1
|
Qu Y, Gu J, Li L, Yan Y, Yan C, Zhang T. Guilu Erxian Jiao remodels dendritic spine morphology through activation of the hippocampal TRPC6-CaMKIV-CREB signaling pathway and suppresses fear memory generalization in rats with post-traumatic stress disorder. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119252. [PMID: 39681200 DOI: 10.1016/j.jep.2024.119252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Guilu Erxian Jiao (GLEXJ) is a renowned traditional Chinese herbal formula used to tonify the kidney. It is employed to treat psychiatric disorders, and alleviate memory impairment, cognitive dysfunction, and behavioral disorders. Modern pharmacological studies have demonstrated GLEXJ's ability to significantly inhibit the fear response in post-traumatic stress disorder (PTSD) and facilitate the extinction of fear memory. However, the underlying pharmacological mechanisms remain elusive. AIM OF THE STUDY Fear memory generalization, a fundamental characteristic of PTSD, remains poorly understood, and optimal pharmacological treatments are lacking. This study aimed to investigate GLEXJ's inhibitory effects on fear memory generalization in PTSD rats and elucidate its underlying mechanisms. MATERIALS AND METHODS PTSD rats were induced using the single prolonged stress and electrical stimulation (SPS&S) protocol and treated with GLEXJ or paroxetine (PRX). Fear memory generalization was assessed using a contextual fear memory test. Hippocampal dendritic spine morphology was analyzed using Golgi-Cox staining. The chemical composition of GLEXJ was determined using ultra-high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS). Network pharmacology was employed to predict GLEXJ's therapeutic mechanism in PTSD treatment. Western blotting and immunofluorescence were used to measure indicators of the transient receptor potential channel 6 (TRPC6)-mediated calcium/calmodulin-dependent protein kinase IV-cAMP response element-binding protein (CaMKIV-CREB) signaling pathway. In vitro, TRPC6 was suppressed in rat adrenal pheochromocytoma (PC12) cells using lentiviral vectors, and phalloidin staining was employed to examine changes in Fibros actin (F-actin), elucidating the mechanistic effects of GLEXJ-containing serum. RESULTS GLEXJ significantly mitigated fear memory generalization in PTSD rats, even with repeated stress exposure. It also alleviated abnormal hippocampal dendritic spine morphology. Network pharmacology analysis confirmed that GLEXJ was closely related to the Ca2+ signaling pathway in PTSD treatment. PTSD rats exhibited disrupted TRPC6-mediated CaMKIV-CREB signaling and impaired synaptic plasticity. GLEXJ upregulated TRPC6 expression, reactivated the CaMKIV-CREB pathway, and promoted synaptic remodeling. In vitro studies confirmed that TRPC6 suppression reduced F-actin levels while GLEXJ-containing serum increased TRPC6 expression and F-actin content. CONCLUSIONS GLEXJ activates CaMKIV-CREB signaling by upregulating TRPC6 in the hippocampus of PTSD rats, leading to the positive modulation of dendritic spine morphology and synaptic remodeling. This mechanism contributes to the attenuation of fear memory generalization. Given the limitations of current PTSD treatments, these findings offer potential avenues for developing more effective therapeutic strategies.
Collapse
Affiliation(s)
- Yue Qu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Jingna Gu
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Lanxin Li
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Yuqi Yan
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Can Yan
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| | - Tiange Zhang
- Integrative Medicine Research Center, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Outer Ring East Road No. 232, Higher Education Mega Center, Guangzhou, 510006, China.
| |
Collapse
|
2
|
Heesbeen EJ, Bijlsma EY, Risseeuw TA, Hessel EVS, Groenink L. A systematic approach to identify gaps in neuroimmunology: TNF-α and fear learning deficits, a worked example. Brain Behav Immun 2025; 123:752-764. [PMID: 39442635 DOI: 10.1016/j.bbi.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND The pathophysiology of several neurodegenerative and neuropsychiatric disorders is linked to an altered immune system. However, it is often unclear how the immune system specifically affects these disorders since neuroimmune interactions are very complex. In this paper, we introduce an adjusted version of the adverse outcome pathway (AOP) approach from toxicology to the field of neuroimmunology. A review of the effect of TNF-α on fear learning deficits is used as a worked example to demonstrate how an AOP approach can help identify gaps of knowledge and crucial steps in the pathophysiology of neuroimmunological disorders. METHODS The AOP was constructed in five steps. First, the adverse outcome was formulated clearly and specifically. Second, the link between the molecular initiating event and the adverse outcome was established with a preliminary literature search in the Medline database. Third, a systematic literature search was performed in which we identified 95 relevant articles. Fourth, the main biological processes and relevant key events were identified. Fifth, the links between key events were determined and an AOP network was constructed. RESULTS We identified three pathways through which TNF-α may affect fear learning. First, TNF-α receptor activation increases NF-κB levels which increases oxidative stress levels and reduces the activity of glutamate transporters. This alters the synaptic plasticity which is associated with impaired fear acquisition, consolidation, and fear extinction. Second, activation of TNF-α receptors increases the expression and capacity of the serotonin transporter which is linked to impaired fear acquisition, expression, and extinction. Third, TNF-α receptor 1 activation can induce necroptosis, leading to neuroinflammation which is linked to fear learning deficits. CONCLUSION To successfully apply the AOP approach in neuroimmunology we recommend defining adverse outcomes more precisely, establishing stronger connections between key events from various biological processes, incorporating feedforward and feedback loops, and identifying more mechanistic knowledge in later key events. These adjustments are needed to map the complex processes within the field of neuroimmunology and to identify gaps of knowledge.
Collapse
Affiliation(s)
- Elise J Heesbeen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands (the).
| | - Elisabeth Y Bijlsma
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands (the)
| | - Tristan A Risseeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands (the)
| | - Ellen V S Hessel
- Public Health and Health Services, RIVM National Institute for Public Health and the Environment, Bilthoven, Netherlands (the)
| | - Lucianne Groenink
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands (the)
| |
Collapse
|
3
|
Pennington ZT, LaBanca AR, Sompolpong P, Abdel-Raheim SD, Ko B, Christenson Wick Z, Feng Y, Dong Z, Francisco TR, Bacon ME, Chen L, Fulton SL, Maze I, Shuman T, Cai DJ. Dissociable contributions of the amygdala and ventral hippocampus to stress-induced changes in defensive behavior. Cell Rep 2024; 43:114871. [PMID: 39427320 PMCID: PMC11849735 DOI: 10.1016/j.celrep.2024.114871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/01/2024] [Accepted: 09/27/2024] [Indexed: 10/22/2024] Open
Abstract
Stress can have profound consequences on mental health. While much is known about the neural circuits supporting associative memories of stressful events, our understanding of the circuits underlying the non-associative impacts of stress, such as heightened stress sensitivity and anxiety-related behavior, is limited. Here, we demonstrate that the ventral hippocampus (vHC) and basolateral amygdala (BLA) support distinct non-associative behavioral changes following stress. Inhibiting stress-induced protein synthesis in the BLA blocked subsequent increases in stress sensitivity but not anxiety-related behaviors. Conversely, inhibiting stress-induced protein synthesis in the vHC blocked subsequent increases in anxiety-related behavior but not stress sensitivity. Inhibiting neuronal activity in the BLA and vHC during the assessment of stress sensitivity or anxiety-related behavior recapitulated these structures' dissociable contributions to defensive behavior. Lastly, blocking the associative memory of a stressor had no impact on stress-induced changes in anxiety-related behavior. These findings highlight that multiple memory systems support the long-lasting effects of stress.
Collapse
Affiliation(s)
- Zachary T Pennington
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Alexa R LaBanca
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Patlapa Sompolpong
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shereen D Abdel-Raheim
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bumjin Ko
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zoe Christenson Wick
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yu Feng
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Zhe Dong
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Taylor R Francisco
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Madeline E Bacon
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lingxuan Chen
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sasha L Fulton
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ian Maze
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Howard Hughes Medical Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tristan Shuman
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Denise J Cai
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
4
|
Chen C, Li S, Zhou Y, Huang H, Lin JT, Wu WF, Qiu YK, Dong W, Wan J, Liu Q, Zheng H, Wu YQ, Zhou CH. Neuronal excitation-inhibition imbalance in the basolateral amygdala is involved in propofol-mediated enhancement of fear memory. Commun Biol 2024; 7:1408. [PMID: 39472670 PMCID: PMC11522401 DOI: 10.1038/s42003-024-07105-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
Posttraumatic stress disorder (PTSD) is associated with glutamatergic neuron hyperactivation in the basolateral amygdala (BLA) brain area, while GABAergic interneurons in the BLA modulate glutamatergic neuron excitability. Studies have shown that propofol exerts its effects through potentiation of the inhibitory neurotransmitter γ-aminobutyric acid. The neuronal mechanism by which propofol anesthesia modulates fear memory is currently unknown. Here, we used optogenetics and chemogenetics to suppress glutamatergic neurons or activate GABAergic interneurons in the BLA to assess alterations in neuronal excitation-inhibition balance and investigate fear memory. The excitability of glutamatergic neurons in the BLA was significantly reduced by the suppression of glutamatergic neurons or activation of GABAergic interneurons, while propofol-mediated enhancement of fear memory was attenuated. We suggest that propofol anesthesia could reduce the excitability of GABAergic neurons through activation of GABAA receptors, subsequently increasing the excitability of glutamatergic neurons in the mice BLA; the effect of propofol on enhancing mice fear memory might be mediated by strengthening glutamatergic neuronal excitability and decreasing the excitability of GABAergic neurons in the BLA; neuronal excitation-inhibition imbalance in the BLA might be important in mediating the enhancement of fear memory induced by propofol.
Collapse
Affiliation(s)
- Chen Chen
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Zhou
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hui Huang
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Jia-Tao Lin
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Wei-Feng Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Yong-Kang Qiu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Wei Dong
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Jie Wan
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Qiang Liu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yu-Qing Wu
- Jiangsu Province Key Laboratory of Anesthesiology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, Xuzhou, China.
| | - Cheng-Hua Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
5
|
Cheng L, Xiao L, Lin W, Li M, Liu J, Qiu X, Li M, Zheng Y, Xu C, Wang Y, Chen Z. Histamine H 1 receptors in dentate gyrus-projecting cholinergic neurons of the medial septum suppress contextual fear retrieval in mice. Nat Commun 2024; 15:5805. [PMID: 38987240 PMCID: PMC11237085 DOI: 10.1038/s41467-024-50042-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 06/28/2024] [Indexed: 07/12/2024] Open
Abstract
Fear memory is essential for survival and adaptation, yet excessive fear memories can lead to emotional disabilities and mental disorders. Despite previous researches have indicated that histamine H1 receptor (H1R) exerts critical and intricate effects on fear memory, the role of H1R is still not clarified. Here, we show that deletion of H1R gene in medial septum (MS) but not other cholinergic neurons selectively enhances contextual fear memory without affecting cued memory by differentially activating the dentate gyrus (DG) neurons in mice. H1R in cholinergic neurons mediates the contextual fear retrieval rather than consolidation by decreasing acetylcholine release pattern in DG. Furthermore, selective knockdown of H1R in the MS is sufficient to enhance contextual fear memory by manipulating the retrieval-induced neurons in DG. Our results suggest that H1R in MS cholinergic neurons is critical for contextual fear retrieval, and could be a potential therapeutic target for individuals with fear-related disorders.
Collapse
Affiliation(s)
- Li Cheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Ling Xiao
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Wenkai Lin
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Minzhu Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiaying Liu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaoyun Qiu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Menghan Li
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Yanrong Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Cenglin Xu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
6
|
Pennington ZT, LaBanca AR, Sompolpong P, Abdel-Raheim SD, Ko B, Christenson Wick Z, Feng Y, Dong Z, Francisco TR, Bacon ME, Chen L, Fulton SL, Maze I, Shuman T, Cai DJ. Dissociable contributions of the amygdala and ventral hippocampus to stress-induced changes in defensive behavior. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.27.530077. [PMID: 36945605 PMCID: PMC10028838 DOI: 10.1101/2023.02.27.530077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
Abstract
BACKGROUND Severe stress can produce multiple persistent changes in defensive behavior relevant to psychiatric illness. While much is known about the circuits supporting stress-induced associative fear, how stress-induced circuit plasticity supports non-associative changes in defensive behavior remains unclear. METHODS Mice were exposed to an acute severe stressor, and subsequently, both associative and non-associative defensive behavioral responses were assessed. A mixture of local protein synthesis inhibition, pan-neuronal chemogenetic inhibition, and projection-specific chemogenetic inhibition were utilized to isolate the roles of the basolateral amygdala (BLA) and ventral hippocampus (vHC) to the induction and expression of associative and non-associative defensive behavioral changes. RESULTS Stress-induced protein synthesis in the BLA was necessary for enhancements in stress sensitivity but not enhancements in anxiety-related behaviors, whereas protein synthesis in the vHC was necessary for enhancements in anxiety-related behavior but not enhancements in stress sensitivity. Like protein synthesis, neuronal activity of the BLA and vHC were found to differentially support the expression of these same defensive behaviors. Additionally, projection-specific inhibition of BLA-vHC connections failed to alter these behaviors, indicating that these defensive behaviors are regulated by distinct BLA and vHC circuits. Lastly, contributions of the BLA and vHC to stress sensitivity and anxiety-related behavior were independent of their contributions to associative fear. CONCLUSIONS Stress-induced plasticity in the BLA and vHC were found to support dissociable non-associative behavioral changes, with BLA supporting enhancements in stress sensitivity and vHC supporting increased anxiety-related behavior. These findings demonstrate that independent BLA and vHC circuits are critical for stress-induced defensive behaviors, and that differential targeting of BLA and vHC circuits may be needed in disease treatment.
Collapse
|
7
|
Melo MBD, Favaro VM, Oliveira MGM. The contextual fear conditioning consolidation depends on the functional interaction of the dorsal subiculum and basolateral amygdala in rats. Neurobiol Learn Mem 2023; 205:107827. [PMID: 37678544 DOI: 10.1016/j.nlm.2023.107827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/09/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
Fear conditioning tasks enable us to explore the neural basis of adaptative and maladaptive behaviors related to aversive memories. Recently, we provided the first evidence of the dorsal subiculum (DSub) involvement in contextual fear conditioning (CFC) consolidation by showing that the post-training bilateral NMDA (N-methyl-D-aspartate) receptor blockade in DSub impaired the performance of animals in the test session. As the memory consolidation process depends on the coordinated engagement of different brain regions, and the DSub share reciprocal projections with the basolateral amygdala (BLA), which is also involved in CFC, it is possible that the functional interaction between these sites can be relevant for the consolidation of this task. In this sense, the present study aimed to explore the effects of the functional disconnection of the DSub and BLA in the CFC consolidation after NMDA post-training blockade. In addition, to verify if the observed effects were due to spatial representation processes mediated by the DSub, we employed a hippocampal-independent procedure: tone fear conditioning (TFC). Results showed that the functional disconnection of these regions by post-training NMDA blockade impaired CFC consolidation, whereas there was no impairment in TFC. Altogether, the present data suggest that the DSub and BLA would functionally interact through NMDA-related synaptic plasticity to support CFC consolidation probably due to DSub-related spatial processing showing that the TFC consolidation was not disrupted. This work contributes to filling a gap of studies exploring the DSub involvement in fear conditioning by providing a broad framework of the subicular-amygdaloid connection functionality.
Collapse
Affiliation(s)
- Márcio Braga de Melo
- Departamento de Psicobiologia, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | - Vanessa Manchim Favaro
- Setor de Investigação de Doenças Neuromusculares, Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| | | |
Collapse
|
8
|
Veres JM, Fekete Z, Müller K, Andrasi T, Rovira-Esteban L, Barabas B, Papp OI, Hajos N. Fear learning and aversive stimuli differentially change excitatory synaptic transmission in perisomatic inhibitory cells of the basal amygdala. Front Cell Neurosci 2023; 17:1120338. [PMID: 37731462 PMCID: PMC10507864 DOI: 10.3389/fncel.2023.1120338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
Inhibitory circuits in the basal amygdala (BA) have been shown to play a crucial role in associative fear learning. How the excitatory synaptic inputs received by BA GABAergic interneurons are influenced by memory formation, a network parameter that may contribute to learning processes, is still largely unknown. Here, we investigated the features of excitatory synaptic transmission received by the three types of perisomatic inhibitory interneurons upon cue-dependent fear conditioning and aversive stimulus and tone presentations without association. Acute slices were prepared from transgenic mice: one group received tone presentation only (conditioned stimulus, CS group), the second group was challenged by mild electrical shocks unpaired with the CS (unsigned unconditioned stimulus, unsigned US group) and the third group was presented with the CS paired with the US (signed US group). We found that excitatory synaptic inputs (miniature excitatory postsynaptic currents, mEPSCs) recorded in distinct interneuron types in the BA showed plastic changes with different patterns. Parvalbumin (PV) basket cells in the unsigned US and signed US group received mEPSCs with reduced amplitude and rate in comparison to the only CS group. Coupling the US and CS in the signed US group caused a slight increase in the amplitude of the events in comparison to the unsigned US group, where the association of CS and US does not take place. Excitatory synaptic inputs onto cholecystokinin (CCK) basket cells showed a markedly different change from PV basket cells in these behavioral paradigms: only the decay time was significantly faster in the unsigned US group compared to the only CS group, whereas the amplitude of mEPSCs increased in the signed US group compared to the only CS group. Excitatory synaptic inputs received by PV axo-axonic cells showed the least difference in the three behavioral paradigm: the only significant change was that the rate of mEPSCs increased in the signed US group when compared to the only CS group. These results collectively show that associative learning and aversive stimuli unpaired with CS cause different changes in excitatory synaptic transmission in BA perisomatic interneuron types, supporting the hypothesis that they play distinct roles in the BA network operations upon pain information processing and fear memory formation.
Collapse
Affiliation(s)
- Judit M. Veres
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
| | - Zsuzsanna Fekete
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Kinga Müller
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Tibor Andrasi
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
| | - Laura Rovira-Esteban
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
| | - Bence Barabas
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai School of Neurosciences, Semmelweis University, Budapest, Hungary
| | - Orsolya I. Papp
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
| | - Norbert Hajos
- Laboratory of Network Neurophysiology, ELRN Institute of Experimental Medicine, Budapest, Hungary
- The Linda and Jack Gill Center for Molecular Bioscience, Indiana University Bloomington, Bloomington, IN, United States
- Program in Neuroscience, Department of Psychological and Brain Sciences, Indiana University Bloomington, Bloomington, IN, United States
| |
Collapse
|
9
|
Kenna M, Marek R, Sah P. Insights into the encoding of memories through the circuitry of fear. Curr Opin Neurobiol 2023; 80:102712. [PMID: 37003106 DOI: 10.1016/j.conb.2023.102712] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 04/03/2023]
Abstract
Associative learning induces physical changes to a network of cells, known as the memory engram. Fear is widely used as a model to understand the circuit motifs that underpin associative memories. Recent advances suggest that the distinct circuitry engaged by different conditioned stimuli (e.g. tone vs. context) can provide insights into what information is being encoded in the fear engram. Moreover, as the fear memory matures, the circuitry engaged indicates how information is remodelled after learning and hints at potential mechanisms for consolidation. Finally, we propose that the consolidation of fear memories involves plasticity of engram cells through coordinated activity between brain regions, and the inherent characteristics of the circuitry may mediate this process.
Collapse
Affiliation(s)
- Matthew Kenna
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Roger Marek
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Pankaj Sah
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
10
|
Goodman J, Leong KC, Packard MG. NMDA receptor blockade in the dorsolateral striatum impairs consolidation but not retrieval of habit memory. Neurobiol Learn Mem 2023; 197:107709. [PMID: 36503101 DOI: 10.1016/j.nlm.2022.107709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/24/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022]
Abstract
The present study investigated whether N-methyl-d-aspartate (NMDA) receptors in the dorsolateral striatum (DLS) mediate consolidation and retrieval of habit memory. Adult male Long-Evans rats were trained in a response learning version of a water plus-maze task in which rats were reinforced to make a habitual and consistent body-turn response at the maze choice point in order to mount a hidden escape platform. Prior research indicates that acquisition, consolidation, and retrieval in this task requires DLS function. The present study consisted of two experiments. In Experiment 1, rats received intra-DLS post-training injections of the NMDA receptor antagonist 2-amino-5- phosphonopentanoic acid (AP5; 2 µg/side) to examine the role of NMDA receptors in consolidation of habit memory. In Experiment 2, different groups of rats received a single pre-retrieval injection of AP5 in the DLS (AP5; 2 µg/side) during the last day of maze training to examine the potential role of NMDA receptors in retrieval of habit memory. Results indicated that post-training intra-DLS AP5 injections impaired memory consolidation. However, administration of AP5 at the same dose that impaired consolidation had no effect on memory retrieval. The findings are consistent with previous research indicating a role for NMDA receptors in the DLS in memory consolidation, and suggest that NMDA-dependent synaptic activity in the DLS may not be a critical component of habit memory retrieval.
Collapse
Affiliation(s)
- Jarid Goodman
- Department of Psychology, Delaware State University, Dover, DE, United States
| | - Kah-Chung Leong
- Department of Psychology, Trinity University, San Antonio, TX, United States
| | - Mark G Packard
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
11
|
Van Assche IA, Padilla MS, Stupart OSRP, Milton AL. Refinement of the stress-enhanced fear learning model of post-traumatic stress disorder: a behavioral and molecular analysis. Lab Anim (NY) 2022; 51:293-300. [PMID: 36266512 DOI: 10.1038/s41684-022-01054-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 08/15/2022] [Indexed: 11/05/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating mental health condition for which current treatments have long-term efficacy in 50% of patients. There is a clear need for better understanding of the mechanisms underlying PTSD and the development of new treatment approaches. Analog trauma procedures in animals, such as the stress-enhanced fear learning (SEFL) procedure, can be used to produce behavioral and neurobiological changes that have validity in modeling PTSD. However, by necessity, the modeling of PTSD in animals requires them to potentially experience pain and suffering. Consistent with the '3Rs' (reduction, refinement and replacement) of animal research, this study aimed to determine whether the SEFL procedure can be refined to reduce potential animal pain and suffering while retaining the same behavioral and neurobiological changes. Here we showed that PTSD-relevant changes could be produced in both behavior and the brain of rats that were group- rather than single-housed and that received lower-magnitude electric shocks in the 'trauma analog' session. We also varied the number of shock exposures in the trauma analog session, finding SEFL-susceptible and SEFL-resilient populations at all levels of shock exposure, but with greater levels of shock increasing the proportion of rats showing the SEFL-susceptible phenotype. These data demonstrate that the SEFL procedure can be used as an animal analog of PTSD with reduced potential pain and suffering to the animals and that variations in the procedure could be used to generate specific proportions of SEFL-susceptible and SEFL-resilient animals in future studies.
Collapse
Affiliation(s)
- Indra A Van Assche
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, UK.,Biomedical Sciences Group: Woman and Child, KU Leuven, Leuven, Belgium
| | - Mc Stephen Padilla
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, UK
| | | | - Amy L Milton
- Department of Psychology, University of Cambridge, Downing Site, Cambridge, UK.
| |
Collapse
|
12
|
Jiang YL, Wang XS, Li XB, Liu A, Fan QY, Yang L, Feng B, Zhang K, Lu L, Qi JY, Yang F, Song DK, Wu YM, Zhao MG, Liu SB. Tanshinone IIA improves contextual fear- and anxiety-like behaviors in mice via the CREB/BDNF/TrkB signaling pathway. Phytother Res 2022; 36:3932-3948. [PMID: 35801985 DOI: 10.1002/ptr.7540] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 05/27/2022] [Accepted: 06/11/2022] [Indexed: 11/11/2022]
Abstract
Posttraumatic stress disorder (PTSD) is one of the most common psychiatric diseases, which is characterized by the typical symptoms such as re-experience, avoidance, and hyperarousal. However, there are few drugs for PTSD treatment. In this study, conditioned fear and single-prolonged stress were employed to establish PTSD mouse model, and we investigated the effects of Tanshinone IIA (TanIIA), a natural product isolated from traditional Chinese herbal Salvia miltiorrhiza, as well as the underlying mechanisms in mice. The results showed that the double stress exposure induced obvious PTSD-like symptoms, and TanIIA administration significantly decreased freezing time in contextual fear test and relieved anxiety-like behavior in open field and elevated plus maze tests. Moreover, TanIIA increased the spine density and upregulated synaptic plasticity-related proteins as well as activated CREB/BDNF/TrkB signaling pathway in the hippocampus. Blockage of CREB remarkably abolished the effects of TanIIA in PTSD model mice and reversed the upregulations of p-CREB, BDNF, TrkB, and synaptic plasticity-related protein induced by TanIIA. The molecular docking simulation indicated that TanIIA could interact with the CREB-binding protein. These findings indicate that TanIIA ameliorates PTSD-like behaviors in mice by activating the CREB/BDNF/TrkB pathway, which provides a basis for PTSD treatment.
Collapse
Affiliation(s)
- Yong-Li Jiang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin-Shang Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Xu-Bo Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - An Liu
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Qing-Yu Fan
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Le Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Ban Feng
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Liang Lu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Jing-Yu Qi
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Fan Yang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Da-Ke Song
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| | - Ming-Gao Zhao
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
13
|
Effects of Intra-BLA Administration of PPAR Antagonists on Formalin-Evoked Nociceptive Behaviour, Fear-Conditioned Analgesia, and Conditioned Fear in the Presence or Absence of Nociceptive Tone in Rats. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27062021. [PMID: 35335382 PMCID: PMC8949000 DOI: 10.3390/molecules27062021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 11/16/2022]
Abstract
There is evidence for the involvement of peroxisome proliferator-activated receptors (PPARs) in pain, cognition, and anxiety. However, their role in pain–fear interactions is unknown. The amygdala plays a key role in pain, conditioned fear, and fear-conditioned analgesia (FCA). We investigated the effects of intra-basolateral amygdala (BLA) administration of PPARα, PPARβ/δ, and PPARγ antagonists on nociceptive behaviour, FCA, and conditioned fear in the presence or absence of nociceptive tone. Male Sprague-Dawley (SD) rats received footshock (FC) or no footshock (NFC) in a conditioning arena. Twenty-three and a half hours later, rats received an intraplantar injection of formalin or saline and, 15 min later, intra-BLA microinjections of vehicle, PPARα (GW6471) PPARβ/δ (GSK0660), or PPARγ (GW9662) antagonists before arena re-exposure. Pain and fear-related behaviour were assessed, and neurotransmitters/endocannabinoids measured post-mortem. Intra-BLA administration of PPARα or PPARγ antagonists potentiated freezing in the presence of nociceptive tone. Blockade of all PPAR subtypes in the BLA increased freezing and BLA dopamine levels in NFC rats in the absence of nociceptive tone. Administration of intra-BLA PPARα and PPARγ antagonists increased levels of dopamine in the BLA compared with the vehicle-treated counterparts. In conclusion, PPARα and PPARγ in the BLA play a role in the expression or extinction of conditioned fear in the presence or absence of nociceptive tone.
Collapse
|
14
|
Namkung H, Thomas KL, Hall J, Sawa A. Parsing neural circuits of fear learning and extinction across basic and clinical neuroscience: Towards better translation. Neurosci Biobehav Rev 2022; 134:104502. [PMID: 34921863 DOI: 10.1016/j.neubiorev.2021.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 12/22/2022]
Abstract
Over the past decades, studies of fear learning and extinction have advanced our understanding of the neurobiology of threat and safety learning. Animal studies can provide mechanistic/causal insights into human brain regions and their functional connectivity involved in fear learning and extinction. Findings in humans, conversely, may further enrich our understanding of neural circuits in animals by providing macroscopic insights at the level of brain-wide networks. Nevertheless, there is still much room for improvement in translation between basic and clinical research on fear learning and extinction. Through the lens of neural circuits, in this article, we aim to review the current knowledge of fear learning and extinction in both animals and humans, and to propose strategies to fill in the current knowledge gap for the purpose of enhancing clinical benefits.
Collapse
Affiliation(s)
- Ho Namkung
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Kerrie L Thomas
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK; School of Biosciences, Cardiff University, Cardiff, UK
| | - Jeremy Hall
- Neuroscience and Mental Health Research Institute, Cardiff University, Cardiff, UK; School of Medicine, Cardiff University, Cardiff, UK
| | - Akira Sawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, 21287, USA.
| |
Collapse
|
15
|
Souto JJ, Silva GM, Almeida NL, Shoshina II, Santos NA, Fernandes TP. Age-related episodic memory decline and the role of amyloid-β: a systematic review. Dement Neuropsychol 2021; 15:299-313. [PMID: 34630918 PMCID: PMC8485646 DOI: 10.1590/1980-57642021dn15-030002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 02/22/2021] [Indexed: 12/25/2022] Open
Abstract
Aging has been associated with the functional decline of episodic memory (EM). Unanswered questions are whether the decline of EM occurs even during healthy aging and whether this decline is related to amyloid-β (Aβ) deposition in the hippocampus. Objective The main purpose of this study was to investigate data on the relationship between the age-related EM decline and Aβ deposition. Methods We searched the Cochrane, MEDLINE, Scopus, and Web of Science databases and reference lists of retrieved articles that were published in the past 10 years. The initial literature search identified 517 studies. After screening the title, abstract, key words, and reference lists, 56 studies met the inclusion criteria. Results The overall results revealed that increases in Aβ are related to lower hippocampal volume and worse performance on EM tests. The results of this systematic review revealed that high levels of Aβ may be related to EM deficits and the progression to Alzheimer's disease. Conclusions We discussed the strengths and pitfalls of various tests and techniques used for investigating EM and Aβ deposition, methodological issues, and potential directions for future research.
Collapse
Affiliation(s)
- Jandirlly Julianna Souto
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| | - Gabriella Medeiros Silva
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| | - Natalia Leandro Almeida
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| | | | - Natanael Antonio Santos
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| | - Thiago Paiva Fernandes
- Department of Psychology, Universidade Federal da Paraíba - João Pessoa, PB, Brazil.,Perception, Neuroscience and Behaviour Laboratory, Universidade Federal da Paraíba - João Pessoa, Brazil
| |
Collapse
|
16
|
Alexander C, Vasefi M. Cannabidiol and the corticoraphe circuit in post-traumatic stress disorder. IBRO Neurosci Rep 2021; 11:88-102. [PMID: 34485973 PMCID: PMC8408530 DOI: 10.1016/j.ibneur.2021.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/04/2021] [Accepted: 08/18/2021] [Indexed: 01/06/2023] Open
Abstract
Post-Traumatic Stress Disorder (PTSD), characterized by re-experiencing, avoidance, negative affect, and impaired memory processing, may develop after traumatic events. PTSD is complicated by impaired plasticity and medial prefrontal cortex (mPFC) activity, hyperactivity of the amygdala, and impaired fear extinction. Cannabidiol (CBD) is a promising candidate for treatment due to its multimodal action that enhances plasticity and calms hyperexcitability. CBD’s mechanism in the mPFC of PTSD patients has been explored extensively, but literature on the mechanism in the dorsal raphe nucleus (DRN) is lacking. Following the PRISMA guidelines, we examined current literature regarding CBD in PTSD and overlapping symptomologies to propose a mechanism by which CBD treats PTSD via corticoraphe circuit. Acute CBD inhibits excess 5-HT release from DRN to amygdala and releases anandamide (AEA) onto amygdala inputs. By first reducing amygdala and DRN hyperactivity, CBD begins to ameliorate activity disparity between mPFC and amygdala. Chronic CBD recruits the mPFC, creating harmonious corticoraphe signaling. DRN releases enough 5-HT to ameliorate mPFC hypoactivity, while the mPFC continuously excites DRN 5-HT neurons via glutamate. Meanwhile, AEA regulates corticoraphe activity to stabilize signaling. AEA prevents DRN GABAergic interneurons from inhibiting 5-HT release so the DRN can assist the mPFC in overcoming its hypoactivity. DRN-mediated restoration of mPFC activity underlies CBD’s mechanism on fear extinction and learning of stress coping. CBD reduces PTSD symptoms via the DRN and corticoraphe circuit. Acute effects of CBD reduce DRN-amygdala excitatory signaling to lessen the activity disparity between amygdala and mPFC. Chronic CBD officially resolves mPFC hypoactivity by facilitating 5-HT release from DRN to mPFC. CBD-facilitated endocannabinoid signaling stabilizes DRN activity and restores mPFC inhibitory control. Chronically administered CBD acts via the corticoraphe circuit to favor fear extinction over fear memory reconsolidation.
Collapse
Key Words
- 2-AG, 2-arachidonoylglycerol
- 5-HT, Serotonin
- 5-HT1AR, 5-HT Receptor Type 1A
- 5-HT2AR, 5-HT Receptor Type 2 A
- AEA, Anandamide
- CB1R, Cannabinoid Receptor Type 1
- CB2R, Cannabinoid Receptor Type 2
- CBD, Cannabidiol
- COVID-19, SARS-CoV-2
- Cannabidiol
- DRN, Dorsal Raphe Nucleus
- ERK1/2, Extracellular Signal-Related Kinases Type 1 or Type 2
- FAAH, Fatty Acid Amide Hydrolase
- GABA, Gamma-Aminobutyric Acid
- GPCRs, G-Protein Coupled Receptors
- NMDAR, N-Methyl-D-aspartate Receptors
- PET, Positron Emission Tomography
- PFC, DRN and Raphe
- PFC, Prefrontal Cortex
- PTSD
- PTSD, Post-Traumatic Stress Disorder
- SSNRI, Selective Norepinephrine Reuptake Inhibitor
- SSRI, Selective Serotonin Reuptake Inhibitor
- Serotonin
- TRPV1, Transient Receptor Potential Vanilloid 1 Channels
- Traumatic Stress
- fMRI, Functional Magnetic Resonance Imaging
- mPFC, Medial Prefrontal Cortex
Collapse
Affiliation(s)
- Claire Alexander
- Department of Biology, Lamar University, Beaumont, TX 77710, USA
| | - Maryam Vasefi
- Department of Biology, Lamar University, Beaumont, TX 77710, USA
| |
Collapse
|
17
|
Rajbhandari AK, Octeau CJ, Gonzalez S, Pennington ZT, Mohamed F, Trott J, Chavez J, Ngyuen E, Keces N, Hong WZ, Neve RL, Waschek J, Khakh BS, Fanselow MS. A Basomedial Amygdala to Intercalated Cells Microcircuit Expressing PACAP and Its Receptor PAC1 Regulates Contextual Fear. J Neurosci 2021; 41:3446-3461. [PMID: 33637560 PMCID: PMC8051692 DOI: 10.1523/jneurosci.2564-20.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 11/21/2022] Open
Abstract
Trauma can cause dysfunctional fear regulation leading some people to develop disorders, such as post-traumatic stress disorder (PTSD). The amygdala regulates fear, whereas PACAP (pituitary adenylate activating peptide) and PAC1 receptors are linked to PTSD symptom severity at genetic/epigenetic levels, with a strong link in females with PTSD. We discovered a PACAPergic projection from the basomedial amygdala (BMA) to the medial intercalated cells (mICCs) in adult mice. In vivo optogenetic stimulation of this pathway increased CFOS expression in mICCs, decreased fear recall, and increased fear extinction. Selective deletion of PAC1 receptors from the mICCs in females reduced fear acquisition, but enhanced fear generalization and reduced fear extinction in males. Optogenetic stimulation of the BMA-mICC PACAPergic pathway produced EPSCs in mICC neurons, which were enhanced by the PAC1 receptor antagonist, PACAP 6-38. Our findings show that mICCs modulate contextual fear in a dynamic and sex-dependent manner via a microcircuit containing the BMA and mICCs, and in a manner that was dependent on behavioral state.SIGNIFICANCE STATEMENT Traumatic stress can affect different aspects of fear behaviors, including fear learning, generalization of learned fear to novel contexts, how the fear of the original context is recalled, and how fear is reduced over time. While the amygdala has been studied for its role in regulation of different aspects of fear, the molecular circuitry of this structure is quite complex. In addition, aspects of fear can be modulated differently in males and females. Our findings show that a specific circuitry containing the neuropeptide PACAP and its receptor, PAC1, regulates various aspects of fear, including acquisition, generalization, recall, and extinction in a sexually dimorphic manner, characterizing a novel pathway that modulates traumatic fear.
Collapse
Affiliation(s)
- Abha K Rajbhandari
- Department of Psychology, University of California, Los Angeles, California 90095
- Staglin Center for Brain and Behavior, University of California, Los Angeles, California 90095
| | - Christopher J Octeau
- Department of Physiology, University of California, Los Angeles, California 90095
| | - Sarah Gonzalez
- Department of Psychology, University of California, Los Angeles, California 90095
- Staglin Center for Brain and Behavior, University of California, Los Angeles, California 90095
| | - Zachary T Pennington
- Department of Psychology, University of California, Los Angeles, California 90095
| | - Farzanna Mohamed
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Jeremy Trott
- Department of Psychology, University of California, Los Angeles, California 90095
- Staglin Center for Brain and Behavior, University of California, Los Angeles, California 90095
| | - Jasmine Chavez
- Department of Psychology, University of California, Los Angeles, California 90095
| | - Erin Ngyuen
- Department of Psychology, University of California, Los Angeles, California 90095
| | - Natasha Keces
- Department of Psychology, University of California, Los Angeles, California 90095
| | - Weizhe Z Hong
- Department of Neurobiology, University of California, Los Angeles, California 90095
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, Boston, Massachusetts, California 02114
| | - James Waschek
- Department of Psychiatry, University of California, Los Angeles, California 90095
| | - Baljit S Khakh
- Department of Physiology, University of California, Los Angeles, California 90095
- Department of Neurobiology, University of California, Los Angeles, California 90095
| | - Michael S Fanselow
- Department of Psychology, University of California, Los Angeles, California 90095
- Staglin Center for Brain and Behavior, University of California, Los Angeles, California 90095
| |
Collapse
|
18
|
Yegla B, Boles J, Kumar A, Foster TC. Partial microglial depletion is associated with impaired hippocampal synaptic and cognitive function in young and aged rats. Glia 2021; 69:1494-1514. [PMID: 33586813 DOI: 10.1002/glia.23975] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/11/2022]
Abstract
The role of microglia in mediating age-related changes in cognition and hippocampal synaptic function was examined by microglial depletion and replenishment using PLX3397. We observed age-related differences in microglial number and morphology, as well as increased Iba-1 expression, indicating microglial activation. PLX3397 treatment decreased microglial number, with aged rats exhibiting the lowest density. Young rats exhibited increased expression of pro-inflammatory cytokines during depletion and repopulation and maintenance of Iba-1 levels despite reduced microglial number. For aged rats, several cytokines increased with depletion and recovered during repopulation; however, aged rats did not fully recover microglial cell number or Iba-1 expression during repopulation, with a recovery comparable to young control levels rather than aged controls. Hippocampal CA3-CA1 synaptic transmission was impaired with age, and microglial depletion was associated with decreased total synaptic transmission in young and aged rats. A robust decline in N-methyl-d-aspartate-receptor-mediated synaptic transmission arose in young depleted rats specifically. Microglial replenishment normalized depletion-induced synaptic function to control levels; however, recovery of aged animals did not mirror young. Microglial depletion was associated with decreased context-object discrimination memory in both age groups, which recovered with microglial repopulation. Aged rats displayed impaired contextual and cued fear memory, and microglial replenishment did not recover their memory to the level of young. The current study indicates that cognitive function and synaptic transmission benefit from the support of aged microglia and are hindered by removal of these cells. Replenishment of microglia in aging did not ameliorate age-related cognitive impairments or senescent synaptic function.
Collapse
Affiliation(s)
- Brittney Yegla
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Jake Boles
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Ashok Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA
| | - Thomas C Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, Florida, USA.,Genetics and Genomics Program, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
19
|
León LA, Brandão ML, Cardenas FP, Parra D, Krahe TE, Cruz APM, Landeira-Fernandez J. Distinct patterns of brain Fos expression in Carioca High- and Low-conditioned Freezing Rats. PLoS One 2020; 15:e0236039. [PMID: 32702030 PMCID: PMC7377485 DOI: 10.1371/journal.pone.0236039] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 06/28/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The bidirectional selection of high and low anxiety-like behavior is a valuable tool for understanding the neurocircuits that are responsible for anxiety disorders. Our group developed two breeding lines of rats, known as Carioca High- and Low-conditioned Freezing (CHF and CLF), based on defensive freezing in the contextual fear conditioning paradigm. A random selected line was employed as a control (CTL) comparison group for both CHF and CLF lines of animals. The present study performed Fos immunochemistry to investigate changes in neural activity in different brain structures among CHF and CLF rats when they were exposed to contextual cues that were previously associated with footshock. RESULTS The study indicated that CHF rats expressed high Fos expression in the locus coeruleus, periventricular nucleus of the hypothalamus (PVN), and lateral portion of the septal area and low Fos expression in the medial portion of the septal area, dentate gyrus, and prelimbic cortex (PL) compared to CTL animals. CLF rats exhibited a decrease in Fos expression in the PVN, PL, and basolateral nucleus of the amygdala and increase in the cingulate and perirhinal cortices compared to CTL animals. CONCLUSIONS Both CHF and CLF rats displayed Fos expression changes key regions of the anxiety brain circuitry. The two bidirectional lines exhibit different pattern of neural activation and inhibition with opposing influences on the PVN, the main structure involved in regulating the hypothalamic-pituitary-adrenal neuroendocrine responses observed in anxiety disorders.
Collapse
Affiliation(s)
- Laura A. León
- Laboratory of Neuropsychopharmacology, FFCLRP, Behavioral Neuroscience Institute (INeC), São Paulo University, Campus USP, Ribeirão Preto, São Paulo, Brazil
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
- Programa de Psicología, Universidad Sergio Arboleda, Bogotá, Colombia
| | - Marcus L. Brandão
- Laboratory of Neuropsychopharmacology, FFCLRP, Behavioral Neuroscience Institute (INeC), São Paulo University, Campus USP, Ribeirão Preto, São Paulo, Brazil
| | - Fernando P. Cardenas
- Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Bogotá, Colombia
| | - Diana Parra
- Laboratorio de Neurociencia y Comportamiento, Universidad de los Andes, Bogotá, Colombia
| | - Thomas E. Krahe
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - J. Landeira-Fernandez
- Department of Psychology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
20
|
Harnett NG, Goodman AM, Knight DC. PTSD-related neuroimaging abnormalities in brain function, structure, and biochemistry. Exp Neurol 2020; 330:113331. [PMID: 32343956 DOI: 10.1016/j.expneurol.2020.113331] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 04/06/2020] [Accepted: 04/24/2020] [Indexed: 12/20/2022]
Abstract
Although approximately 90% of the U.S. population will experience a traumatic event within their lifetime, only a fraction of those traumatized individuals will develop posttraumatic stress disorder (PTSD). In fact, approximately 7 out of 100 people in the U.S. will be afflicted by this debilitating condition, which suggests there is substantial inter-individual variability in susceptibility to PTSD. This uncertainty regarding who is susceptible to PTSD necessitates a thorough understanding of the neurobiological processes that underlie PTSD development in order to build effective predictive models for the disorder. In turn, these predictive models may lead to the development of improved diagnostic markers, early intervention techniques, and targeted treatment approaches for PTSD. Prior research has characterized a fear learning and memory network, centered on the prefrontal cortex, hippocampus, and amygdala, that plays a key role in the pathology of PTSD. Importantly, changes in the function, structure, and biochemistry of this network appear to underlie the cognitive-affective dysfunction observed in PTSD. The current review discusses prior research that has demonstrated alterations in brain function, structure, and biochemistry associated with PTSD. Further, the potential for future research to address current gaps in our understanding of the neural processes that underlie the development of PTSD is discussed. Specifically, this review emphasizes the need for multimodal neuroimaging research and investigations into the acute effects of posttraumatic stress. The present review provides a framework to move the field towards a comprehensive neurobiological model of PTSD.
Collapse
Affiliation(s)
- Nathaniel G Harnett
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adam M Goodman
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - David C Knight
- Department of Psychology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
21
|
Dulka BN, Bagatelas ED, Bress KS, Grizzell JA, Cannon MK, Whitten CJ, Cooper MA. Chemogenetic activation of an infralimbic cortex to basolateral amygdala projection promotes resistance to acute social defeat stress. Sci Rep 2020; 10:6884. [PMID: 32327679 PMCID: PMC7181792 DOI: 10.1038/s41598-020-63879-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 04/06/2020] [Indexed: 12/15/2022] Open
Abstract
Tremendous individual differences exist in stress responsivity and social defeat stress is a key approach for identifying cellular mechanisms of stress susceptibility and resilience. Syrian hamsters show reliable territorial aggression, but after social defeat they exhibit a conditioned defeat (CD) response characterized by increased submission and an absence of aggression in future social interactions. Hamsters that achieve social dominance prior to social defeat exhibit greater defeat-induced neural activity in infralimbic (IL) cortex neurons that project to the basolateral amygdala (BLA) and reduced CD response compared to subordinate hamsters. Here, we hypothesize that chemogenetic activation of an IL-to-BLA neural projection during acute social defeat will reduce the CD response in subordinate hamsters and thereby produce dominant-like behavior. We confirmed that clozapine-N-oxide (CNO) itself did not alter the CD response and validated a dual-virus, Cre-dependent, chemogenetic approach by showing that CNO treatment increased c-Fos expression in the IL and decreased it in the BLA. We found that CNO treatment during social defeat reduced the acquisition of CD in subordinate, but not dominant, hamsters. This project extends our understanding of the neural circuits underlying resistance to acute social stress, which is an important step toward delineating circuit-based approaches for the treatment of stress-related psychopathologies.
Collapse
Affiliation(s)
- Brooke N Dulka
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA.,Department of Psychology, University of Wisconsin-Milwaukee, Milwaukee, WI, 53211, USA
| | - Elena D Bagatelas
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Kimberly S Bress
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA
| | - J Alex Grizzell
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Megan K Cannon
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Conner J Whitten
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA
| | - Matthew A Cooper
- Department of Psychology, University of Tennessee, Knoxville, TN, 37996, USA.
| |
Collapse
|
22
|
Encoding of contextual fear memory in hippocampal-amygdala circuit. Nat Commun 2020; 11:1382. [PMID: 32170133 PMCID: PMC7069961 DOI: 10.1038/s41467-020-15121-2] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 02/15/2020] [Indexed: 01/12/2023] Open
Abstract
In contextual fear conditioning, experimental subjects learn to associate a neutral context with an aversive stimulus and display fear responses to a context that predicts danger. Although the hippocampal–amygdala pathway has been implicated in the retrieval of contextual fear memory, the mechanism by which fear memory is encoded in this circuit has not been investigated. Here, we show that activity in the ventral CA1 (vCA1) hippocampal projections to the basal amygdala (BA), paired with aversive stimuli, contributes to encoding conditioned fear memory. Contextual fear conditioning induced selective strengthening of a subset of vCA1–BA synapses, which was prevented under anisomycin-induced retrograde amnesia. Moreover, a subpopulation of BA neurons receives stronger monosynaptic inputs from context-responding vCA1 neurons, whose activity was required for contextual fear learning and synaptic potentiation in the vCA1–BA pathway. Our study suggests that synaptic strengthening of vCA1 inputs conveying contextual information to a subset of BA neurons contributes to encoding adaptive fear memory for the threat-predictive context. Previous studies implicate the hippocampal–amygdala pathway in contextual fear conditioning, in which animals learn to associate a neutral context with an aversive stimulus and display fear responses to dangerous situations. Here the authors show that selective strengthening of hippocampal–amygdala pathway contributes to encoding adaptive fear memory for threat-predictive context.
Collapse
|
23
|
Stress reactivity after traumatic brain injury: implications for comorbid post-traumatic stress disorder. Behav Pharmacol 2020; 30:115-121. [PMID: 30640181 DOI: 10.1097/fbp.0000000000000461] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Most people have or will experience traumatic stress at some time over the lifespan, but only a subset of traumatized individuals develop post-traumatic stress disorder (PTSD). Clinical research supports high rates of traumatic brain injury (TBI)-PTSD comorbidity and demonstrates TBI as a significant predictor of the development of PTSD. Biological factors impacted following brain injury that may contribute to increased PTSD risk are unknown. Heightened stress reactivity and dysregulated hypothalamic-pituitary-adrenal (HPA) axis function are common to both TBI and PTSD, and affect amygdalar structure and function, which is implicated in PTSD. In this review, we summarize a growing body of literature that shows HPA axis dysregulation, as well as enhanced fear and amygdalar function after TBI. We present the hypothesis that altered stress reactivity as a result of brain injury impacts the amygdala and defense systems to be vulnerable to increased fear and PTSD development from traumatic stress. Identifying biological mechanisms that underlie this vulnerability, such as dysregulated HPA axis function, may lead to better targeted treatments and preventive measures to support psychological health after TBI.
Collapse
|
24
|
Helfer P, Shultz TR. A computational model of systems memory consolidation and reconsolidation. Hippocampus 2019; 30:659-677. [PMID: 31872960 DOI: 10.1002/hipo.23187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 10/05/2019] [Accepted: 12/04/2019] [Indexed: 12/27/2022]
Abstract
In the mammalian brain, newly acquired memories depend on the hippocampus (HPC) for maintenance and recall, but over time, the neocortex takes over these functions, rendering memories HPC-independent. The process responsible for this transformation is called systems memory consolidation. Reactivation of a well-consolidated memory can trigger a temporary return to a HPC-dependent state, a phenomenon known as systems memory reconsolidation. The neural mechanisms underlying systems memory consolidation and reconsolidation are not well understood. Here, we propose a neural model based on well-documented mechanisms of synaptic plasticity and stability and describe a computational implementation that demonstrates the model's ability to account for a range of findings from the systems consolidation and reconsolidation literature. We derive several predictions from the computational model and suggest experiments that may test its validity.
Collapse
Affiliation(s)
- Peter Helfer
- Department of Psychology, McGill University, 2001 McGill College, Montreal, QC, Canada
| | - Thomas R Shultz
- Department of Psychology, McGill University, 2001 McGill College, Montreal, QC, Canada
| |
Collapse
|
25
|
Johnson LR, Battle AR, Martinac B. Remembering Mechanosensitivity of NMDA Receptors. Front Cell Neurosci 2019; 13:533. [PMID: 31866826 PMCID: PMC6906178 DOI: 10.3389/fncel.2019.00533] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
An increase in post-synaptic Ca2+ conductance through activation of the ionotropic N-methyl-D-aspartate receptor (NMDAR) and concomitant structural changes are essential for the initiation of long-term potentiation (LTP) and memory formation. Memories can be initiated by coincident events, as occurs in classical conditioning, where the NMDAR can act as a molecular coincidence detector. Binding of glutamate and glycine, together with depolarization of the postsynaptic cell membrane to remove the Mg2+ channel pore block, results in NMDAR opening for Ca2+ conductance. Accumulating evidence has implicated both force-from-lipids and protein tethering mechanisms for mechanosensory transduction in NMDAR, which has been demonstrated by both, membrane stretch and application of amphipathic molecules such as arachidonic acid (AA). The contribution of mechanosensitivity to memory formation and consolidation may be to increase activity of the NMDAR leading to facilitated memory formation. In this review we look back at the progress made toward understanding the physiological and pathological role of NMDA receptor channels in mechanobiology of the nervous system and consider these findings in like of their potential functional implications for memory formation. We examine recent studies identifying mechanisms of both NMDAR and other mechanosensitive channels and discuss functional implications including gain control of NMDA opening probability. Mechanobiology is a rapidly growing area of biology with many important implications for understanding form, function and pathology in the nervous system.
Collapse
Affiliation(s)
- Luke R Johnson
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia.,Division of Psychology, School of Medicine, University of Tasmania, Launceston, TAS, Australia.,Department of Psychiatry, Center for the Study of Traumatic Stress, Uniformed Services University of the Health Sciences, Bethesda, MD, United States.,School of Biomedical Sciences, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, QLD, Australia
| | - Andrew R Battle
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology, Brisbane, QLD, Australia.,Prince Charles Hospital Northside Clinical Unit, School of Clinical Medicine, The University of Queensland, Brisbane, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia
| | - Boris Martinac
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia.,St. Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, Australia
| |
Collapse
|
26
|
Wong FS, Westbrook RF, Holmes NM. 'Online' integration of sensory and fear memories in the rat medial temporal lobe. eLife 2019; 8:e47085. [PMID: 31180324 PMCID: PMC6592679 DOI: 10.7554/elife.47085] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/10/2019] [Indexed: 12/04/2022] Open
Abstract
How does a stimulus never associated with danger become frightening? The present study addressed this question using a sensory preconditioning task with rats. In this task, rats integrate a sound-light memory formed in stage 1 with a light-danger memory formed in stage 2, as they show fear when tested with the sound in stage 3. Here we show that this integration occurs 'online' during stage 2: when activity in the region that consolidated the sound-light memory (perirhinal cortex) was inhibited during formation of the light-danger memory, rats no longer showed fear when tested with the sound but continued to fear the light. Thus, fear that accrues to a stimulus paired with danger simultaneously spreads to its past associates, thereby roping those associates into a fear memory network.
Collapse
Affiliation(s)
- Francesca S Wong
- School of PsychologyUniversity of New South WalesSydneyAustralia
| | - R Fred Westbrook
- School of PsychologyUniversity of New South WalesSydneyAustralia
| | - Nathan M Holmes
- School of PsychologyUniversity of New South WalesSydneyAustralia
| |
Collapse
|
27
|
Raber J, Arzy S, Bertolus JB, Depue B, Haas HE, Hofmann SG, Kangas M, Kensinger E, Lowry CA, Marusak HA, Minnier J, Mouly AM, Mühlberger A, Norrholm SD, Peltonen K, Pinna G, Rabinak C, Shiban Y, Soreq H, van der Kooij MA, Lowe L, Weingast LT, Yamashita P, Boutros SW. Current understanding of fear learning and memory in humans and animal models and the value of a linguistic approach for analyzing fear learning and memory in humans. Neurosci Biobehav Rev 2019; 105:136-177. [PMID: 30970272 DOI: 10.1016/j.neubiorev.2019.03.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 01/30/2019] [Accepted: 03/18/2019] [Indexed: 01/04/2023]
Abstract
Fear is an emotion that serves as a driving factor in how organisms move through the world. In this review, we discuss the current understandings of the subjective experience of fear and the related biological processes involved in fear learning and memory. We first provide an overview of fear learning and memory in humans and animal models, encompassing the neurocircuitry and molecular mechanisms, the influence of genetic and environmental factors, and how fear learning paradigms have contributed to treatments for fear-related disorders, such as posttraumatic stress disorder. Current treatments as well as novel strategies, such as targeting the perisynaptic environment and use of virtual reality, are addressed. We review research on the subjective experience of fear and the role of autobiographical memory in fear-related disorders. We also discuss the gaps in our understanding of fear learning and memory, and the degree of consensus in the field. Lastly, the development of linguistic tools for assessments and treatment of fear learning and memory disorders is discussed.
Collapse
Affiliation(s)
- Jacob Raber
- Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA; Departments of Neurology and Radiation Medicine, and Division of Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA.
| | - Shahar Arzy
- Department of Medical Neurobiology, Hebrew University, Jerusalem 91904, Israel
| | | | - Brendan Depue
- Departments of Psychological and Brain Sciences and Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, USA
| | - Haley E Haas
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Stefan G Hofmann
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Maria Kangas
- Department of Psychology, Macquarie University, Sydney, Australia
| | | | - Christopher A Lowry
- Department of Integrative Physiology and Center for Neuroscience, University of Colorado Boulder, Boulder, CO, USA
| | - Hilary A Marusak
- Department of Pharmacy Practice, Wayne State University, Detroit, MI, USA
| | - Jessica Minnier
- School of Public Health, Oregon Health & Science University, Portland, OR, USA
| | - Anne-Marie Mouly
- Lyon Neuroscience Research Center, CNRS-UMR 5292, INSERM U1028, Université Lyon, Lyon, France
| | - Andreas Mühlberger
- Department of Psychology (Clinical Psychology and Psychotherapy), University of Regensburg, Regensburg, Germany; PFH - Private University of Applied Sciences, Department of Psychology (Clinical Psychology and Psychotherapy Research), Göttingen, Germany
| | - Seth Davin Norrholm
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Kirsi Peltonen
- Faculty of Social Sciences/Psychology, Tampere University, Tampere, Finland
| | - Graziano Pinna
- The Psychiatric Institute, Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, USA
| | - Christine Rabinak
- Department of Pharmacy Practice, Wayne State University, Detroit, MI, USA
| | - Youssef Shiban
- Department of Psychology (Clinical Psychology and Psychotherapy), University of Regensburg, Regensburg, Germany; PFH - Private University of Applied Sciences, Department of Psychology (Clinical Psychology and Psychotherapy Research), Göttingen, Germany
| | - Hermona Soreq
- Department of Biological Chemistry, Edmond and Lily Safra Center of Brain Science and The Institute of Life Sciences, Hebrew University, Jerusalem 91904, Israel
| | - Michael A van der Kooij
- Translational Psychiatry, Department of Psychiatry and Psychotherapy, Universitatsmedizin der Johannes Guttenberg University Medical Center, Mainz, Germany
| | | | - Leah T Weingast
- Department of Psychiatry and Behavioral Science, Emory University School of Medicine, Atlanta, GA, USA
| | - Paula Yamashita
- School of Public Health, Oregon Health & Science University, Portland, OR, USA
| | - Sydney Weber Boutros
- Department of Behavioral Neuroscience, ONPRC, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
28
|
Pagani MR, Merlo E. Kinase and Phosphatase Engagement Is Dissociated Between Memory Formation and Extinction. Front Mol Neurosci 2019; 12:38. [PMID: 30842725 PMCID: PMC6391346 DOI: 10.3389/fnmol.2019.00038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/31/2019] [Indexed: 01/18/2023] Open
Abstract
Associative long-term memories (LTMs) support long-lasting behavioral changes resulting from sensory experiences. Retrieval of a stable LTM by means of a large number of conditioned stimulus (CS) alone presentations produces inhibition of the original memory through extinction. Currently, there are two opposing hypotheses to account for the neural mechanisms supporting extinction. The unlearning hypothesis posits that extinction affects the original memory trace by reverting the synaptic changes supporting LTM. On the contrary, the new learning hypothesis proposes that extinction is simply the formation of a new associative memory that inhibits the expression of the original one. We propose that detailed analysis of extinction-associated molecular mechanisms could help distinguish between these hypotheses. Here we will review experimental evidence regarding the role of protein kinases and phosphatases (K&P) on LTM formation and extinction. Even though K&P regulate both memory processes, their participation appears to be dissociated. LTM formation recruits kinases, but is constrained by phosphatases. Memory extinction presents a more diverse molecular landscape, requiring phosphatases and some kinases, but also being constrained by kinase activity. Based on the available evidence, we propose a new theoretical model for memory extinction: a neuronal segregation of K&P supports a combination of time-dependent reversible inhibition of the original memory [CS-unconditioned stimulus (US)], with establishment of a new associative memory trace (CS-noUS).
Collapse
Affiliation(s)
- Mario Rafael Pagani
- Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)-Houssay, Facultad de Medicina, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Emiliano Merlo
- Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)-Houssay, Facultad de Medicina, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.,Department of Psychology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
29
|
Lee JLC, Amorim FE, Cassini LF, Amaral OB. Different temporal windows for CB1 receptor involvement in contextual fear memory destabilisation in the amygdala and hippocampus. PLoS One 2019; 14:e0205781. [PMID: 30645588 PMCID: PMC6333379 DOI: 10.1371/journal.pone.0205781] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/26/2018] [Indexed: 12/24/2022] Open
Abstract
Reconsolidation is a process in which re-exposure to a reminder causes a previously acquired memory to undergo a process of destabilisation followed by subsequent restabilisation. Different molecular mechanisms have been postulated for destabilisation in the amygdala and hippocampus, including CB1 receptor activation, protein degradation and AMPA receptor exchange; however, most of the amygdala studies have used pre-reexposure interventions, while those in the hippocampus have usually performed them after reexposure. To test whether the temporal window for destabilisation is similar across both structures, we trained Lister Hooded rats in a contextual fear conditioning task, and 1 day later performed memory reexposure followed by injection of either the NMDA antagonist MK-801 (0.1 mg/kg) or saline in order to block reconsolidation. In parallel, we also performed local injections of either the CB1 antagonist SR141716A or its vehicle in the hippocampus or in the amygdala, either immediately before or immediately after reactivation. Infusion of SR141716A in the hippocampus prevented the reconsolidation-blocking effect of MK-801 when performed after reexposure, but not before it. In the amygdala, meanwhile, pre-reexposure infusions of SR141716A impaired reconsolidation blockade by MK-801, although the time-dependency of this effect was not as clear as in the hippocampus. Our results suggest the temporal windows for CB1-receptor-mediated memory destabilisation during reconsolidation vary between brain structures. Whether this reflects different time windows for engagement of these structures or different roles played by CB1 receptors in destabilisation across structures remains an open question for future studies.
Collapse
MESH Headings
- Amygdala/drug effects
- Amygdala/physiology
- Animals
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Cannabinoid Receptor Antagonists/administration & dosage
- Conditioning, Classical/drug effects
- Dizocilpine Maleate/administration & dosage
- Excitatory Amino Acid Antagonists/administration & dosage
- Fear/drug effects
- Fear/physiology
- Hippocampus/drug effects
- Hippocampus/physiology
- Male
- Memory/drug effects
- Memory/physiology
- Models, Animal
- Rats
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, AMPA/antagonists & inhibitors
- Receptors, AMPA/metabolism
- Rimonabant/administration & dosage
- Time Factors
Collapse
Affiliation(s)
- Jonathan L. C. Lee
- University of Birmingham, School of Psychology, Edgbaston, Birmingham, United Kingdom
- * E-mail:
| | - Felippe E. Amorim
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lindsey F. Cassini
- University of Birmingham, School of Psychology, Edgbaston, Birmingham, United Kingdom
| | - Olavo B. Amaral
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
30
|
Carew SJ, Mukherjee B, MacIntyre ITK, Ghosh A, Li S, Kirouac GJ, Harley CW, Yuan Q. Pheromone-Induced Odor Associative Fear Learning in Rats. Sci Rep 2018; 8:17701. [PMID: 30532054 PMCID: PMC6286391 DOI: 10.1038/s41598-018-36023-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/14/2018] [Indexed: 12/02/2022] Open
Abstract
Alarm pheromones alert conspecifics to the presence of danger. Can pheromone communication aid in learning specific cues? Such facilitation has an evident evolutionary advantage. We use two associative learning paradigms to test this hypothesis. The first is stressed cage mate-induced conditioning. One pair-housed adult rat received 4 pairings of terpinene + shock over 30 min. Ten minutes after return to the home cage, its companion rat was removed and exposed to terpinene. Single-housed controls were exposed to either terpinene or shock only. Companion rats showed terpinene-specific freezing, which was prevented by β-adrenoceptor blockade. Using Arc to index neuronal activation in response to terpinene re-exposure, stressed cage-mate induced associative learning was measured. Companion rats showed increased neuronal activity in the accessory olfactory bulb, while terpinene + shock-conditioned rats showed increased activity in the main olfactory bulb. Both groups had enhanced activity in the anterior basolateral amygdala and central amygdala. To test involvement of pheromone mediation, in the 2nd paradigm, we paired terpinene with soiled bedding from odor + shock rats or a rat alarm pheromone. Both conditioning increased rats’ freezing to terpinene. Blocking NMDA receptors in the basolateral amygdala prevented odor-specific learning suggesting shock and pheromone-paired pathways converge in the amygdala. An alarm pheromone thus enables cue-specific learning as well as signalling danger.
Collapse
Affiliation(s)
- Samantha J Carew
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, A1B 3V6, Canada
| | - Bandhan Mukherjee
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, A1B 3V6, Canada
| | - Iain T K MacIntyre
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, A1B 3V6, Canada
| | - Abhinaba Ghosh
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, A1B 3V6, Canada
| | - Sa Li
- Department of Oral Biology and Psychiatry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E 0W2, Canada
| | - Gilbert J Kirouac
- Department of Oral Biology and Psychiatry, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, R3E 0W2, Canada
| | - Carolyn W Harley
- Psychology Department, Faculty of Science, Memorial University of Newfoundland, St. John's, A1B 3X9, Canada
| | - Qi Yuan
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, A1B 3V6, Canada.
| |
Collapse
|
31
|
Ventral CA3 Activation Mediates Prophylactic Ketamine Efficacy Against Stress-Induced Depressive-like Behavior. Biol Psychiatry 2018; 84:846-856. [PMID: 29615190 PMCID: PMC6107435 DOI: 10.1016/j.biopsych.2018.02.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/29/2018] [Accepted: 02/13/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND We previously reported that a single injection of ketamine prior to stress protects against the onset of depressive-like behavior and attenuates learned fear. However, the molecular pathways and brain circuits underlying ketamine-induced stress resilience are still largely unknown. METHODS Here, we tested whether prophylactic ketamine administration altered neural activity in the prefrontal cortex and/or hippocampus. Mice were injected with saline or ketamine (30 mg/kg) 1 week before social defeat. Following behavioral tests assessing depressive-like behavior, mice were sacrificed and brains were processed to quantify ΔFosB expression. In a second set of experiments, mice were stereotaxically injected with viral vectors into ventral CA3 (vCA3) in order to silence or overexpress ΔFosB prior to prophylactic ketamine administration. In a third set of experiments, ArcCreERT2 mice, a line that allows for the indelible labeling of neural ensembles activated by a single experience, were used to quantify memory traces representing a contextual fear conditioning experience following prophylactic ketamine administration. RESULTS Prophylactic ketamine administration increased ΔFosB expression in the ventral dentate gyrus and vCA3 of social defeat mice but not of control mice. Transcriptional silencing of ΔFosB activity in vCA3 inhibited prophylactic ketamine efficacy, while overexpression of ΔFosB mimicked and occluded ketamine's prophylactic effects. In ArcCreERT2 mice, ketamine administration altered memory traces representing the contextual fear conditioning experience in vCA3 but not in the ventral dentate gyrus. CONCLUSIONS Our data indicate that prophylactic ketamine may be protective against a stressor by altering neural activity, specifically the neural ensembles representing an individual stressor in vCA3.
Collapse
|
32
|
Chaaya N, Battle AR, Johnson LR. An update on contextual fear memory mechanisms: Transition between Amygdala and Hippocampus. Neurosci Biobehav Rev 2018; 92:43-54. [DOI: 10.1016/j.neubiorev.2018.05.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 03/02/2018] [Accepted: 05/08/2018] [Indexed: 12/27/2022]
|
33
|
Weston CSE. Amygdala Represents Diverse Forms of Intangible Knowledge, That Illuminate Social Processing and Major Clinical Disorders. Front Hum Neurosci 2018; 12:336. [PMID: 30186129 PMCID: PMC6113401 DOI: 10.3389/fnhum.2018.00336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 08/02/2018] [Indexed: 01/21/2023] Open
Abstract
Amygdala is an intensively researched brain structure involved in social processing and multiple major clinical disorders, but its functions are not well understood. The functions of a brain structure are best hypothesized on the basis of neuroanatomical connectivity findings, and of behavioral, neuroimaging, neuropsychological and physiological findings. Among the heaviest neuroanatomical interconnections of amygdala are those with perirhinal cortex (PRC), but these are little considered in the theoretical literature. PRC integrates complex, multimodal, meaningful and fine-grained distributed representations of objects and conspecifics. Consistent with this connectivity, amygdala is hypothesized to contribute meaningful and fine-grained representations of intangible knowledge for integration by PRC. Behavioral, neuroimaging, neuropsychological and physiological findings further support amygdala mediation of a diversity of such representations. These representations include subjective valence, impact, economic value, noxiousness, importance, ingroup membership, social status, popularity, trustworthiness and moral features. Further, the formation of amygdala representations is little understood, and is proposed to be often implemented through embodied cognition mechanisms. The hypothesis builds on earlier work, and makes multiple novel contributions to the literature. It highlights intangible knowledge, which is an influential but insufficiently researched factor in social and other behaviors. It contributes to understanding the heavy but neglected amygdala-PRC interconnections, and the diversity of amygdala-mediated intangible knowledge representations. Amygdala is a social brain region, but it does not represent species-typical social behaviors. A novel proposal to clarify its role is postulated. The hypothesis is also suggested to illuminate amygdala's involvement in several core symptoms of autism spectrum disorder (ASD). Specifically, novel and testable explanations are proposed for the ASD symptoms of disorganized visual scanpaths, apparent social disinterest, preference for concrete cognition, aspects of the disorder's heterogeneity, and impairment in some activities of daily living. Together, the presented hypothesis demonstrates substantial explanatory potential in the neuroscience, social and clinical domains.
Collapse
|
34
|
Miller LA, Heroux NA, Stanton ME. Differential involvement of amygdalar NMDA receptors across variants of contextual fear conditioning in adolescent rats. Behav Brain Res 2018; 356:236-242. [PMID: 30142395 DOI: 10.1016/j.bbr.2018.08.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 07/30/2018] [Accepted: 08/14/2018] [Indexed: 11/30/2022]
Abstract
In standard contextual fear conditioning (sCFC), learning of the context and formation of the context-shock association occur in the same training session whereas in the context preexposure facilitation effect (CPFE) learning the context (preexposure) and the context-shock association (training) are separated by 24 h. In both procedures conditioned freezing can be measured immediately (post-shock test) or during a 24-hour retention test. In adult rats, disrupting basolateral amygdala (BLA) activity or plasticity during training on sCFC impairs both post-shock and retention freezing [Maren et al, 1996; 1]. This manipulation on the training day of the CPFE disrupts retention freezing but effects on post-shock freezing are unknown [Matus-Amat et al, 2007; 2]. Experiment 1 extended this literature from adult to adolescent rats and to the role of BLA activity and plasticity in post-shock freezing during the CPFE. Intra-BLA infusions of muscimol prior to the training day of the CPFE disrupted both post-shock and retention freezing in Postnatal Day (PD) 31-33 rats. In the second two experiments, intra-BLA infusions of APV prior to the training day of sCFC disrupted retention but not post-shock freezing, while infusions of APV prior to training of the CPFE disrupt both post-shock and retention freezing. Our findings suggest that the BLA plasticity plays a different role in the CPFE vs. sCFC. Its role in the CPFE is similar in both adolescent and adult rats, while the role of the BLA in post-shock freezing during sCFC may differ across age or across studies that employ different procedures or parameters.
Collapse
|
35
|
Sprowles JLN, Amos-Kroohs RM, Braun AA, Sugimoto C, Vorhees CV, Williams MT. Developmental manganese, lead, and barren cage exposure have adverse long-term neurocognitive, behavioral and monoamine effects in Sprague-Dawley rats. Neurotoxicol Teratol 2018; 67:50-64. [PMID: 29631003 DOI: 10.1016/j.ntt.2018.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 03/30/2018] [Accepted: 04/04/2018] [Indexed: 12/17/2022]
Abstract
Developmental stress, including low socioeconomic status (SES), can induce dysregulation of the hypothalamic-pituitary-adrenal axis and result in long-term changes in stress reactivity. Children in lower SES households experience more stress and are more likely to be exposed to environmental neurotoxins such as lead (Pb) and manganese (Mn) than children in higher SES households. Co-exposure to stress, Pb, and Mn during early development may increase the risk of central nervous system dysfunction compared with unexposed children. To investigate the potential interaction of these factors, Sprague-Dawley rats were bred, and litters born in-house were culled on postnatal day (P)1 to 6 males and 6 females. One male and female within each litter were assigned to one of the following groups: 0 (vehicle), 10 mg/kg Pb, 100 mg/kg Mn, or 10 mg/kg Pb + 100 mg/kg Mn (PbMn), water gavage, and handled only from P4-28 with half the litters reared in cages with standard bedding (29 litters) and half with no bedding (Barren; 27 litters). Mn and PbMn groups had decreased anxiety, reduced acoustic startle, initial open-field hypoactivity, increased activity following (+)-methamphetamine, deficits in egocentric learning in the Cincinnati water maze (CWM), and deficits in latent inhibition conditioning. Pb increased anxiety and reduced open-field activity. Barren-reared rats had decreased anxiety, CWM deficits, increased startle, and initial open-field hyperactivity. Mn, PbMn, Pb Barren-reared groups had impaired Morris water maze performance. Pb altered neostriatal serotonin and norepinephrine, Mn increased hippocampal serotonin in males, Mn + Barren-rearing increased neostriatal serotonin, and Barren-rearing decreased neostriatal dopamine in males. At the doses used here, most effects were in the Mn and PbMn groups. Few interactions between Mn, Pb, and rearing stress were found, indicating that the interaction of these three variables is not as impactful as hypothesized.
Collapse
Affiliation(s)
- Jenna L N Sprowles
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States; Rhodes College, Department of Psychology, 2000 North Parkway, Memphis, TN 38112, United States.
| | - Robyn M Amos-Kroohs
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States; Virginia Department of Forensic Science, 700 North Fifth St, Richmond, VA 23219, United States
| | - Amanda A Braun
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States
| | - Chiho Sugimoto
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine, Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| |
Collapse
|
36
|
Crestani AP, Sierra RO, Machado A, Haubrich J, Scienza KM, de Oliveira Alvares L, Quillfeldt JA. Hippocampal plasticity mechanisms mediating experience-dependent learning change over time. Neurobiol Learn Mem 2018; 150:56-63. [PMID: 29501525 DOI: 10.1016/j.nlm.2018.02.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 01/02/2018] [Accepted: 02/19/2018] [Indexed: 01/15/2023]
Abstract
The requirement of NMDA receptor (NMDAR) activity for memory formation is well described. However, the plasticity mechanisms for memory can be modified by experience, such that a future similar learning becomes independent of NMDARs. This effect has often been reported in learning events conducted with a few days interval. In this work, we asked whether the NMDAR-independency is permanent or the brain regions and plasticity mechanisms of experience-dependent learning may change over time. Considering that contextual memories undergo a gradual reorganization over time, becoming progressively independent from the hippocampus and dependent upon cortical regions, we investigated the brain regions mediating a new related learning conducted at a remote time-point, when the first memory was already cortically established. First, we demonstrated that anterior cingulate cortex was not able to support a learning subsequent to a previous systems-level consolidated memory; it did require at least one functional subregion of the hippocampus (ventral or dorsal). Moreover, after replicating findings showing that a few days interval between trainings induces a NMDAR-independent learning, we managed to show that a learning following a longer interval once again becomes dependent on NMDARs in the hippocampus. These findings suggest that while the previous memory grows independent from the hippocampus over time, an experience-dependent learning following a systems-consolidated memory once again engages the hippocampus and a NMDAR-dependent plasticity mechanism.
Collapse
Affiliation(s)
- Ana Paula Crestani
- Psychobiology and Neurocomputation Lab, Biophysics Department, Bioscience Institute, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Neurosciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Rodrigo Ordoñez Sierra
- Psychobiology and Neurocomputation Lab, Biophysics Department, Bioscience Institute, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Neurosciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Adriano Machado
- Psychobiology and Neurocomputation Lab, Biophysics Department, Bioscience Institute, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Neurosciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Josué Haubrich
- Psychobiology and Neurocomputation Lab, Biophysics Department, Bioscience Institute, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Neurosciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Krislei Martin Scienza
- Psychobiology and Neurocomputation Lab, Biophysics Department, Bioscience Institute, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Neurosciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Lucas de Oliveira Alvares
- Neurosciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil; Neurobiology of Memory Lab, Biophysics Department, Bioscience Institute, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Jorge Alberto Quillfeldt
- Psychobiology and Neurocomputation Lab, Biophysics Department, Bioscience Institute, Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil; Neurosciences Graduate Program, Federal University of Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
37
|
Beyeler A, Chang CJ, Silvestre M, Lévêque C, Namburi P, Wildes CP, Tye KM. Organization of Valence-Encoding and Projection-Defined Neurons in the Basolateral Amygdala. Cell Rep 2018; 22:905-918. [PMID: 29386133 PMCID: PMC5891824 DOI: 10.1016/j.celrep.2017.12.097] [Citation(s) in RCA: 187] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/30/2017] [Accepted: 12/26/2017] [Indexed: 01/03/2023] Open
Abstract
The basolateral amygdala (BLA) mediates associative learning for both
fear and reward. Accumulating evidence supports the notion that different BLA
projections distinctly alter motivated behavior, including projections to the
nucleus accumbens (NAc), medial aspect of the central amygdala (CeM), and
ventral hippocampus (vHPC). Although there is consensus regarding the existence
of distinct subsets of BLA neurons encoding positive or negative valence,
controversy remains regarding the anatomical arrangement of these populations.
First, we map the location of more than 1,000 neurons distributed across the BLA
and recorded during a Pavlovian discrimination task. Next, we determine the
location of projection-defined neurons labeled with retrograde tracers and use
CLARITY to reveal the axonal path in 3-dimensional space. Finally, we examine
the local influence of each projection-defined populations within the BLA.
Understanding the functional and topographical organization of circuits
underlying valence assignment could reveal fundamental principles about
emotional processing.
Collapse
Affiliation(s)
- Anna Beyeler
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Neurocentre Magendie, INSERM, U1215, University of Bordeaux, 146 rue Léo Saignat, 33077 Bordeaux Cedex, France.
| | - Chia-Jung Chang
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Margaux Silvestre
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Clémentine Lévêque
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Praneeth Namburi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Craig P Wildes
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kay M Tye
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
38
|
Pattwell SS, Bath KG. Emotional learning, stress, and development: An ever-changing landscape shaped by early-life experience. Neurobiol Learn Mem 2017; 143:36-48. [PMID: 28458034 PMCID: PMC5540880 DOI: 10.1016/j.nlm.2017.04.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 12/19/2022]
Abstract
The capacity to learn to associate cues with negative outcomes is a highly adaptive process that appears to be conserved across species. However, when the cue is no longer a valid predictor of danger, but the emotional response persists, this can result in maladaptive behaviors, and in humans contribute to debilitating emotional disorders. Over the past several decades, work in neuroscience, psychiatry, psychology, and biology have uncovered key processes underlying, and structures governing, emotional responding and learning, as well as identified disruptions in the structural and functional integrity of these brain regions in models of pathology. In this review, we highlight some of this elegant body of work as well as incorporate emerging findings from the field of developmental neurobiology to emphasize how development contributes to changes in the ability to learn and express emotional responses, and how early experiences, such as stress, shape the development and functioning of these circuits.
Collapse
Affiliation(s)
- Siobhan S Pattwell
- Department of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109, United States.
| | - Kevin G Bath
- Department of Cognitive, Linguistic, and Psychological Sciences, Brown University, Providence, RI 02912, United States
| |
Collapse
|
39
|
Schaefer N, Rotermund C, Blumrich EM, Lourenco MV, Joshi P, Hegemann RU, Jamwal S, Ali N, García Romero EM, Sharma S, Ghosh S, Sinha JK, Loke H, Jain V, Lepeta K, Salamian A, Sharma M, Golpich M, Nawrotek K, Paidi RK, Shahidzadeh SM, Piermartiri T, Amini E, Pastor V, Wilson Y, Adeniyi PA, Datusalia AK, Vafadari B, Saini V, Suárez-Pozos E, Kushwah N, Fontanet P, Turner AJ. The malleable brain: plasticity of neural circuits and behavior - a review from students to students. J Neurochem 2017. [PMID: 28632905 DOI: 10.1111/jnc.14107] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One of the most intriguing features of the brain is its ability to be malleable, allowing it to adapt continually to changes in the environment. Specific neuronal activity patterns drive long-lasting increases or decreases in the strength of synaptic connections, referred to as long-term potentiation and long-term depression, respectively. Such phenomena have been described in a variety of model organisms, which are used to study molecular, structural, and functional aspects of synaptic plasticity. This review originated from the first International Society for Neurochemistry (ISN) and Journal of Neurochemistry (JNC) Flagship School held in Alpbach, Austria (Sep 2016), and will use its curriculum and discussions as a framework to review some of the current knowledge in the field of synaptic plasticity. First, we describe the role of plasticity during development and the persistent changes of neural circuitry occurring when sensory input is altered during critical developmental stages. We then outline the signaling cascades resulting in the synthesis of new plasticity-related proteins, which ultimately enable sustained changes in synaptic strength. Going beyond the traditional understanding of synaptic plasticity conceptualized by long-term potentiation and long-term depression, we discuss system-wide modifications and recently unveiled homeostatic mechanisms, such as synaptic scaling. Finally, we describe the neural circuits and synaptic plasticity mechanisms driving associative memory and motor learning. Evidence summarized in this review provides a current view of synaptic plasticity in its various forms, offers new insights into the underlying mechanisms and behavioral relevance, and provides directions for future research in the field of synaptic plasticity. Read the Editorial Highlight for this article on page 788. Cover Image for this issue: doi: 10.1111/jnc.13815.
Collapse
Affiliation(s)
- Natascha Schaefer
- Institute for Clinical Neurobiology, Julius-Maximilians-University of Wuerzburg, Würzburg, Germany
| | - Carola Rotermund
- German Center of Neurodegenerative Diseases, University of Tuebingen, Tuebingen, Germany
| | - Eva-Maria Blumrich
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, Bremen, Germany.,Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pooja Joshi
- Inserm UMR 1141, Robert Debre Hospital, Paris, France
| | - Regina U Hegemann
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - Sumit Jamwal
- Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Nilufar Ali
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | | | - Sorabh Sharma
- Neuropharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Rajasthan, India
| | - Shampa Ghosh
- National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Tarnaka, Hyderabad, India
| | - Jitendra K Sinha
- National Institute of Nutrition (NIN), Indian Council of Medical Research (ICMR), Tarnaka, Hyderabad, India
| | - Hannah Loke
- Hudson Institute of Medical Research, Melbourne, Victoria, Australia.,Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Vishal Jain
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Katarzyna Lepeta
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ahmad Salamian
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mahima Sharma
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mojtaba Golpich
- Department of Medicine, University Kebangsaan Malaysia Medical Centre (HUKM), Cheras, Kuala Lumpur, Malaysia
| | - Katarzyna Nawrotek
- Department of Process Thermodynamics, Faculty of Process and Environmental Engineering, Lodz University of Technology, Lodz, Poland
| | - Ramesh K Paidi
- CSIR-Indian Institute of Chemical Biology, Jadavpur, Kolkata, India
| | - Sheila M Shahidzadeh
- Department of Biology, Program in Neuroscience, Syracuse University, Syracuse, New York, USA
| | - Tetsade Piermartiri
- Programa de Pós-Graduação em Neurociências, Universidade Federal de Santa Catarina (UFSC), Florianópolis, Brazil
| | - Elham Amini
- Department of Medicine, University Kebangsaan Malaysia Medical Centre (HUKM), Cheras, Kuala Lumpur, Malaysia
| | - Veronica Pastor
- Instituto de Biología Celular y Neurociencia Prof. Eduardo De Robertis, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Yvette Wilson
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| | - Philip A Adeniyi
- Cell Biology and Neurotoxicity Unit, Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University, Ado - Ekiti, Ekiti State, Nigeria
| | | | - Benham Vafadari
- Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Vedangana Saini
- Department of Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Edna Suárez-Pozos
- Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Toxicología, México
| | - Neetu Kushwah
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Paula Fontanet
- Division of Molecular and Cellular Neuroscience, Institute of Cellular Biology and Neuroscience (IBCN), CONICET-UBA, School of Medicine, Buenos Aires, Argentina
| | - Anthony J Turner
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
40
|
Harnett NG, Wood KH, Ference EW, Reid MA, Lahti AC, Knight AJ, Knight DC. Glutamate/glutamine concentrations in the dorsal anterior cingulate vary with Post-Traumatic Stress Disorder symptoms. J Psychiatr Res 2017; 91:169-176. [PMID: 28478230 DOI: 10.1016/j.jpsychires.2017.04.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/24/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023]
Abstract
Trauma and stress-related disorders (e.g., Acute Stress Disorder; ASD and Post-Traumatic Stress Disorder; PTSD) that develop following a traumatic event are characterized by cognitive-affective dysfunction. The cognitive and affective functions disrupted by stress disorder are mediated, in part, by glutamatergic neural systems. However, it remains unclear whether neural glutamate concentrations, measured acutely following trauma, vary with ASD symptoms and/or future PTSD symptom expression. Therefore, the current study utilized proton magnetic resonance spectroscopy (1H-MRS) to investigate glutamate/glutamine (Glx) concentrations within the dorsal anterior cingulate cortex (ACC) of recently (i.e., within one month) traumatized individuals and non-traumatized controls. Although Glx concentrations within dorsal ACC did not differ between recently traumatized and non-traumatized control groups, a positive linear relationship was observed between Glx concentrations and current stress disorder symptoms in traumatized individuals. Further, Glx concentrations showed a positive linear relationship with future stress disorder symptoms (i.e., assessed 3 months post-trauma). The present results suggest glutamate concentrations may play a role in both acute and future post-traumatic stress symptoms following a traumatic experience. The current results expand our understanding of the neurobiology of stress disorder and suggest glutamate within the dorsal ACC plays an important role in cognitive-affective dysfunction following a traumatic experience.
Collapse
Affiliation(s)
- Nathaniel G Harnett
- Department of Psychology, University of Alabama at Birmingham, United States
| | - Kimberly H Wood
- Department of Psychology, University of Alabama at Birmingham, United States
| | - Edward W Ference
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, United States
| | - Meredith A Reid
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, United States; Department of Electrical and Computer Engineering, Auburn University, United States
| | - Adrienne C Lahti
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, United States
| | - Amy J Knight
- Department of Physical Medicine and Rehabilitation, University of Alabama at Birmingham, United States
| | - David C Knight
- Department of Psychology, University of Alabama at Birmingham, United States.
| |
Collapse
|
41
|
Functional Heterogeneity in the Bed Nucleus of the Stria Terminalis. J Neurosci 2017; 36:8038-49. [PMID: 27488624 DOI: 10.1523/jneurosci.0856-16.2016] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/05/2016] [Indexed: 11/21/2022] Open
Abstract
Early work stressed the differing involvement of the central amygdala (CeA) and bed nucleus of the stria terminalis (BNST) in the genesis of fear versus anxiety, respectively. In 2009, Walker, Miles, and Davis proposed a model of amygdala-BNST interactions to explain these functional differences. This model became extremely influential and now guides a new wave of studies on the role of BNST in humans. Here, we consider evidence for and against this model, in the process highlighting central principles of BNST organization. This analysis leads us to conclude that BNST's influence is not limited to the generation of anxiety-like responses to diffuse threats, but that it also shapes the impact of discrete threatening stimuli. It is likely that BNST-CeA interactions are involved in modulating responses to such threats. In addition, whereas current views emphasize the contributions of the anterolateral BNST region in anxiety, accumulating data indicate that the anteromedial and anteroventral regions also play a critical role. The presence of multiple functional subregions within the small volume of BNST raises significant technical obstacles for functional imaging studies in humans.
Collapse
|
42
|
McGowan JC, LaGamma CT, Lim SC, Tsitsiklis M, Neria Y, Brachman RA, Denny CA. Prophylactic Ketamine Attenuates Learned Fear. Neuropsychopharmacology 2017; 42:1577-1589. [PMID: 28128336 PMCID: PMC5518899 DOI: 10.1038/npp.2017.19] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 01/13/2017] [Accepted: 01/18/2017] [Indexed: 12/19/2022]
Abstract
Ketamine has been reported to be an efficacious antidepressant for major depressive disorder and posttraumatic stress disorder. Most recently, ketamine has also been shown to be prophylactic against stress-induced depressive-like behavior in mice. It remains unknown, however, when ketamine should be administered relative to a stressor in order to maximize its antidepressant and/or prophylactic effects. Moreover, it is unknown whether ketamine can be prophylactic against subsequent stressors. We systematically administered ketamine at different time points relative to a fear experience, in order to determine when ketamine is most effective at reducing fear expression or preventing fear reactivation. Using a contextual fear conditioning (CFC) paradigm, mice were administered a single dose of saline or ketamine (30 mg/kg) at varying time points before or after CFC. Mice administered prophylactic ketamine 1 week, but not 1 month or 1 h before CFC, exhibited reduced freezing behavior when compared with mice administered saline. In contrast, ketamine administration following CFC or during extinction did not alter subsequent fear expression. However, ketamine administered before reinstatement increased the number of rearing bouts in an open field, possibly suggesting an increase in attentiveness. These data indicate that ketamine can buffer a fear response when given a week before as prophylactic, but not when given immediately before or after a stress-inducing episode. Thus, ketamine may be most useful in the clinic if administered in a prophylactic manner 1 week before a stressor, in order to protect against heightened fear responses to aversive stimuli.
Collapse
Affiliation(s)
- Josephine C McGowan
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, USA,Barnard College of Columbia University, New York, NY, USA
| | - Christina T LaGamma
- Barnard College of Columbia University, New York, NY, USA,Division of Integrative Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, USA
| | - Sean C Lim
- Division of Integrative Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, USA
| | - Melina Tsitsiklis
- Doctoral Program in Neurobiology and Behavior, Columbia University, New York, NY, USA
| | - Yuval Neria
- Department of Psychiatry, Columbia University, NYSPI Kolb Research Annex, New York, NY, USA,Department of Epidemiology, Columbia University, New York, NY, USA
| | - Rebecca A Brachman
- Department of Psychiatry, Columbia University, NYSPI Kolb Research Annex, New York, NY, USA
| | - Christine A Denny
- Division of Integrative Neuroscience, Research Foundation for Mental Hygiene, Inc. (RFMH)/New York State Psychiatric Institute (NYSPI), New York, NY, USA,Department of Psychiatry, Columbia University, NYSPI Kolb Research Annex, New York, NY, USA,Department of Psychiatry, Columbia University, NYSPI Kolb Research Annex, Room 777, 1051 Riverside Drive, Unit 87, New York, NY 10032, USA, Tel: +1 646 774 7100, Fax: +1 646 774 7102, E-mail:
| |
Collapse
|
43
|
Coimbra NC, Calvo F, Almada RC, Freitas RL, Paschoalin-Maurin T, dos Anjos-Garcia T, Elias-Filho DH, Ubiali WA, Lobão-Soares B, Tracey I. Opioid neurotransmission modulates defensive behavior and fear-induced antinociception in dangerous environments. Neuroscience 2017; 354:178-195. [DOI: 10.1016/j.neuroscience.2017.04.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 10/19/2022]
|
44
|
Mikics E, Toth M, Biro L, Bruzsik B, Nagy B, Haller J. The role of GluN2B-containing NMDA receptors in short- and long-term fear recall. Physiol Behav 2017; 177:44-48. [PMID: 28400283 DOI: 10.1016/j.physbeh.2017.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/24/2017] [Accepted: 04/07/2017] [Indexed: 12/27/2022]
Abstract
N-methyl-d-aspartate (NMDA) receptors are crucial synaptic elements in long-term memory formation, including the associative learning of fearful events. Although NMDA blockers were consistently shown to inhibit fear memory acquisition and recall, the clinical use of general NMDA blockers is hampered by their side effects. Recent studies revealed significant heterogeneity in the distribution and neurophysiological characteristics of NMDA receptors with different GluN2 (NR2) subunit composition, which may have differential role in fear learning and recall. To investigate the specific role of NMDA receptor subpopulations with different GluN2 subunit compositions in the formation of lasting traumatic memories, we contrasted the effects of general NMDA receptor blockade with GluN2A-, GluN2B-, and GluN2C/D subunit selective antagonists (MK-801, PEAQX, Ro25-6981, PPDA, respectively). To investigate acute and lasting consequences, behavioral responses were investigated 1 and 28days after fear conditioning. We found that MK-801 (0.05 and 0.1mg/kg) decreased fear recall at both time points. GluN2B receptor subunit blockade produced highly similar effects, albeit efficacy was somewhat smaller 28days after fear conditioning. Unlike MK-801, Ro25-6981 (3 and 10mg/kg) did not affect locomotor activity in the open-field. In contrast, GluN2A and GluN2C/D blockers (6 and 20mg/kg PEAQX; 3 and 10mg/kg PPDA, respectively) had no effect on conditioned fear recall at any time point and dose. This sharp contrast between GluN2B- and other subunit-containing NMDA receptor function indicates that GluN2B receptor subunits are intimately involved in fear memory formation, and may provide a novel pharmacological target in post-traumatic stress disorder or other fear-related disorders.
Collapse
Affiliation(s)
- Eva Mikics
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary.
| | - Mate Toth
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| | - Laszlo Biro
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| | - Biborka Bruzsik
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| | - Boglarka Nagy
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| | - Jozsef Haller
- Institute of Experimental Medicine, Department of Behavioral Neuroscience, P.O. Box 67, H-1450 Budapest, Hungary
| |
Collapse
|
45
|
Paré D, Quirk GJ. WHEN SCIENTIFIC PARADIGMS LEAD TO TUNNEL VISION: LESSONS FROM THE STUDY OF FEAR. NPJ SCIENCE OF LEARNING 2017; 2:6. [PMID: 30294453 PMCID: PMC6171770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/07/2017] [Accepted: 02/21/2017] [Indexed: 10/05/2023]
Abstract
For the past 30 years, research on the amygdala has largely focused on the genesis of defensive behaviors as its main function. This focus originated from early lesion studies and was supported by extensive anatomical, physiological, and pharmacological data. Here we argue that while much data is consistent with the fear model of amygdala function, it has never been directly tested, in part due to overreliance on the fear conditioning task. In support of the fear model, amygdala neurons appear to signal threats and/or stimuli predictive of threats. However, recent studies in a natural threat setting show that amygdala activity does not correlate with threats, but simply with the movement of the rat, independent of valence. This was true for both natural threats as well as conditioned stimuli; indeed there was no evidence of threat signaling in amygdala neurons. Similar findings are emerging for prefrontal neurons that modulate the amygdala. These recent developments lead us to propose a new conceptualization of amygdala function whereby the amygdala inhibits behavioral engagement. Moreover, we propose that the goal of understanding the amygdala will be best served by shifting away from fear conditioning toward naturalistic approach and avoidance paradigms that involve decision making and a larger repertoire of spontaneous and learned behaviors, all the while keeping an open mind.
Collapse
Affiliation(s)
- Denis Paré
- Center for Molecular and Behavioral Neuroscience, Rutgers State University, Newark, NJ 07102 USA
| | - Gregory J. Quirk
- University of Puerto Rico School of Medicine, San Juan, PR 00936-5067 USA
| |
Collapse
|
46
|
Paré D, Quirk GJ. WHEN SCIENTIFIC PARADIGMS LEAD TO TUNNEL VISION: LESSONS FROM THE STUDY OF FEAR. NPJ SCIENCE OF LEARNING 2017; 2:6. [PMID: 30294453 PMCID: PMC6171770 DOI: 10.1038/s41539-017-0007-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/07/2017] [Accepted: 02/21/2017] [Indexed: 06/08/2023]
Abstract
For the past 30 years, research on the amygdala has largely focused on the genesis of defensive behaviors as its main function. This focus originated from early lesion studies and was supported by extensive anatomical, physiological, and pharmacological data. Here we argue that while much data is consistent with the fear model of amygdala function, it has never been directly tested, in part due to overreliance on the fear conditioning task. In support of the fear model, amygdala neurons appear to signal threats and/or stimuli predictive of threats. However, recent studies in a natural threat setting show that amygdala activity does not correlate with threats, but simply with the movement of the rat, independent of valence. This was true for both natural threats as well as conditioned stimuli; indeed there was no evidence of threat signaling in amygdala neurons. Similar findings are emerging for prefrontal neurons that modulate the amygdala. These recent developments lead us to propose a new conceptualization of amygdala function whereby the amygdala inhibits behavioral engagement. Moreover, we propose that the goal of understanding the amygdala will be best served by shifting away from fear conditioning toward naturalistic approach and avoidance paradigms that involve decision making and a larger repertoire of spontaneous and learned behaviors, all the while keeping an open mind.
Collapse
Affiliation(s)
- Denis Paré
- Center for Molecular and Behavioral Neuroscience, Rutgers State University, Newark, NJ 07102 USA
| | - Gregory J. Quirk
- University of Puerto Rico School of Medicine, San Juan, PR 00936-5067 USA
| |
Collapse
|
47
|
Acca GM, Mathew AS, Jin J, Maren S, Nagaya N. Allopregnanolone induces state-dependent fear via the bed nucleus of the stria terminalis. Horm Behav 2017; 89:137-144. [PMID: 28104355 PMCID: PMC5381271 DOI: 10.1016/j.yhbeh.2017.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/09/2017] [Accepted: 01/15/2017] [Indexed: 12/22/2022]
Abstract
Gonadal steroids and their metabolites have been shown to be important modulators of emotional behavior. Allopregnanolone (ALLO), for example, is a metabolite of progesterone that has been linked to anxiety-related disorders such as posttraumatic stress disorder. In rodents, it has been shown to reduce anxiety in a number of behavioral paradigms including Pavlovian fear conditioning. We have recently found that expression of conditioned contextual (but not auditory) freezing in rats can be suppressed by infusion of ALLO into the bed nucleus of the stria terminalis (BNST). To further explore the nature of this effect, we infused ALLO into the BNST of male rats prior to both conditioning and testing. We found that suppression of contextual fear occurred when the hormone was present during either conditioning or testing but not during both procedures, suggesting that ALLO acts in a state-dependent manner within the BNST. A shift in interoceptive context during testing for animals conditioned under ALLO provided further support for this mechanism of hormonal action on contextual fear. Interestingly, infusions of ALLO into the basolateral amygdala produced a state-independent suppression of both conditioned contextual and auditory freezing. Altogether, these results suggest that ALLO can influence the acquisition and expression of fear memories by both state-dependent and state-independent mechanisms.
Collapse
Affiliation(s)
- Gillian M Acca
- Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - Abel S Mathew
- Department of Psychology, Texas A&M University, College Station, TX, USA
| | - Jingji Jin
- Institute for Neuroscience, Texas A&M University, College Station, TX, USA
| | - Stephen Maren
- Institute for Neuroscience, Texas A&M University, College Station, TX, USA; Department of Psychology, Texas A&M University, College Station, TX, USA
| | - Naomi Nagaya
- Institute for Neuroscience, Texas A&M University, College Station, TX, USA; Department of Psychology, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
48
|
Jasnow AM, Lynch JF, Gilman TL, Riccio DC. Perspectives on fear generalization and its implications for emotional disorders. J Neurosci Res 2016; 95:821-835. [PMID: 27448175 DOI: 10.1002/jnr.23837] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 12/28/2022]
Abstract
Although generalization to conditioned stimuli is not a new phenomenon, renewed interest in understanding its biological underpinning has stemmed from its association with a number of anxiety disorders. Generalization as it relates to fear processing is a temporally dynamic process in which animals, including humans, display fear in response to similar yet distinct cues or contexts as the time between training and testing increases. This Review surveys the literature on contextual fear generalization and its relation to several views of memory, including systems consolidation, forgetting, and transformation hypothesis, which differentially implicate roles of the hippocampus and neocortex in memory consolidation and retrieval. We discuss recent evidence on the neurobiological mechanisms contributing to the increase in fear generalization over time and how generalized responding may be modulated by acquisition, consolidation, and retrieval mechanisms. Whereas clinical perspectives of generalization emphasize a lack of fear inhibition to CS- cues or fear toward intermediate CS cues, the time-dependent nature of generalization and its relation to traditional views on memory consolidation and retrieval are often overlooked. Understanding the time-dependent increase in fear generalization has important implications not only for understanding how generalization contributes to anxiety disorders but also for understanding basic long-term memory function. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Aaron M Jasnow
- Department of Psychological Sciences, Kent State University, Kent, Ohio
| | - Joseph F Lynch
- Department of Psychological Sciences, Kent State University, Kent, Ohio
| | - T Lee Gilman
- Department of Psychological Sciences, Kent State University, Kent, Ohio
| | - David C Riccio
- Department of Psychological Sciences, Kent State University, Kent, Ohio
| |
Collapse
|
49
|
Epinephrine increases contextual learning through activation of peripheral β2-adrenoceptors. Psychopharmacology (Berl) 2016; 233:2099-2108. [PMID: 26935825 DOI: 10.1007/s00213-016-4254-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 02/22/2016] [Indexed: 02/07/2023]
Abstract
RATIONALE Phenylethanolamine-N-methyltransferase knockout (Pnmt-KO) mice are unable to synthesize epinephrine and display reduced contextual fear. However, the precise mechanism responsible for impaired contextual fear learning in these mice is unknown. OBJECTIVES Our aim was to study the mechanism of epinephrine-dependent contextual learning. METHODS Wild-type (WT) or Pnmt-KO (129x1/SvJ) mice were submitted to a fear conditioning test either in the absence or in the presence of epinephrine, isoprenaline (non-selective β-adrenoceptor agonist), fenoterol (selective β2-adrenoceptor agonist), epinephrine plus sotalol (non-selective β-adrenoceptor antagonist), and dobutamine (selective β1-adrenoceptor agonist). Catecholamines were separated by reverse-phase HPLC and quantified by electrochemical detection. Blood glucose was measured by coulometry. RESULTS Re-exposure to shock context induced higher freezing in WT and Pnmt-KO mice treated with epinephrine and fenoterol than in mice treated with vehicle. In addition, freezing response in Pnmt-KO mice was much lower than in WT mice. Freezing induced by epinephrine was blocked by sotalol in Pnmt-KO mice. Epinephrine and fenoterol treatment restored glycemic response in Pnmt-KO mice. Re-exposure to shock context did not induce a significant difference in freezing in Pnmt-KO mice treated with dobutamine and vehicle. CONCLUSIONS Aversive memories are best retained if moderately high plasma epinephrine concentrations occur at the same moment as the aversive stimulus. In addition, epinephrine increases context fear learning by acting on peripheral β2-adrenoceptors, which may induce high levels of blood glucose. Since glucose crosses the blood-brain barrier, it may enhance hippocampal-dependent contextual learning.
Collapse
|
50
|
Dulka BN, Ford EC, Lee MA, Donnell NJ, Goode TD, Prosser R, Cooper MA. Proteolytic cleavage of proBDNF into mature BDNF in the basolateral amygdala is necessary for defeat-induced social avoidance. ACTA ACUST UNITED AC 2016; 23:156-60. [PMID: 26980783 PMCID: PMC4793198 DOI: 10.1101/lm.040253.115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 01/21/2016] [Indexed: 02/02/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is essential for memory processes. The present study tested whether proteolytic cleavage of proBDNF into mature BDNF (mBDNF) within the basolateral amygdala (BLA) regulates the consolidation of defeat-related memories. We found that acute social defeat increases the expression of mBDNF, but not proBDNF, in the BLA/central amygdala. We also showed that blocking plasmin in the BLA with microinjection of α2-antiplasmin immediately following social defeat decreases social avoidance 24 h later. These data suggest the proteolytic cleavage of BDNF in the BLA is necessary for defeat-induced social avoidance.
Collapse
Affiliation(s)
- Brooke N Dulka
- University of Tennessee, Knoxville, Tennessee 37996, USA
| | - Ellen C Ford
- University of Tennessee, Knoxville, Tennessee 37996, USA
| | - Melissa A Lee
- University of Tennessee, Knoxville, Tennessee 37996, USA
| | | | - Travis D Goode
- University of Tennessee, Knoxville, Tennessee 37996, USA
| | | | | |
Collapse
|