [PMID: 30877788 PMCID: PMC6918929 DOI: 10.1093/cercor/bhz033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 01/16/2019] [Accepted: 02/08/2019] [Indexed: 01/02/2023] Open
Abstract
Preterm-born children suffer from neurological and behavioral disorders. Herein, we hypothesized that premature birth and non-maternal care of preterm newborns might disrupt neurobehavioral function, hippocampal dendritic arborization, and dendritic spine density. Additionally, we assessed whether 17β-estradiol (E2) replacement or the TrkB receptor agonist, 7,8-dihydroxyflavone (DHF), would reverse compromised dendritic development and cognitive function in preterm newborns. These hypotheses were tested by comparing preterm (E28.5) rabbit kits cared and gavage-fed by laboratory personnel and term-kits reared and breast-fed by their mother doe at an equivalent postconceptional age. Neurobehavioral tests showed that both premature-birth and formula-feeding with non-maternal care led to increased anxiety behavior, poor social interaction, and lack of novelty preference compared with term-kits. Dendritic branching and number of total or mushroom dendritic spines were reduced in the CA1 field of preterm-kits compared with term controls. While CDC42 and Rac1/2/3 expression levels were lower, RhoA-activity was higher in preterm-kits compared with term controls. Both E2 and DHF treatment reversed prematurity-induced reduction in spine density, reduced total RhoA-GTPase levels, and enhanced cognitive function. Hence, prematurity and non-maternal care result in cognitive deficits, and reduced dendritic arbors and spines in CA1. E2 replacement or DHF treatment might reverse changes in dendritic spines and improve neurodevelopment in premature infants.
Collapse
Affiliation(s)
- Damon Klebe
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Mahima Tibrewal
- Department of Pediatrics, New York Medical College, Valhalla NY, USA
| | - Deep R Sharma
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Rachna Vanaparthy
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Sunil Krishna
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Merina Varghese
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Bokun Cheng
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Peter R Mouton
- Department of Pathology and Cell Biology, College of Medicine, University of South Florida, Tampa FL, USA
| | - Jana Velíšková
- Departments of Cell Biology & Anatomy, Neurology, and Obstetrics & Gynecology, New York Medical College, Valhalla NY, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| |
Collapse
|
19
|
Feng X, Zhao Y, Yang T, Song M, Wang C, Yao Y, Fan H. Glucocorticoid-Driven NLRP3 Inflammasome Activation in Hippocampal Microglia Mediates Chronic Stress-Induced Depressive-Like Behaviors. Front Mol Neurosci 2019; 12:210. [PMID: 31555091 PMCID: PMC6727781 DOI: 10.3389/fnmol.2019.00210] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/16/2019] [Indexed: 12/17/2022] Open
Abstract
Chronic stress is a key risk factor for depression, and microglia have been implicated in the pathogenesis of the disease. Recent studies show that the Nod-like receptor protein 3 (NLRP3) inflammasome is expressed in microglia and may play a crucial role in depression. However, the mechanism of NLRP3 inflammasome activation in hippocampal microglia and its role in depressive-like behaviors remain poorly understood. In this study, rats were subjected to 6 h of restraint stress per day for 21 days to produce a model of stress-induced depression. Behavioral tests and serum corticosterone were used to assess the success of the model. Furthermore, HAPI cells were pretreated with dexamethasone (5 × 10-7 M) to assess stress-induced changes in microglial cells in culture. The microglial marker Iba-1, reactive oxygen species (ROS), nuclear factor kappa B (NF-κB) and key components of the NLRP3 inflammasome and its downstream inflammatory effectors (IL-1β and IL-18) were measured. Chronic stress induced depressive-like behavior, increased serum corticosterone levels and produced hippocampal structural changes. Chronic stress and dexamethasone both increased Iba-1 expression and ROS formation and also elevated levels of NF-κB, NLRP3, cleaved caspase-1, IL-1β and IL-18. After use of the NF-κB inhibitor BAY 117082 and knocked out NLRP3 in vitro decreased ROS formation and the expression of Iba-1, NF-κB and NLRP3 as well as levels of cleaved caspase-1, IL-1β and IL-18. These findings suggest that activation of the glucocorticoid receptor-NF-κB-NLRP3 pathway in hippocampal microglia mediates chronic stress-induced hippocampal neuroinflammation and depression-like behavior.
Collapse
Affiliation(s)
- Xiujing Feng
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Manyu Song
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Chaoran Wang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yujie Yao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
20
|
Moradi-Kor N, Ghanbari A, Rashidipour H, Yousefi B, Bandegi AR, Rashidy-Pour A. Beneficial effects of Spirulina platensis, voluntary exercise and environmental enrichment against adolescent stress induced deficits in cognitive functions, hippocampal BDNF and morphological remolding in adult female rats. Horm Behav 2019; 112:20-31. [PMID: 30917909 DOI: 10.1016/j.yhbeh.2019.03.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 03/16/2019] [Accepted: 03/22/2019] [Indexed: 11/21/2022]
Abstract
Chronic exposure to stress during adolescent period has been demonstrated to impair cognitive functions and the dendritic morphology of pyramidal neurons in the rat hippocampal CA3 area. The present study investigated the combined protective effects of Spirulina platensis (SP), a supplement made from blue-green algae with neuroprotective properties, voluntary exercise (EX) and environmental enrichment (EE) against cognitive deficits, alternations in hippocampal BDNF levels, and abnormal neuronal remodeling in adult female rats (PND 60) induced by exposure to chronic restraint stress during adolescent period (PND 30-40). Rats were exposed to restraint stress (2 h/day for 10 days, PND 30-40). Then, the animals were subjected to treatment with SP (200 mg/kg/day), EX, EE and the combined treatments (SP + EX, and SP + EE) between PND 41 and 55 of age. Following the interventions, spatial learning and memory, passive avoidance performance, hippocampal dendritic morphology and BDNF levels were assessed. Results showed that plasma corticosterone levels increased at PND 40 and remained elevated at PND 55 and 70 in the stressed rats. Stressed rats showed deficits in spatial learning and memory and passive avoidance performance, decreased BDNF levels in the hippocampus, and reduced apical dendritic length and branch points of the CA3 pyramidal neurons. These deficits were alleviated by the SP, EX and EE, and the combined treatments, which accompanied with a decline in serum corticosterone in stressed animals. Some treatments even enhanced cognitive functions, and BDNF levels and neuroanatomical remodeling in the hippocampus of non-stressed animals. Our findings provide important evidences that physical activity, exposure to EE, and the SP treatment during adolescent period can protect against adolescent stress induced behavioral, biochemical and neuroanatomical impairments in adulthood.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Avoidance Learning/drug effects
- Avoidance Learning/physiology
- Brain-Derived Neurotrophic Factor/metabolism
- Cell Extracts/pharmacology
- Cognition/drug effects
- Cognition/physiology
- Cognition Disorders/etiology
- Cognition Disorders/pathology
- Cognition Disorders/physiopathology
- Cognition Disorders/prevention & control
- Conditioning, Psychological/drug effects
- Conditioning, Psychological/physiology
- Female
- Hippocampus/drug effects
- Hippocampus/metabolism
- Hippocampus/pathology
- Hippocampus/physiopathology
- Memory/drug effects
- Memory/physiology
- Neuronal Plasticity/drug effects
- Physical Conditioning, Animal/physiology
- Rats
- Rats, Wistar
- Restraint, Physical/physiology
- Restraint, Physical/psychology
- Sexual Maturation/drug effects
- Sexual Maturation/physiology
- Social Environment
- Spatial Learning/drug effects
- Spirulina/chemistry
- Stress, Psychological/complications
- Stress, Psychological/metabolism
- Stress, Psychological/pathology
- Stress, Psychological/psychology
Collapse
Affiliation(s)
- Nasroallah Moradi-Kor
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran; Laboratory of Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Ghanbari
- Laboratory of Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Hadi Rashidipour
- School of Veterinary Medicine, Islamic Azad University, Garmsar Branch, Garmsar, Iran
| | - Behpour Yousefi
- Department of Anatomical Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Ahmad Reza Bandegi
- Laboratory of Endocrine Research, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Rashidy-Pour
- Laboratory of Learning and Memory, Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
21
|
Jonckheere J, Deloulme JC, Dall’Igna G, Chauliac N, Pelluet A, Nguon AS, Lentini C, Brocard J, Denarier E, Brugière S, Couté Y, Heinrich C, Porcher C, Holtzmann J, Andrieux A, Suaud-Chagny MF, Gory-Fauré S. Short- and long-term efficacy of electroconvulsive stimulation in animal models of depression: The essential role of neuronal survival. Brain Stimul 2018; 11:1336-1347. [DOI: 10.1016/j.brs.2018.08.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/12/2018] [Accepted: 08/03/2018] [Indexed: 12/28/2022] Open
|
22
|
Gong Q, Su YA, Wu C, Si TM, Deussing JM, Schmidt MV, Wang XD. Chronic Stress Reduces Nectin-1 mRNA Levels and Disrupts Dendritic Spine Plasticity in the Adult Mouse Perirhinal Cortex. Front Cell Neurosci 2018; 12:67. [PMID: 29593501 PMCID: PMC5859075 DOI: 10.3389/fncel.2018.00067] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 12/28/2022] Open
Abstract
In adulthood, chronic exposure to stressful experiences disrupts synaptic plasticity and cognitive function. Previous studies have shown that perirhinal cortex-dependent object recognition memory is impaired by chronic stress. However, the stress effects on molecular expression and structural plasticity in the perirhinal cortex remain unclear. In this study, we applied the chronic social defeat stress (CSDS) paradigm and measured the mRNA levels of nectin-1, nectin-3 and neurexin-1, three synaptic cell adhesion molecules (CAMs) implicated in the adverse stress effects, in the perirhinal cortex of wild-type (WT) and conditional forebrain corticotropin-releasing hormone receptor 1 conditional knockout (CRHR1-CKO) mice. Chronic stress reduced perirhinal nectin-1 mRNA levels in WT but not CRHR1-CKO mice. In conditional forebrain corticotropin-releasing hormone conditional overexpression (CRH-COE) mice, perirhinal nectin-1 mRNA levels were also reduced, indicating that chronic stress modulates nectin-1 expression through the CRH-CRHR1 system. Moreover, chronic stress altered dendritic spine morphology in the main apical dendrites and reduced spine density in the oblique apical dendrites of perirhinal layer V pyramidal neurons. Our data suggest that chronic stress disrupts cell adhesion and dendritic spine plasticity in perirhinal neurons, which may contribute to stress-induced impairments of perirhinal cortex-dependent memory.
Collapse
Affiliation(s)
- Qian Gong
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun-Ai Su
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Chen Wu
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Mei Si
- National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital/Institute of Mental Health, Beijing, China.,Key Laboratory of Mental Health, Ministry of Health, Peking University, Beijing, China
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry (MPG), Munich, Germany
| | - Mathias V Schmidt
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry (MPG), Munich, Germany
| | - Xiao-Dong Wang
- Department of Neurobiology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
23
|
Csabai D, Wiborg O, Czéh B. Reduced Synapse and Axon Numbers in the Prefrontal Cortex of Rats Subjected to a Chronic Stress Model for Depression. Front Cell Neurosci 2018; 12:24. [PMID: 29440995 PMCID: PMC5797661 DOI: 10.3389/fncel.2018.00024] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 01/16/2018] [Indexed: 12/28/2022] Open
Abstract
Stressful experiences can induce structural changes in neurons of the limbic system. These cellular changes contribute to the development of stress-induced psychopathologies like depressive disorders. In the prefrontal cortex of chronically stressed animals, reduced dendritic length and spine loss have been reported. This loss of dendritic material should consequently result in synapse loss as well, because of the reduced dendritic surface. But so far, no one studied synapse numbers in the prefrontal cortex of chronically stressed animals. Here, we examined synaptic contacts in rats subjected to an animal model for depression, where animals are exposed to a chronic stress protocol. Our hypothesis was that long term stress should reduce the number of axo-spinous synapses in the medial prefrontal cortex. Adult male rats were exposed to daily stress for 9 weeks and afterward we did a post mortem quantitative electron microscopic analysis to quantify the number and morphology of synapses in the infralimbic cortex. We analyzed asymmetric (Type I) and symmetric (Type II) synapses in all cortical layers in control and stressed rats. We also quantified axon numbers and measured the volume of the infralimbic cortex. In our systematic unbiased analysis, we examined 21,000 axon terminals in total. We found the following numbers in the infralimbic cortex of control rats: 1.15 × 109 asymmetric synapses, 1.06 × 108 symmetric synapses and 1.00 × 108 myelinated axons. The density of asymmetric synapses was 5.5/μm3 and the density of symmetric synapses was 0.5/μm3. Average synapse membrane length was 207 nm and the average axon terminal membrane length was 489 nm. Stress reduced the number of synapses and myelinated axons in the deeper cortical layers, while synapse membrane lengths were increased. These stress-induced ultrastructural changes indicate that neurons of the infralimbic cortex have reduced cortical network connectivity. Such reduced network connectivity is likely to form the anatomical basis for the impaired functioning of this brain area. Indeed, impaired functioning of the prefrontal cortex, such as cognitive deficits are common in stressed individuals as well as in depressed patients.
Collapse
Affiliation(s)
- Dávid Csabai
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Ove Wiborg
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Boldizsár Czéh
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary.,Institute of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
24
|
Fluoxetine, not donepezil, reverses anhedonia, cognitive dysfunctions and hippocampal proteome changes during repeated social defeat exposure. Eur Neuropsychopharmacol 2018; 28:195-210. [PMID: 29174946 DOI: 10.1016/j.euroneuro.2017.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 09/17/2017] [Accepted: 11/03/2017] [Indexed: 12/11/2022]
Abstract
While anhedonia is considered a core symptom of major depressive disorder (MDD), less attention has been paid to cognitive dysfunctions. We evaluated the behavioural and molecular effects of a selective serotonin re-uptake inhibitor (SSRI, fluoxetine) and an acetylcholinesterase inhibitor (AChEI, donepezil) on emotional-cognitive endophenotypes of depression and the hippocampal proteome. A chronic social defeat (SD) procedure was followed up by "reminder" sessions of direct and indirect SD. Anhedonia-related behaviour was assessed longitudinally by intracranial self-stimulation (ICSS). Cognitive dysfunction was analysed by an object recognition test (ORT) and extinction of fear memory. Tandem mass spectrometry (MSE) and protein-protein-interaction (PPI) network modelling were used to characterise the underlying biological processes of SD and SSRI/AChEI treatment. Independent selected reaction monitoring (SRM) was conducted for molecular validation. Repeated SD resulted in a stable increase of anhedonia-like behaviour as measured by ICSS. Fluoxetine treatment reversed this phenotype, whereas donepezil showed no effect. Fluoxetine improved recognition memory and inhibitory learning in a stressor-related context, whereas donepezil only improved fear extinction. MSE and PPI network analysis highlighted functional SD stress-related hippocampal proteome changes including reduced glutamatergic neurotransmission and learning processes, which were reversed by fluoxetine, but not by donepezil. SRM validation of molecular key players involved in these pathways confirmed the hypothesis that fluoxetine acts via increased AMPA receptor signalling and Ca2+-mediated neuroplasticity in the amelioration of stress-impaired reward processing and memory consolidation. Our study highlights molecular mediators of SD stress reversed by SSRI treatment, identifying potential viable future targets to improve cognitive dysfunctions in MDD patients.
Collapse
|
25
|
The Effect of Glucocorticoid and Glucocorticoid Receptor Interactions on Brain, Spinal Cord, and Glial Cell Plasticity. Neural Plast 2017; 2017:8640970. [PMID: 28928988 PMCID: PMC5591892 DOI: 10.1155/2017/8640970] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/14/2017] [Indexed: 12/15/2022] Open
Abstract
Stress, injury, and disease trigger glucocorticoid (GC) elevation. Elevated GCs bind to the ubiquitously expressed glucocorticoid receptor (GR). While GRs are in every cell in the nervous system, the expression level varies, suggesting that diverse cell types react differently to GR activation. Stress/GCs induce structural plasticity in neurons, Schwann cells, microglia, oligodendrocytes, and astrocytes as well as affect neurotransmission by changing the release and reuptake of glutamate. While general nervous system plasticity is essential for adaptation and learning and memory, stress-induced plasticity is often maladaptive and contributes to neuropsychiatric disorders and neuropathic pain. In this brief review, we describe the evidence that stress/GCs activate GR to promote cell type-specific changes in cellular plasticity throughout the nervous system.
Collapse
|
26
|
Pérez MÁ, Peñaloza-Sancho V, Ahumada J, Fuenzalida M, Dagnino-Subiabre A. n-3 Polyunsaturated fatty acid supplementation restored impaired memory and GABAergic synaptic efficacy in the hippocampus of stressed rats. Nutr Neurosci 2017; 21:556-569. [DOI: 10.1080/1028415x.2017.1323609] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Miguel Ángel Pérez
- Laboratory of Stress Neurobiology, Center for Neurobiology and Brain Plasticity, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
| | - Valentín Peñaloza-Sancho
- Laboratory of Stress Neurobiology, Center for Neurobiology and Brain Plasticity, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
| | - Juan Ahumada
- Laboratory of Neural Plasticity, Center for Neurobiology and Brain Plasticity, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
| | - Marco Fuenzalida
- Laboratory of Neural Plasticity, Center for Neurobiology and Brain Plasticity, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
| | - Alexies Dagnino-Subiabre
- Laboratory of Stress Neurobiology, Center for Neurobiology and Brain Plasticity, Faculty of Sciences, Institute of Physiology, Universidad de Valparaíso, Valparaíso, Chile
- Auditory and Cognition Center, AUCO, Santiago, Chile
| |
Collapse
|
27
|
Vose LR, Stanton PK. Synaptic Plasticity, Metaplasticity and Depression. Curr Neuropharmacol 2017; 15:71-86. [PMID: 26830964 PMCID: PMC5327460 DOI: 10.2174/1570159x14666160202121111] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 08/13/2015] [Accepted: 01/30/2016] [Indexed: 01/30/2023] Open
Abstract
The development of a persistent depressive affective state has for some time been thought to result from persistent alterations in neurotransmitter-mediated synaptic transmission. While the identity of those transmitters has changed over the years, the literature has lacked mechanistic connections between the neurophysiological mechanisms they regulate, and how these mechanisms alter neuronal function, and, hence, affective homeostasis. This review will examine recent work that suggests that both long-term activity-dependent changes in synaptic strength (“plasticity”), and shifting set points for the ease of induction of future long-term changes (“metaplasticity”), may be critical to establishing and reversing a depressive behavioral state. Activity-dependent long-term synaptic plasticity involves both strengthening and weakening of synaptic connections associated with a dizzying array of neurochemical alterations that include synaptic insertion and removal of a number of subtypes of AMPA, NMDA and metabotropic glutamate receptors, changes in presynaptic glutamate release, and structural changes in dendritic spines. Cellular mechanisms of metaplasticity are far less well understood. Here, we will review the growing evidence that long-term synaptic changes in glutamatergic transmission, in brain regions that regulate mood, are key determinants of affective homeostasis and therapeutic targets with immense potential for drug development.
Collapse
Affiliation(s)
| | - Patric K Stanton
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, 10595, USA
| |
Collapse
|
28
|
Malan-Müller S, Hemmings S. The Big Role of Small RNAs in Anxiety and Stress-Related Disorders. ANXIETY 2017; 103:85-129. [DOI: 10.1016/bs.vh.2016.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
29
|
Ammassari-Teule M. Is structural remodeling in regions governing memory an univocal correlate of memory? Neurobiol Learn Mem 2016; 136:28-33. [DOI: 10.1016/j.nlm.2016.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 09/12/2016] [Accepted: 09/14/2016] [Indexed: 12/16/2022]
|
30
|
Kezhu W, Pan X, Cong L, Liming D, Beiyue Z, Jingwei L, Yanyan Y, Xinmin L. Effects of Ginsenoside Rg1 on Learning and Memory in a Reward-directed Instrumental Conditioning Task in Chronic Restraint Stressed Rats. Phytother Res 2016; 31:81-89. [DOI: 10.1002/ptr.5733] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/07/2016] [Accepted: 09/14/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Wang Kezhu
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Xu Pan
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Lu Cong
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Dong Liming
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Zhang Beiyue
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
- Beijing University of Chinese Medicine; Chaoyang District, Beijing 100029 China
| | - Lu Jingwei
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | - Yang Yanyan
- China Astronauts Research and Training Center; Beijing 100094 China
| | - Liu Xinmin
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| |
Collapse
|
31
|
Csabai D, Seress L, Varga Z, Ábrahám H, Miseta A, Wiborg O, Czéh B. Electron Microscopic Analysis of Hippocampal Axo-Somatic Synapses in a Chronic Stress Model for Depression. Hippocampus 2016; 27:17-27. [PMID: 27571571 PMCID: PMC5215622 DOI: 10.1002/hipo.22650] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2016] [Indexed: 01/01/2023]
Abstract
Stress can alter the number and morphology of excitatory synapses in the hippocampus, but nothing is known about the effect of stress on inhibitory synapses. Here, we used an animal model for depression, the chronic mild stress model, and quantified the number of perisomatic inhibitory neurons and their synapses. We found reduced density of parvalbumin‐positive (PV+) neurons in response to stress, while the density of cholecystokinin‐immunoreactive (CCK+) neurons was unaffected. We did a detailed electron microscopic analysis to quantify the frequency and morphology of perisomatic inhibitory synapses in the hippocampal CA1 area. We analyzed 1100 CA1 pyramidal neurons and 4800 perisomatic terminals in five control and four chronically stressed rats. In the control animals we observed the following parameters: Number of terminals/soma = 57; Number of terminals/100 µm cell perimeter = 10; Synapse/terminal ratio = 32%; Synapse number/100 terminal = 120; Average terminal length = 920nm. None of these parameters were affected by the stress exposure. Overall, these data indicate that despite the depressive‐like behavior and the decrease in the number of perisomatic PV+ neurons in the light microscopic preparations, the number of perisomatic inhibitory synapses on CA1 pyramidal cells was not affected by stress. In the electron microscope, PV+ neurons and the axon terminals appeared to be normal and we did not find any apoptotic or necrotic cells. This data is in sharp contrast to the remarkable remodeling of the excitatory synapses on spines that has been reported in response to stress and depressive‐like behavior. © 2016 The Authors Hippocampus Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dávid Csabai
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, Pécs, 7624, Hungary
| | - László Seress
- Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, 7624, Hungary
| | - Zsófia Varga
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, Pécs, 7624, Hungary
| | - Hajnalka Ábrahám
- Central Electron Microscope Laboratory, University of Pécs, Medical School, Pécs, 7624, Hungary.,Department of Medical Biology, University of Pécs, Medical School, Pécs, 7624, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine, University of Pécs, Medical School, Pécs, 7624, Hungary
| | - Ove Wiborg
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| | - Boldizsár Czéh
- MTA - PTE, Neurobiology of Stress Research Group, Szentágothai Research Center, Pécs, 7624, Hungary.,Department of Laboratory Medicine, University of Pécs, Medical School, Pécs, 7624, Hungary.,Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Risskov, Denmark
| |
Collapse
|
32
|
Abstract
OBJECTIVE Among the 28 metalloproteinases described so far, 23 can be found in the human organism, but only few are expressed in the human brain. The main objective of this study was to analyse the relationship between MMP-2, MMP-9 and TIMP-2 gene expression and cognitive performance. METHODS The study comprised 234 subjects: patients suffering from recurrent depressive disorder (rDD, n=139) and healthy subjects (HS, n=95). The cognitive function assessment was carried out with the help of the following tests: Trail Making Test, The Stroop Test, Verbal Fluency Test and Auditory Verbal Learning Test. Gene expression on the mRNA and protein level was evaluated for MMP-2, MMP-9 and TIMP-2 in both groups using RNA extraction, reverse transcription and enzyme-linked immunosorbent assay. RESULTS Both mRNA and protein expression levels of all the genes were significantly lower in rDD subjects as compared with HS. Having analysed the entire experimental group (N=234), significant interrelations were found between the expression of the analysed genes and the results of the tests used to measure cognitive functions. Increased expression on both the mRNA and the protein level was associated in each case with better performance of all the tests conducted. After carrying out a separate analysis on the people from the rDD group and the HS group, similar dependencies were still observed. CONCLUSIONS The results of our study show decreased expression of MMP-2, MMP-9 and TIMP-2 genes on both mRNA and protein levels in depression. Elevated expression of MMP-2, MMP-9, TIMP-2 positively affects cognitive efficiency: working memory, executive functions, attention functions, direct and delayed auditory-verbal memory, the effectiveness of learning processes and verbal fluency. The study highlights the important role of peripheral matrix metalloproteinases genes in depression and cognitive functions.
Collapse
|
33
|
Milior G, Lecours C, Samson L, Bisht K, Poggini S, Pagani F, Deflorio C, Lauro C, Alboni S, Limatola C, Branchi I, Tremblay ME, Maggi L. Fractalkine receptor deficiency impairs microglial and neuronal responsiveness to chronic stress. Brain Behav Immun 2016; 55:114-125. [PMID: 26231972 DOI: 10.1016/j.bbi.2015.07.024] [Citation(s) in RCA: 180] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/25/2015] [Accepted: 07/26/2015] [Indexed: 12/25/2022] Open
Abstract
Chronic stress is one of the most relevant triggering factors for major depression. Microglial cells are highly sensitive to stress and, more generally, to environmental challenges. However, the role of these brain immune cells in mediating the effects of stress is still unclear. Fractalkine signaling - which comprises the chemokine CX3CL1, mainly expressed by neurons, and its receptor CX3CR1, almost exclusively present on microglia in the healthy brain - has been reported to critically regulate microglial activity. Here, we investigated whether interfering with microglial function by deleting the Cx3cr1 gene affects the brain's response to chronic stress. To this purpose, we housed Cx3cr1 knockout and wild-type adult mice in either control or stressful environments for 2weeks, and investigated the consequences on microglial phenotype and interactions with synapses, synaptic transmission, behavioral response and corticosterone levels. Our results show that hampering neuron-microglia communication via the CX3CR1-CX3CL1 pathway prevents the effects of chronic unpredictable stress on microglial function, short- and long-term neuronal plasticity and depressive-like behavior. Overall, the present findings suggest that microglia-regulated mechanisms may underlie the differential susceptibility to stress and consequently the vulnerability to diseases triggered by the experience of stressful events, such as major depression.
Collapse
Affiliation(s)
- Giampaolo Milior
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | - Cynthia Lecours
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Louis Samson
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Kanchan Bisht
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada
| | - Silvia Poggini
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Francesca Pagani
- Center for Life Nanoscience, Istituto Italiano di Tecnologia@Sapienza, Rome, Italy
| | - Cristina Deflorio
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy; Département de Neuroscience, Institut Pasteur, Unité Neurobiologie Intégrative des Systèmes Cholinergiques, Paris Cedex 15, Paris, France
| | - Clotilde Lauro
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | - Silvia Alboni
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy; IRCCS Neuromed, Pozzilli, IS, Italy
| | - Igor Branchi
- Section of Behavioural Neurosciences, Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, 2705, boulevard Laurier, Québec, Canada.
| | - Laura Maggi
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| |
Collapse
|
34
|
Zhu X, Gao R, Liu Z, Cheng Z, Qi Y, Fan C, Yu SY. Ginsenoside Rg1 reverses stress-induced depression-like behaviours and brain-derived neurotrophic factor expression within the prefrontal cortex. Eur J Neurosci 2016; 44:1878-85. [DOI: 10.1111/ejn.13255] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 02/17/2016] [Accepted: 04/05/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Xiuzhi Zhu
- Department of Physiology; School of Medicine; Shandong University; Wenhuaxilu Road Jinan Shandong Province 250012 China
| | - Rui Gao
- Department of Physiology; School of Medicine; Shandong University; Wenhuaxilu Road Jinan Shandong Province 250012 China
- Department of Microorganism; Jinan Nursing Vocational College; Jinan Shandong Province China
| | - Zhuxi Liu
- Department of Physiology; School of Medicine; Shandong University; Wenhuaxilu Road Jinan Shandong Province 250012 China
| | - Ziyi Cheng
- Department of Physiology; School of Medicine; Shandong University; Wenhuaxilu Road Jinan Shandong Province 250012 China
| | - Yihang Qi
- Department of Physiology; School of Medicine; Shandong University; Wenhuaxilu Road Jinan Shandong Province 250012 China
| | - Cuiqin Fan
- Department of Physiology; School of Medicine; Shandong University; Wenhuaxilu Road Jinan Shandong Province 250012 China
| | - Shu Yan Yu
- Department of Physiology; School of Medicine; Shandong University; Wenhuaxilu Road Jinan Shandong Province 250012 China
- Shandong Provincial Key Laboratory of Mental Disorders; School of Medicine; Jinan Shandong Province China
| |
Collapse
|
35
|
van der Kooij MA, Masana M, Rust MB, Müller MB. The stressed cytoskeleton: How actin dynamics can shape stress-related consequences on synaptic plasticity and complex behavior. Neurosci Biobehav Rev 2016; 62:69-75. [DOI: 10.1016/j.neubiorev.2015.12.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 12/01/2015] [Accepted: 12/07/2015] [Indexed: 01/06/2023]
|
36
|
Dendritic Spines in Depression: What We Learned from Animal Models. Neural Plast 2016; 2016:8056370. [PMID: 26881133 PMCID: PMC4736982 DOI: 10.1155/2016/8056370] [Citation(s) in RCA: 301] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 11/26/2015] [Indexed: 02/07/2023] Open
Abstract
Depression, a severe psychiatric disorder, has been studied for decades, but the underlying mechanisms still remain largely unknown. Depression is closely associated with alterations in dendritic spine morphology and spine density. Therefore, understanding dendritic spines is vital for uncovering the mechanisms underlying depression. Several chronic stress models, including chronic restraint stress (CRS), chronic unpredictable mild stress (CUMS), and chronic social defeat stress (CSDS), have been used to recapitulate depression-like behaviors in rodents and study the underlying mechanisms. In comparison with CRS, CUMS overcomes the stress habituation and has been widely used to model depression-like behaviors. CSDS is one of the most frequently used models for depression, but it is limited to the study of male mice. Generally, chronic stress causes dendritic atrophy and spine loss in the neurons of the hippocampus and prefrontal cortex. Meanwhile, neurons of the amygdala and nucleus accumbens exhibit an increase in spine density. These alterations induced by chronic stress are often accompanied by depression-like behaviors. However, the underlying mechanisms are poorly understood. This review summarizes our current understanding of the chronic stress-induced remodeling of dendritic spines in the hippocampus, prefrontal cortex, orbitofrontal cortex, amygdala, and nucleus accumbens and also discusses the putative underlying mechanisms.
Collapse
|
37
|
Glasper ER, Hyer MM, Katakam J, Harper R, Ameri C, Wolz T. Fatherhood contributes to increased hippocampal spine density and anxiety regulation in California mice. Brain Behav 2016; 6:e00416. [PMID: 27110439 PMCID: PMC4834941 DOI: 10.1002/brb3.416] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/07/2015] [Accepted: 10/15/2015] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Parenting alters the hippocampus, an area of the brain that undergoes significant experience-induced plasticity and contributes to emotional regulation. While the relationship between maternal care and hippocampal neuroplasticity has been characterized, the extent to which fatherhood alters the structure and function of the hippocampus is far less understood. METHODS Here, we investigated to what extent fatherhood altered anxiety regulation and dendritic morphology of the hippocampus using the highly paternal California mouse (Peromyscus californicus). RESULTS Fathers spent significantly more time on the open arms of the elevated plus maze, compared to non-fathers. Total distance traveled in the EPM was not changed by paternal experience, which suggests that the increased time spent on the open arms of the maze indicates decreased anxiety-like behavior. Fatherhood also increased dendritic spine density of granule cells in the dentate gyrus and basal dendrites of pyramidal cells in area CA1 of the hippocampus. CONCLUSIONS These findings parallel those observed in maternal rodents, suggesting that the hippocampus of fathers and mothers respond similarly to offspring.
Collapse
Affiliation(s)
- Erica R Glasper
- Department of Psychology University of Maryland College Park Maryland 20742; Program in Neuroscience and Cognitive Science University of Maryland College Park Maryland 20742
| | - Molly M Hyer
- Program in Neuroscience and Cognitive Science University of Maryland College Park Maryland 20742
| | - Jhansi Katakam
- Department of Psychology University of Maryland College Park Maryland 20742
| | - Robyn Harper
- Department of Psychology University of Maryland College Park Maryland 20742
| | - Cyrus Ameri
- Department of Psychology University of Maryland College Park Maryland 20742
| | - Thomas Wolz
- Department of Psychology University of Maryland College Park Maryland 20742
| |
Collapse
|
38
|
Zhang G, Chen L, Yang L, Hua X, Zhou B, Miao Z, Li J, Hu H, Namaka M, Kong J, Xu X. Combined use of spatial restraint stress and middle cerebral artery occlusion is a novel model of post-stroke depression in mice. Sci Rep 2015; 5:16751. [PMID: 26572587 PMCID: PMC4648085 DOI: 10.1038/srep16751] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/19/2015] [Indexed: 12/22/2022] Open
Abstract
Post stroke depression (PSD) is one of the most common complications of ischemic stroke. At present, the underlying mechanisms are unclear, largely because there are no reliable, valid and reproducible animal models of PSD. Here we report a novel animal model of PSD that displays consistent and reliable clinical features of hemiplegic stroke. The animal model encompasses a combination of the middle cerebral artery occlusion (MCAO) and spatial restraint stress. We found that a 60-minute MCAO followed by spatial restraint stress for 2 h daily for 2 to 4 weeks from the fourth day after MCAO induced PSD-like depressive phenotypes in mice. Importantly, the mice showed exacerbated deficits of neurological functions and decreased body weights, which were accompanied with reduced levels of brain derived neurotrophic factor and neurotransmitters including serotonin and dopamine. In addition, we identified increased levels of serum cortisol in our PSD mice. Finally, we found that mice with PSD were responsive to the tri-cyclic antidepressant imipramine as evidenced by their attenuated depressive behaviors, increased body weights, recovered brain serotonin levels, and decreased serum cortisol levels. This mouse model replicates multiple features of human post-stroke depression and thus provides a new model for the investigation of PSD.
Collapse
Affiliation(s)
- Gaocai Zhang
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China.,Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Li Chen
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China
| | - Lingli Yang
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China
| | - Xiaodong Hua
- Department of Biochemistry, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Beiqun Zhou
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China.,Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou City, China
| | - Jizhen Li
- Department of Neurology, Suzhou Kowloon Hospital, 118 Wansheng Street, Suzhou City, China
| | - Hua Hu
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China
| | - Michael Namaka
- College of Pharmacy and Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Xingshun Xu
- Department of Neurology, the Second Affiliated Hospital of Soochow University, Suzhou City, China.,Institute of Neuroscience, Soochow University, Suzhou City, China
| |
Collapse
|
39
|
Li XL, Yuan YG, Xu H, Wu D, Gong WG, Geng LY, Wu FF, Tang H, Xu L, Zhang ZJ. Changed Synaptic Plasticity in Neural Circuits of Depressive-Like and Escitalopram-Treated Rats. Int J Neuropsychopharmacol 2015; 18:pyv046. [PMID: 25899067 PMCID: PMC4648155 DOI: 10.1093/ijnp/pyv046] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Although progress has been made in the detection and characterization of neural plasticity in depression, it has not been fully understood in individual synaptic changes in the neural circuits under chronic stress and antidepressant treatment. METHODS Using electron microscopy and Western-blot analyses, the present study quantitatively examined the changes in the Gray's Type I synaptic ultrastructures and the expression of synapse-associated proteins in the key brain regions of rats' depressive-related neural circuit after chronic unpredicted mild stress and/or escitalopram administration. Meanwhile, their depressive behaviors were also determined by several tests. RESULTS The Type I synapses underwent considerable remodeling after chronic unpredicted mild stress, which resulted in the changed width of the synaptic cleft, length of the active zone, postsynaptic density thickness, and/or synaptic curvature in the subregions of medial prefrontal cortex and hippocampus, as well as the basolateral amygdaloid nucleus of the amygdala, accompanied by changed expression of several synapse-associated proteins. Chronic escitalopram administration significantly changed the above alternations in the chronic unpredicted mild stress rats but had little effect on normal controls. Also, there was a positive correlation between the locomotor activity and the maximal synaptic postsynaptic density thickness in the stratum radiatum of the Cornu Ammonis 1 region and a negative correlation between the sucrose preference and the length of the active zone in the basolateral amygdaloid nucleus region in chronic unpredicted mild stress rats. CONCLUSION These findings strongly indicate that chronic stress and escitalopram can alter synaptic plasticity in the neural circuits, and the remodeled synaptic ultrastructure was correlated with the rats' depressive behaviors, suggesting a therapeutic target for further exploration.
Collapse
Affiliation(s)
- Xiao-Li Li
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Yong-Gui Yuan
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Hua Xu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Di Wu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Wei-Gang Gong
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Lei-Yu Geng
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Fang-Fang Wu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Hao Tang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Lin Xu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work
| | - Zhi-Jun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute and Medical School of Southeast University, Nanjing, Jiangsu, China (Drs Li, Yuan, H. Xu, D. Wu, Gong, Geng, F-F. Wu, Tang, and Zhang); Key Laboratory of Animal Models and Human Disease Mechanisms, Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China (Dr L. Xu); Graduate School of Chinese Academy of Sciences, Beijing, China (Dr L. Xu)X-L.L. and Y-G.Y. contributed equally to this work.
| |
Collapse
|
40
|
Flupirtine attenuates chronic restraint stress-induced cognitive deficits and hippocampal apoptosis in male mice. Behav Brain Res 2015; 288:1-10. [PMID: 25869780 DOI: 10.1016/j.bbr.2015.04.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 04/01/2015] [Accepted: 04/04/2015] [Indexed: 02/07/2023]
Abstract
Chronic restraint stress (CRS) causes hippocampal neurodegeneration and hippocampus-dependent cognitive deficits. Flupirtine represents neuroprotective effects and we have previously shown that flupirtine can protect against memory impairment induced by acute stress. The present study aimed to investigate whether flupirtine could alleviate spatial learning and memory impairment and hippocampal apoptosis induced by CRS. CRS mice were restrained in well-ventilated Plexiglass tubes for 6h daily beginning from 10:00 to 16:00 for 21 consecutive days. Mice were injected with flupirtine (10mg/kg and 25mg/kg) or vehicle (10% DMSO) 30min before restraint stress for 21 days. After stressor cessation, the spatial learning and memory, dendritic spine density, injured neurons and the levels of Bcl-2, Bax, p-Akt, p-GSK-3β, p-Erk1/2 and synaptophysin of hippocampal tissues were examined. Our results showed that flupirtine significantly prevented spatial learning and memory impairment induced by CRS in the Morris water maze. In addition, flupirtine (10mg/kg and 25mg/kg) treatment alleviated neuronal apoptosis and the reduction of dendritic spine density and synaptophysin expression in the hippocampal CA1 region of CRS mice. Furthermore, flupirtine (10mg/kg and 25mg/kg) treatment significantly decreased the expression of Bax and increased the p-Akt and p-GSK-3β, and flupirtine (25mg/kg) treatment up-regulated the p-Erk1/2 in the hippocampus of CRS mice. These results suggested that flupirtine exerted protective effects on the CRS-induced cognitive impairment and hippocampal neuronal apoptosis, which is possibly associated with the activation of Akt/GSK-3β and Erk1/2 signaling pathways.
Collapse
|
41
|
Pietranera L, Brocca ME, Roig P, Lima A, Garcia-Segura LM, De Nicola AF. Estrogens are neuroprotective factors for hypertensive encephalopathy. J Steroid Biochem Mol Biol 2015; 146:15-25. [PMID: 24736028 DOI: 10.1016/j.jsbmb.2014.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 10/25/2022]
Abstract
Estrogens are neuroprotective factors for brain diseases, including hypertensive encephalopathy. In particular, the hippocampus is highly damaged by high blood pressure, with several hippocampus functions being altered in humans and animal models of hypertension. Working with a genetic model of primary hypertension, the spontaneously hypertensive rat (SHR), we have shown that SHR present decreased dentate gyrus neurogenesis, astrogliosis, low expression of brain derived neurotrophic factor (BDNF), decreased number of neurons in the hilus of the dentate gyrus, increased basal levels of the estrogen-synthesizing enzyme aromatase, and atrophic dendritic arbor with low spine density in the CA1 region compared to normotensive Wistar Kyoto (WKY) ratsl. Changes also occur in the hypothalamus of SHR, with increased expression of the hypertensinogenic peptide arginine vasopressin (AVP) and its V1b receptor. Following chronic estradiol treatment, SHR show decreased blood pressure, enhanced hippocampus neurogenesis, decreased the reactive astrogliosis, increased BDNF mRNA and protein expression in the dentate gyrus, increased neuronal number in the hilus of the dentate gyrus, further increased the hyperexpression of aromatase and replaced spine number with remodeling of the dendritic arbor of the CA1 region. We have detected by qPCR the estradiol receptors ERα and ERβ in hippocampus from both SHR and WKY rats, suggesting direct effects of estradiol on brain cells. We hypothesize that a combination of exogenously given estrogens plus those locally synthesized by estradiol-stimulated aromatase may better alleviate the hippocampal and hypothalamic encephalopathy of SHR. This article is part of a Special Issue entitled "Sex steroids and brain disorders".
Collapse
Affiliation(s)
- Luciana Pietranera
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina; Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina
| | - Maria Elvira Brocca
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Paulina Roig
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Analia Lima
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina
| | - Luis Miguel Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Avenida Doctor Arce 37, E-28002 Madrid, Spain
| | - Alejandro F De Nicola
- Laboratory of Neuroendocrine Biochemistry, Instituto de Biología y Medicina Experimental, Obligado 2490, 1428 Buenos Aires, Argentina; Department of Human Biochemistry, Faculty of Medicine, University of Buenos Aires, Paraguay 2155, 1425 Buenos Aires, Argentina.
| |
Collapse
|
42
|
Role for NUP62 depletion and PYK2 redistribution in dendritic retraction resulting from chronic stress. Proc Natl Acad Sci U S A 2014; 111:16130-5. [PMID: 25349423 DOI: 10.1073/pnas.1418896111] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Genetic evidence suggests cell-type-specific functions for certain nucleoporins, and gene expression profiling has revealed that nucleoporin p62 (NUP62) transcripts are decreased in the prefrontal cortex of major depressives. Chronic stress, which can precipitate depression, induces changes in the architecture and plasticity of apical dendrites that are particularly evident in the CA3 region of the hippocampus. Genetically targeted translating ribosome affinity purification revealed a selective reduction in translated Nup62 transcripts in CA3 of chronically stressed mice, and the Nup62 protein content of nuclei extracted from whole hippocampus was found to be decreased in chronically stressed rats. In cultured cells, phosphorylation of a FAK/proline-rich tyrosine kinase 2 (PYK2) consensus site in the alpha-helical domain of NUP62 (human Y422) is shown to be associated with shedding of NUP62 from the nuclear pore complex (NPC) and/or retention of NUP62 in the cytoplasm. Increased levels of phospho-Y425 Nup62 were observed in cytoplasmic fractions of hippocampi from chronically stressed rats, and immunofluorescence microscopy revealed redistribution of activated Pyk2 to the perinuclear region of stressed pyramidal neurons. Depletion of Nup62 from cultured embryonic day 18 rat hippocampal and cortical neurons resulted in simplification and retraction of dendritic arbors, without disruption of axon initial segment integrity. Thus, at least two types of mechanisms--one affecting expression and the other association with the NPC--could contribute to loss of NUP62 from CA3 pyramidal neurons during chronic stress. Their combined actions may account for the enhanced responsiveness of CA3 apical dendrites to chronic stress and may either be pathogenic or serve to protect CA3 neurons from permanent damage.
Collapse
|
43
|
van der Kooij MA, Fantin M, Rejmak E, Grosse J, Zanoletti O, Fournier C, Ganguly K, Kalita K, Kaczmarek L, Sandi C. Role for MMP-9 in stress-induced downregulation of nectin-3 in hippocampal CA1 and associated behavioural alterations. Nat Commun 2014; 5:4995. [PMID: 25232752 PMCID: PMC4199199 DOI: 10.1038/ncomms5995] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Accepted: 08/15/2014] [Indexed: 01/08/2023] Open
Abstract
Chronic stress is a risk factor for the development of psychopathologies characterized by cognitive dysfunction and deregulated social behaviours. Emerging evidence suggests a role for cell adhesion molecules, including nectin-3, in the mechanisms that underlie the behavioural effects of stress. We tested the hypothesis that proteolytic processing of nectins by matrix metalloproteinases (MMPs), an enzyme family that degrades numerous substrates, including cell adhesion molecules, is involved in hippocampal effects induced by chronic restraint stress. A reduction in nectin-3 in the perisynaptic CA1, but not in the CA3, compartment is observed following chronic stress and is implicated in the effects of stress in social exploration, social recognition and a CA1-dependent cognitive task. Increased MMP-9-related gelatinase activity, involving N-methyl-D-aspartate receptor, is specifically found in the CA1 and involved in nectin-3 cleavage and chronic stress-induced social and cognitive alterations. Thus, MMP-9 proteolytic processing emerges as an important mediator of stress effects in brain function and behaviour.
Collapse
Affiliation(s)
- Michael A. van der Kooij
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne 1015, Switzerland
| | - Martina Fantin
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne 1015, Switzerland
| | - Emilia Rejmak
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street 02-093 Warsaw, Poland
| | - Jocelyn Grosse
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne 1015, Switzerland
| | - Olivia Zanoletti
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne 1015, Switzerland
| | - Celine Fournier
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne 1015, Switzerland
| | - Krishnendu Ganguly
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street 02-093 Warsaw, Poland
| | - Katarzyna Kalita
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street 02-093 Warsaw, Poland
| | - Leszek Kaczmarek
- Laboratory of Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street 02-093 Warsaw, Poland
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, EPFL, Lausanne 1015, Switzerland
| |
Collapse
|
44
|
Zalosnik MI, Pollano A, Trujillo V, Suárez MM, Durando PE. Effect of maternal separation and chronic stress on hippocampal-dependent memory in young adult rats: evidence for the match-mismatch hypothesis. Stress 2014; 17:445-50. [PMID: 24930801 DOI: 10.3109/10253890.2014.936005] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adverse experiences early in life may sensitize the hippocampus to subsequent stressors throughout the individual's life. We analyzed in male rats, whether, the interaction between early maternal separation and chronic stress affects: (1) the volume of the dorsal hippocampus, (2) CA1, CA2/3 and dentate gyrus (DG) and (3) hippocampal-dependent memory in adulthood. Male Wistar rats were subjected to daily maternal separation for 4.5 h between postnatal days 1-21. From postnatal day 50, animals were exposed to a chronic unpredictable stress paradigm during 24 days. The volumes of the dorsal hippocampus, their areas or strata did not reveal significant differences between treatments. Non-maternally separated and stressed animals showed poor hippocampal performance in a contextual fear conditioning test, with a significant reduction in freezing behavior during post-conditioning compared with control and maternally separated and stressed animals. Also, memory retrieval 24 h after conditioning was significantly weaker in this group than in control animals. Memory performance in maternally separated and stressed rats was similar to control animals. Our results show an interaction between early environment experiences and chronic variable stress in young adulthood as evidence that early stressful experiences do not necessarily lead to a negative outcome but can help in maintaining brain plasticity and increase fitness when animals reach adulthood.
Collapse
Affiliation(s)
- M I Zalosnik
- Laboratorio de Fisiología Animal, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba , Córdoba , Argentina
| | | | | | | | | |
Collapse
|
45
|
Qiao H, An SC, Ren W, Ma XM. Progressive alterations of hippocampal CA3-CA1 synapses in an animal model of depression. Behav Brain Res 2014; 275:191-200. [PMID: 25192638 DOI: 10.1016/j.bbr.2014.08.040] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 08/08/2014] [Accepted: 08/20/2014] [Indexed: 12/11/2022]
Abstract
Major depressive disorder is the most prevalent psychiatric condition, but the cellular and molecular mechanisms underlying this disorder are largely unknown, although multiple hypotheses have been proposed. The aim of this study was to characterize the progressive alteration of neuronal plasticity in the male rat hippocampus during depression induced by chronic unpredictable mild stress (CUMS), an established animal model of depression. The data in the hippocampus were collected on days 7, 14 and 21 after the onset of three-week CUMS. When analyzed on day 21, three-week CUMS induced typically depressive-like behaviors, impaired LTP induction, and decreased basal synaptic transmission at hippocampal CA3-CA1 synapses recorded in vivo, which was accompanied by decreased density of dendritic spines in CA1 and CA3 pyramidal neurons. The levels of both Kalirin-7 and brain-derived neurotrophic factor (BDNF) in the hippocampus were decreased at the same time. On day 14 (middle phase), some depressive-like behaviors were observed, which was accompanied by depressed basal synaptic transmission and enhanced LTP induction at the CA3-CA1 synapses. However, BDNF expression was decreased without alteration of Kalirin7 expression in comparison with no-stress control. Depressed basal synaptic transmission occurred in the middle phase of CUMS may contribute to decreased expression of BDNF. On day 7, depressive-like behaviors were not observed, and LTP induction, spine density, Kalirin-7 and BDNF expression were not altered by CUMS in comparison with no-stress control. These results showed that the functional changes at CA3-CA1synapses occurred earlier than the structural alteration during three-week CUMS as a strategy of neural adaptation, and rats required three weeks to develop depressive-like behaviors during CUMS. Our results suggest an important role of Kalirin-7 in CUMS-mediated alterations in spine density, synaptic function and overall depressive-like behaviors on day 21.
Collapse
Affiliation(s)
- Hui Qiao
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, PR China
| | - Shu-Cheng An
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, PR China.
| | - Wei Ren
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, PR China
| | - Xin-Ming Ma
- College of Life Science, Shaanxi Normal University, Xi'an, Shaanxi Province 710062, PR China; University of Connecticut Health Center, Department of Neuroscience, Farmington, CT 06030, USA
| |
Collapse
|
46
|
Van Kempen TA, Gorecka J, Gonzalez AD, Soeda F, Milner TA, Waters EM. Characterization of neural estrogen signaling and neurotrophic changes in the accelerated ovarian failure mouse model of menopause. Endocrinology 2014; 155:3610-23. [PMID: 24926825 PMCID: PMC4138565 DOI: 10.1210/en.2014-1190] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Accelerated ovarian failure (AOF) can be induced in young mice with low doses of 4-vinylcyclohexene diepoxide (VCD), modeling the hormone changes observed across menopause. We assessed markers of synaptic plasticity in the hippocampus, anxiety-like behavior, and spatial learning longitudinally at 4 time points across the AOF model: premenopause, early perimenopause, late perimenopause, and postmenopause (POST). As others have shown, VCD administration decreased ovarian follicle counts and increased acyclicity as the model progressed to POST but with no impact on organ or body weights. The morphology of Iba1 immunoreactive microglia did not differ between vehicle- and VCD-administered mice. Hippocampal postsynaptic density 95 levels were minimally altered across the AOF model but decreased at POST in CA3b 24 hours after exogenous estradiol benzoate (EB). In contrast, hippocampal phosphorylated AKT levels transiently decreased in premenopause but increased at POST after 24 hours of EB in select subregions. Electron microscopy revealed fewer estrogen receptor α containing dendritic spines and terminals in CA1 stratum radiatum at POST. mRNA levels of most brain-derived neurotrophic factor exons (except V and VI) were lower in POST compared with ovariectomized mice. Exon V was sensitive to 24 hours of EB administration in POST-VCD. Anxiety-like behavior was unaffected at any menopause phase. Spatial learning was unaffected in all groups, but POST-VCD mice performed below chance. Our results suggest that the AOF model is suitable for longitudinal studies of neurobiological changes across the menopause transition in mice. Our findings also point to complex interactions between estrogen receptors and pathways involved in synaptic plasticity.
Collapse
Affiliation(s)
- Tracey A Van Kempen
- Brain and Mind Research Institute (T.A.V.K., T.A.M.) and Graduate Program in Neuroscience (T.A.V.K., A.D.G.), Weill Cornell Medical College, and Laboratory of Neuroendocrinology (J.G., T.A.M., E.M.W.), The Rockefeller University, New York, New York 10065; and Department of Environmental and Molecular Health Sciences (F.S.), Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Grizzell JA, Iarkov A, Holmes R, Mori T, Echeverria V. Cotinine reduces depressive-like behavior, working memory deficits, and synaptic loss associated with chronic stress in mice. Behav Brain Res 2014; 268:55-65. [DOI: 10.1016/j.bbr.2014.03.047] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/28/2014] [Accepted: 03/31/2014] [Indexed: 12/21/2022]
|
48
|
Borrie SC, Sartori SB, Lehmann J, Sah A, Singewald N, Bandtlow CE. Loss of Nogo receptor homolog NgR2 alters spine morphology of CA1 neurons and emotionality in adult mice. Front Behav Neurosci 2014; 8:175. [PMID: 24860456 PMCID: PMC4030173 DOI: 10.3389/fnbeh.2014.00175] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2014] [Accepted: 04/25/2014] [Indexed: 01/08/2023] Open
Abstract
Molecular mechanisms which stabilize dendrites and dendritic spines are essential for regulation of neuronal plasticity in development and adulthood. The class of Nogo receptor proteins, which are critical for restricting neurite outgrowth inhibition signaling, have been shown to have roles in developmental, experience and activity induced plasticity. Here we investigated the role of the Nogo receptor homolog NgR2 in structural plasticity in a transgenic null mutant for NgR2. Using Golgi-Cox staining to analyze morphology, we show that loss of NgR2 alters spine morphology in adult CA1 pyramidal neurons of the hippocampus, significantly increasing mushroom-type spines, without altering dendritic tree complexity. Furthermore, this shift is specific to apical dendrites in distal CA1 stratum radiatum (SR). Behavioral alterations in NgR2(-/-) mice were investigated using a battery of standardized tests and showed that whilst there were no alterations in learning and memory in NgR2(-/-) mice compared to littermate controls, NgR2(-/-) displayed reduced fear expression in the contextual conditioned fear test, and exhibited reduced anxiety- and depression-related behaviors. This suggests that the loss of NgR2 results in a specific phenotype of reduced emotionality. We conclude that NgR2 has role in maintenance of mature spines and may also regulate fear and anxiety-like behaviors.
Collapse
Affiliation(s)
- Sarah C Borrie
- Division of Neurobiochemistry, Biocenter, Innsbruck Medical University Innsbruck, Austria
| | - Simone B Sartori
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Centre for Molecular Biosciences Innsbruck, University of Innsbruck Innsbruck, Austria
| | - Julian Lehmann
- Division of Neurobiochemistry, Biocenter, Innsbruck Medical University Innsbruck, Austria
| | - Anupam Sah
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Centre for Molecular Biosciences Innsbruck, University of Innsbruck Innsbruck, Austria
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Centre for Molecular Biosciences Innsbruck, University of Innsbruck Innsbruck, Austria
| | - Christine E Bandtlow
- Division of Neurobiochemistry, Biocenter, Innsbruck Medical University Innsbruck, Austria
| |
Collapse
|
49
|
Effects of curcumin on chronic, unpredictable, mild, stress-induced depressive-like behaviour and structural plasticity in the lateral amygdala of rats. Int J Neuropsychopharmacol 2014; 17:793-806. [PMID: 24405689 DOI: 10.1017/s1461145713001661] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Depression is a neuropsychiatric disease associated with wide ranging disruptions in neuronal plasticity throughout the brain. Curcumin, a natural polyphenolic compound of curcuma loga, has been demonstrated to be effective in the treatment of depressive-like disorders. The present study aimed to investigate the mechanisms underlying the antidepressant-like effects of curcumin in a rat model of chronic, unpredictable, mild, stress (CUMS) -induced depression. The results showed that CUMS produced depressive-like behaviours in rats, which were associated with ultra-structural changes in neurons within the lateral amygdala (LA). In addition, the expression of synapse-associated proteins such as brain-derived neurotrophic factor (BDNF), PSD-95 and synaptophysin were significantly decreased in the LA of CUMS-treated rats. Chronic administration of curcumin (40 mg/kg, i.p. 6 wk) before stress exposure significantly prevented these neuronal and biochemical alterations induced by CUMS, and suppressed depressive-like behaviours, suggesting that this neuronal dysregulation may be related to the depressive-like behaviours caused by CUMS. Together with our previous results, the current findings demonstrate that curcumin exhibits neuroprotection and antidepressant-like effects in the CUMS-induced depression model. Furthermore, this antidepressant-like action of curcumin appears to be mediated by modulating synapse-associated proteins within the LA. These findings provide new insights into the underlying mechanisms leading to neural dysfunction in depression and reveal the therapeutic potential for curcumin use in clinical trials.
Collapse
|
50
|
Scollo A, Gottardo F, Contiero B, Edwards SA. Does stocking density modify affective state in pigs as assessed by cognitive bias, behavioural and physiological parameters? Appl Anim Behav Sci 2014. [DOI: 10.1016/j.applanim.2014.01.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|