1
|
Zhang J, Yang CQ, Liu ZQ, Wu SP, Li ZG, Zhang LM, Fan HW, Guo ZY, Man HY, Li X, Lu YM, Zhu LQ, Liu D. Cpeb1 remodels cell type-specific translational program to promote fear extinction. SCIENCE ADVANCES 2025; 11:eadr8687. [PMID: 39792668 PMCID: PMC11721575 DOI: 10.1126/sciadv.adr8687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
Protein translation is crucial for fear extinction, a process vital for adaptive behavior and mental health, yet the underlying cell-specific mechanisms remain elusive. Using a Tet-On 3G genetic approach, we achieved precise temporal control over protein translation in the infralimbic medial prefrontal cortex (IL) during fear extinction. In addition, our results reveal that the disruption of cytoplasmic polyadenylation element binding protein 1 (Cpeb1) leads to notable alterations in cell type-specific translational programs, thereby affecting fear extinction. Specifically, Cpeb1 deficiency in neurons activates the translation of heterochromatin protein 1 binding protein 3, which enhances microRNA networks, whereas in microglia, it suppresses the translation of chemokine receptor 1 (Cx3cr1), resulting in an aged-like microglial phenotype. These coordinated alterations impair spine formation and plasticity. Our study highlights the critical role of cell type-specific protein translation in fear extinction and provides an insight into therapeutic targets for disorders with extinction deficits.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Chun-Qing Yang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zhi-Qiang Liu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Shi-Ping Wu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zu-Guang Li
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Luo-Man Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450002, China
| | - Hong-Wei Fan
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Zi-Yuan Guo
- Center for Stem Cell and Organoid Medicine (CuSTOM), Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Heng-Ye Man
- Department of Biology, Boston University, Boston, MA 02215, USA
| | - Xiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
- Brain Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
- Medical Research Institute, Wuhan University, Wuhan, Hubei 430030, China
| | - You-Ming Lu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Dan Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
2
|
Furriel BCRS, Furriel GP, Cunha Xavier Pinto M, Lemos RP. Computational modeling of fear and stress responses: validation using consolidated fear and stress protocols. Front Syst Neurosci 2024; 18:1454336. [PMID: 39776892 PMCID: PMC11703847 DOI: 10.3389/fnsys.2024.1454336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025] Open
Abstract
Dysfunction in fear and stress responses is intrinsically linked to various neurological diseases, including anxiety disorders, depression, and Post-Traumatic Stress Disorder. Previous studies using in vivo models with Immediate-Extinction Deficit (IED) and Stress Enhanced Fear Learning (SEFL) protocols have provided valuable insights into these mechanisms and aided the development of new therapeutic approaches. However, assessing these dysfunctions in animal subjects using IED and SEFL protocols can cause significant pain and suffering. To advance the understanding of fear and stress, this study presents a biologically and behaviorally plausible computational architecture that integrates several subregions of key brain structures, such as the amygdala, hippocampus, and medial prefrontal cortex. Additionally, the model incorporates stress hormone curves and employs spiking neural networks with conductance-based integrate-and-fire neurons. The proposed approach was validated using the well-established Contextual Fear Conditioning paradigm and subsequently tested with IED and SEFL protocols. The results confirmed that higher intensity aversive stimuli result in more robust and persistent fear memories, making extinction more challenging. They also underscore the importance of the timing of extinction and the significant influence of stress. To our knowledge, this is the first instance of computational modeling being applied to IED and SEFL protocols. This study validates our computational model's complexity and biological realism in analyzing responses to fear and stress through fear conditioning, IED, and SEFL protocols. Rather than providing new biological insights, the primary contribution of this work lies in its methodological innovation, demonstrating that complex, biologically plausible neural architectures can effectively replicate established findings in fear and stress research. By simulating protocols typically conducted in vivo-often involving significant pain and suffering-in an insilico environment, our model offers a promising tool for studying fear-related mechanisms. These findings support the potential of computational models to reduce the reliance on animal testing while setting the stage for new therapeutic approaches.
Collapse
Affiliation(s)
- Brunna Carolinne Rocha Silva Furriel
- Instituto Federal de Goiás, Goiânia, Brazil
- Universidade Federal de Goias, School of Electrical, Mechanical and Computer Engineering, Goiânia, Brazil
- Imaging Research Center, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | | | - Rodrigo Pinto Lemos
- Universidade Federal de Goias, School of Electrical, Mechanical and Computer Engineering, Goiânia, Brazil
| |
Collapse
|
3
|
Gattuso JJ, Wilson C, Hannan AJ, Renoir T. Psilocybin as a lead candidate molecule in preclinical therapeutic studies of psychiatric disorders: A systematic review. J Neurochem 2024; 168:1687-1720. [PMID: 38019032 DOI: 10.1111/jnc.16017] [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: 09/03/2023] [Revised: 11/02/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023]
Abstract
Psilocybin is the main psychoactive compound found in hallucinogenic/magic mushrooms and can bind to both serotonergic and tropomyosin receptor kinase b (TrkB) receptors. Psilocybin has begun to show efficacy for a range of neuropsychiatric conditions, including treatment-resistant depression and anxiety disorders; however, neurobiological mechanisms are still being elucidated. Clinical research has found that psilocybin can alter functional connectivity patterns in human brains, which is often associated with therapeutic outcomes. However, preclinical research affords the opportunity to assess the potential cellular mechanisms by which psilocybin may exert its therapeutic effects. Preclinical rodent models can also facilitate a more tightly controlled experimental context and minimise placebo effects. Furthermore, where there is a rationale, preclinical researchers can investigate psilocybin administration in neuropsychiatric conditions that have not yet been researched clinically. As a result, we have systematically reviewed the knowledge base, identifying 82 preclinical studies which were screened based on specific criteria. This resulted in the exclusion of 44 articles, with 34 articles being included in the main review and another 2 articles included as Supporting Information materials. We found that psilocybin shows promise as a lead candidate molecule for treating a variety of neuropsychiatric conditions, albeit showing the most efficacy for depression. We discuss the experimental findings, and identify possible mechanisms whereby psilocybin could invoke therapeutic changes. Furthermore, we critically evaluate the between-study heterogeneity and possible future research avenues. Our review suggests that preclinical rodent models can provide valid and translatable tools for researching novel psilocybin-induced molecular and cellular mechanisms, and therapeutic outcomes.
Collapse
Affiliation(s)
- James J Gattuso
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Carey Wilson
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| | - Thibault Renoir
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
- Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Merscher AS, Gamer M. Can I see it in the eyes? An investigation of freezing-like motion patterns in response to avoidable threat. Psychophysiology 2024; 61:e14567. [PMID: 38469631 DOI: 10.1111/psyp.14567] [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: 10/24/2023] [Revised: 02/23/2024] [Accepted: 03/02/2024] [Indexed: 03/13/2024]
Abstract
Freezing is one of the most extensively studied defensive behaviors in rodents. Both reduced body and gaze movements during anticipation of threat also occur in humans and have been discussed as translational indicators of freezing but their relationship remains unclear. We thus set out to elucidate body and eye movements and concomitant autonomic dynamics in anticipation of avoidable threat. Specifically, 50 participants viewed naturalistic pictures that were preceded by a colored fixation cross, signaling them whether to expect an inevitable (shock), no (safety), or a potential shock (flight) that could be avoided by a quick button press. Body sway, eye movements, the heart rate and skin conductance were recorded. We replicated previously described reductions in body sway, gaze dispersion, and the heart rate, and a skin conductance increase in flight trials. Stronger reductions in gaze but not in body sway predicted faster motor reactions on a trial-wise basis, highlighting their functional role in action preparation. We failed to find a trait-like relationship between body and gaze movements across participants, but their temporal profiles were positively related within individuals, suggesting that both metrics partly reflect the same construct. However, future research is desirable to assess these response patterns in naturalistic environments. A more ethological examination of different movement dynamics upon threat would not only warrant better comparability between rodent and human research but also help determine whether and how eye-tracking could be implemented as a proxy for fear-related movements in restricted brain imaging environments.
Collapse
Affiliation(s)
- Alma-Sophia Merscher
- Experimental Clinical Psychology, Department of Psychology, University of Würzburg, Würzburg, Germany
| | - Matthias Gamer
- Experimental Clinical Psychology, Department of Psychology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
5
|
Negrón-Oyarzo I, Dib T, Chacana-Véliz L, López-Quilodrán N, Urrutia-Piñones J. Large-scale coupling of prefrontal activity patterns as a mechanism for cognitive control in health and disease: evidence from rodent models. Front Neural Circuits 2024; 18:1286111. [PMID: 38638163 PMCID: PMC11024307 DOI: 10.3389/fncir.2024.1286111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 03/11/2024] [Indexed: 04/20/2024] Open
Abstract
Cognitive control of behavior is crucial for well-being, as allows subject to adapt to changing environments in a goal-directed way. Changes in cognitive control of behavior is observed during cognitive decline in elderly and in pathological mental conditions. Therefore, the recovery of cognitive control may provide a reliable preventive and therapeutic strategy. However, its neural basis is not completely understood. Cognitive control is supported by the prefrontal cortex, structure that integrates relevant information for the appropriate organization of behavior. At neurophysiological level, it is suggested that cognitive control is supported by local and large-scale synchronization of oscillatory activity patterns and neural spiking activity between the prefrontal cortex and distributed neural networks. In this review, we focus mainly on rodent models approaching the neuronal origin of these prefrontal patterns, and the cognitive and behavioral relevance of its coordination with distributed brain systems. We also examine the relationship between cognitive control and neural activity patterns in the prefrontal cortex, and its role in normal cognitive decline and pathological mental conditions. Finally, based on these body of evidence, we propose a common mechanism that may underlie the impaired cognitive control of behavior.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Tatiana Dib
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Lorena Chacana-Véliz
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Nélida López-Quilodrán
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| | - Jocelyn Urrutia-Piñones
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
- Programa de Doctorado en Ciencias Mención en Neurociencia, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
6
|
Usmani MT, Krattli RP, El-Khatib SM, Le ACD, Smith SM, Baulch JE, Ng DQ, Acharya MM, Chan A. BDNF Augmentation Using Riluzole Reverses Doxorubicin-Induced Decline in Cognitive Function and Neurogenesis. Neurotherapeutics 2023; 20:838-852. [PMID: 36720792 PMCID: PMC10275819 DOI: 10.1007/s13311-022-01339-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2022] [Indexed: 02/02/2023] Open
Abstract
Cancer-related cognitive impairment (CRCI) considerably affects the quality of life of millions of cancer survivors. Brain-derived neurotrophic factor (BDNF) has been shown to promote survival, differentiation, and maintenance of in vivo dentate neurogenesis, and chemotherapy induces a plethora of physiological and cellular alterations, including a decline in neurogenesis and increased neuroinflammation linked with cognitive impairments. In our clinical studies, breast cancer patients treated with doxorubicin (Adriamycin®, ADR) experienced a significant reduction in the blood levels of BDNF that was associated with a higher risk of CRCI. Our past rodent studies in CRCI have also shown a significant reduction in dentate neurogenesis accompanied by cognitive impairment. In this study, using a female mouse model of ADR-induced cognitive decline, we tested the impact of riluzole (RZ), an orally active BDNF-enhancing medication that is FDA-approved for amyotrophic lateral sclerosis. ADR-treated mice receiving RZ in the drinking water for 1 month showed significant improvements in hippocampal-dependent learning and memory function (spatial recognition), fear extinction memory consolidation, and reduced anxiety-like behavior. RZ prevented chemotherapy-induced reductions of BDNF levels in the hippocampus. Importantly, RZ mitigated chemotherapy-induced loss of newly born, immature neurons, dentate neurogenesis, and neuroinflammation. In conclusion, this data provides pre-clinical evidence for a translationally feasible approach to enhance the neuroprotective effects of RZ treatment to prevent CRCI.
Collapse
Affiliation(s)
- Manal T Usmani
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Robert P Krattli
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Sanad M El-Khatib
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Anh C D Le
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA
| | - Sarah M Smith
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA
| | - Janet E Baulch
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA
| | - Ding Quan Ng
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, CA, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, USA.
- Department of Radiation Oncology, School of Medicine, University of California, Irvine, CA, USA.
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, CA, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, CA, USA.
| |
Collapse
|
7
|
The role of estrogen receptor manipulation during traumatic stress on changes in emotional memory induced by traumatic stress. Psychopharmacology (Berl) 2023; 240:1049-1061. [PMID: 36879072 DOI: 10.1007/s00213-023-06342-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/13/2023] [Indexed: 03/08/2023]
Abstract
RATIONALE Traumatic stress leads to persistent fear, which is a core feature of post-traumatic stress disorder (PTSD). Women are more likely than men to develop PTSD after trauma exposure, which suggests women are differentially sensitive to traumatic stress. However, it is unclear how this differential sensitivity manifests. Cyclical changes in vascular estrogen release could be a contributing factor where levels of vascular estrogens (and activation of estrogen receptors) at the time of traumatic stress alter the impact of traumatic stress. METHODS To examine this, we manipulated estrogen receptors at the time of stress and observed the effect this had on fear and extinction memory (within the single prolonged stress (SPS) paradigm) in female rats. In all experiments, freezing and darting were used to measure fear and extinction memory. RESULTS In Experiment 1, SPS enhanced freezing during extinction testing, and this effect was blocked by nuclear estrogen receptor antagonism prior to SPS. In Experiment 2, SPS decreased conditioned freezing during the acquisition and testing of extinction. Administration of 17β-estradiol altered freezing in control and SPS animals during the acquisition of extinction, but this treatment had no effect on freezing during the testing of extinction memory. In all experiments, darting was only observed to footshock onset during fear conditioning. CONCLUSION The results suggest multiple behaviors (or different behavioral paradigms) are needed to characterize the nature of traumatic stress effects on emotional memory in female rats and that nuclear estrogen receptor antagonism prior to SPS blocks SPS effects on emotional memory in female rats.
Collapse
|
8
|
Stress resilience-associated behaviors following predator scent stress are accompanied by upregulated nucleus accumbens mGlu5 transcription in female Sprague Dawley rats. Behav Brain Res 2022; 436:114090. [DOI: 10.1016/j.bbr.2022.114090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 08/19/2022] [Accepted: 08/29/2022] [Indexed: 12/27/2022]
|
9
|
Klein PM, Alaghband Y, Doan NL, Ru N, Drayson OGG, Baulch JE, Kramár EA, Wood MA, Soltesz I, Limoli CL. Acute, Low-Dose Neutron Exposures Adversely Impact Central Nervous System Function. Int J Mol Sci 2021; 22:9020. [PMID: 34445726 PMCID: PMC8396607 DOI: 10.3390/ijms22169020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/17/2021] [Accepted: 08/19/2021] [Indexed: 02/06/2023] Open
Abstract
A recognized risk of long-duration space travel arises from the elevated exposure astronauts face from galactic cosmic radiation (GCR), which is composed of a diverse array of energetic particles. There is now abundant evidence that exposures to many different charged particle GCR components within acute time frames are sufficient to induce central nervous system deficits that span from the molecular to the whole animal behavioral scale. Enhanced spacecraft shielding can lessen exposures to charged particle GCR components, but may conversely elevate neutron radiation levels. We previously observed that space-relevant neutron radiation doses, chronically delivered at dose-rates expected during planned human exploratory missions, can disrupt hippocampal neuronal excitability, perturb network long-term potentiation and negatively impact cognitive behavior. We have now determined that acute exposures to similar low doses (18 cGy) of neutron radiation can also lead to suppressed hippocampal synaptic signaling, as well as decreased learning and memory performance in male mice. Our results demonstrate that similar nervous system hazards arise from neutron irradiation regardless of the exposure time course. While not always in an identical manner, neutron irradiation disrupts many of the same central nervous system elements as acute charged particle GCR exposures. The risks arising from neutron irradiation are therefore important to consider when determining the overall hazards astronauts will face from the space radiation environment.
Collapse
Affiliation(s)
- Peter M. Klein
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (P.M.K.); (I.S.)
| | - Yasaman Alaghband
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA; (Y.A.); (N.-L.D.); (N.R.); (O.G.G.D.); (J.E.B.)
| | - Ngoc-Lien Doan
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA; (Y.A.); (N.-L.D.); (N.R.); (O.G.G.D.); (J.E.B.)
| | - Ning Ru
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA; (Y.A.); (N.-L.D.); (N.R.); (O.G.G.D.); (J.E.B.)
| | - Olivia G. G. Drayson
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA; (Y.A.); (N.-L.D.); (N.R.); (O.G.G.D.); (J.E.B.)
| | - Janet E. Baulch
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA; (Y.A.); (N.-L.D.); (N.R.); (O.G.G.D.); (J.E.B.)
| | - Enikö A. Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA; (E.A.K.); (M.A.W.)
| | - Marcelo A. Wood
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA; (E.A.K.); (M.A.W.)
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA 94305, USA; (P.M.K.); (I.S.)
| | - Charles L. Limoli
- Department of Radiation Oncology, University of California, Irvine, CA 92697, USA; (Y.A.); (N.-L.D.); (N.R.); (O.G.G.D.); (J.E.B.)
| |
Collapse
|
10
|
Ferland-Beckham C, Chaby LE, Daskalakis NP, Knox D, Liberzon I, Lim MM, McIntyre C, Perrine SA, Risbrough VB, Sabban EL, Jeromin A, Haas M. Systematic Review and Methodological Considerations for the Use of Single Prolonged Stress and Fear Extinction Retention in Rodents. Front Behav Neurosci 2021; 15:652636. [PMID: 34054443 PMCID: PMC8162789 DOI: 10.3389/fnbeh.2021.652636] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Posttraumatic stress disorder (PTSD) is a mental health condition triggered by experiencing or witnessing a terrifying event that can lead to lifelong burden that increases mortality and adverse health outcomes. Yet, no new treatments have reached the market in two decades. Thus, screening potential interventions for PTSD is of high priority. Animal models often serve as a critical translational tool to bring new therapeutics from bench to bedside. However, the lack of concordance of some human clinical trial outcomes with preclinical animal efficacy findings has led to a questioning of the methods of how animal studies are conducted and translational validity established. Thus, we conducted a systematic review to determine methodological variability in studies that applied a prominent animal model of trauma-like stress, single prolonged stress (SPS). The SPS model has been utilized to evaluate a myriad of PTSD-relevant outcomes including extinction retention. Rodents exposed to SPS express an extinction retention deficit, a phenotype identified in humans with PTSD, in which fear memory is aberrantly retained after fear memory extinction. The current systematic review examines methodological variation across all phases of the SPS paradigm, as well as strategies for behavioral coding, data processing, statistical approach, and the depiction of data. Solutions for key challenges and sources of variation within these domains are discussed. In response to methodological variation in SPS studies, an expert panel was convened to generate methodological considerations to guide researchers in the application of SPS and the evaluation of extinction retention as a test for a PTSD-like phenotype. Many of these guidelines are applicable to all rodent paradigms developed to model trauma effects or learned fear processes relevant to PTSD, and not limited to SPS. Efforts toward optimizing preclinical model application are essential for enhancing the reproducibility and translational validity of preclinical findings, and should be conducted for all preclinical psychiatric research models.
Collapse
Affiliation(s)
| | - Lauren E Chaby
- Cohen Veterans Bioscience, New York City, NY, United States
| | - Nikolaos P Daskalakis
- Department of Psychiatry, Harvard Medical School, Boston, MA, United States.,McLean Hospital, Belmont, MA, United States
| | - Dayan Knox
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - Israel Liberzon
- Department of Psychiatry, Texas A&M University, Bryan, TX, United States
| | - Miranda M Lim
- Departments of Neurology, Behavioral Neuroscience, Medicine, Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States.,Sleep Disorders Clinic, VA Portland Health Care System, Portland, OR, United States
| | - Christa McIntyre
- Department of Neuroscience, The University of Texas at Dallas, Richardson, TX, United States
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States.,Research Service, John. D. Dingell VA Medical Center, Detroit, MI, United States
| | - Victoria B Risbrough
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, United States.,Center for Excellence in Stress and Mental Health, VA San Diego Healthcare System, San Diego, CA, United States
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, United States
| | | | - Magali Haas
- Cohen Veterans Bioscience, New York City, NY, United States
| |
Collapse
|
11
|
Florido A, Perez-Caballero L, Velasco ER, Molina P, Marin-Blasco I, Andero R. Direct and Indirect Measurements of Sex Hormones in Rodents During Fear Conditioning. Curr Protoc 2021; 1:e102. [PMID: 33950571 DOI: 10.1002/cpz1.102] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Fear conditioning (FC) is a widely accepted tool for the assessment of learning and memory processes in rodents related to normal and dysregulated acquired fear. The study of sex differences in fear learning and memory is vast and currently increasing. Sex hormones have proven to be crucial for fear memory formation in males and females, and several methods have been developed to assess this hormonal state in rats and mice. Herein, we explain a routine FC and extinction protocol, together with the evaluation of sex hormonal state in male and female rodents. We explain three protocols for the evaluation of this hormonal state directly from blood samples extracted during the procedure or indirectly through histological verification of the estrous cycle for females or behavioral assessment of social hierarchies in males. Although females have typically been considered to present great variability in sex hormones, it is highlighted that sex hormone assessment in males is as variable as in females and equally important for fear memory formation. The readout of these protocols has had a great impact on different fields of fear learning and memory study and appears essential when studying FC. The proven interaction with drugs involved in the modulation of these processes makes sex hormone assessment during FC a valuable tool for the development of effective treatments for fear-related disorders in men and women. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Fear conditioning and fear extinction Basic Protocol 2: Blood collection for direct measurement of sex hormone levels in fear conditioning Basic Protocol 3: Indirect measurement of sex hormones in females during fear conditioning Basic Protocol 4: Assessment of dominance status in males before a fear conditioning protocol Support Protocol: Construction of a confrontation tube.
Collapse
Affiliation(s)
- Antonio Florido
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Laura Perez-Caballero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Eric Raúl Velasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Patricia Molina
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Unitat de Fisiologia Animal (Departament de Biologia Cel·lular, Fisiologia i Immunologia), Facultat de Biociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Ignacio Marin-Blasco
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
| | - Raül Andero
- Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Departament de Psicobiologia i de Metodologia de les Ciències de la Salut, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
- Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí (I3PT), Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
- ICREA, Barcelona, Spain
| |
Collapse
|
12
|
Marković V, Vicario CM, Yavari F, Salehinejad MA, Nitsche MA. A Systematic Review on the Effect of Transcranial Direct Current and Magnetic Stimulation on Fear Memory and Extinction. Front Hum Neurosci 2021; 15:655947. [PMID: 33828472 PMCID: PMC8019721 DOI: 10.3389/fnhum.2021.655947] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/25/2021] [Indexed: 12/13/2022] Open
Abstract
Anxiety disorders are among the most prevalent mental disorders. Present treatments such as cognitive behavior therapy and pharmacological treatments show only moderate success, which emphasizes the importance for the development of new treatment protocols. Non-invasive brain stimulation methods such as repetitive transcranial magnetic stimulation (rTMS) and transcranial direct current stimulation (tDCS) have been probed as therapeutic option for anxiety disorders in recent years. Mechanistic information about their mode of action, and most efficient protocols is however limited. Here the fear extinction model can serve as a model of exposure therapies for studying therapeutic mechanisms, and development of appropriate intervention protocols. We systematically reviewed 30 research articles that investigated the impact of rTMS and tDCS on fear memory and extinction in animal models and humans, in clinical and healthy populations. The results of these studies suggest that tDCS and rTMS can be efficient methods to modulate fear memory and extinction. Furthermore, excitability-enhancing stimulation applied over the vmPFC showed the strongest potential to enhance fear extinction. We further discuss factors that determine the efficacy of rTMS and tDCS in the context of the fear extinction model and provide future directions to optimize parameters and protocols of stimulation for research and treatment.
Collapse
Affiliation(s)
- Vuk Marković
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
- International Graduate School of Neuroscience, Ruhr-University-Bochum, Bochum, Germany
| | | | - Fatemeh Yavari
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Mohammad A. Salehinejad
- Department of Psychology and Neurosciences, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Michael A. Nitsche
- International Graduate School of Neuroscience, Ruhr-University-Bochum, Bochum, Germany
- Department of Neurology, University Medical Hospital Bergmannsheil, Bochum, Germany
| |
Collapse
|
13
|
Van Schuerbeek A, Vanderhasselt MA, Baeken C, Pierre A, Smolders I, Van Waes V, De Bundel D. Effects of repeated anodal transcranial direct current stimulation on auditory fear extinction in C57BL/6J mice. Brain Stimul 2021; 14:250-260. [PMID: 33454396 DOI: 10.1016/j.brs.2021.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Trauma-based psychotherapy is a first line treatment for post-traumatic stress disorder (PTSD) but not all patients achieve long-term remission. Transcranial direct current stimulation (tDCS) received considerable attention as a neuromodulation method that may improve trauma-based psychotherapy. OBJECTIVE We explored the effects of repeated anodal tDCS over the prefrontal cortex (PFC) on fear extinction in mice as a preclinical model for trauma-based psychotherapy. METHODS We performed auditory fear conditioning with moderate or high shock intensity on C57BL6/J mice. Next, mice received anodal tDCS (0.2 mA, 20 min) or sham stimulation over the PFC twice daily for five consecutive days. Extinction training was performed by repeatedly exposing mice to the auditory cue the day after the last stimulation session. Early and late retention of extinction were evaluated one day and three weeks after extinction training respectively. RESULTS We observed no significant effect of tDCS on the acquisition or retention of fear extinction in mice subjected to fear conditioning with moderate intensity. However, when the intensity of fear conditioning was high, tDCS significantly lowered freezing during the acquisition of extinction, regardless of the extinction protocol. Moreover, when tDCS was combined with a strong extinction protocol, we also observed a significant improvement of early extinction recall. Finally, we found that tDCS reduced generalized fear induced by contextual cues when the intensity of conditioning is high and extinction training limited. CONCLUSIONS Our data provide a rationale to further explore anodal tDCS over the PFC as potential support for trauma-based psychotherapy for PTSD.
Collapse
Affiliation(s)
- Andries Van Schuerbeek
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Marie-Anne Vanderhasselt
- Department of Experimental Clinical and Health Psychology, Universiteit Gent - C, Heymanslaan 10, 9000, Gent, Belgium.
| | - Chris Baeken
- Department of Psychiatry and Medical Psychology, Ghent Experimental Psychiatry (GHEP) Lab, Universiteit Gent - C, Heymanslaan 10, 9000, Gent, Belgium; Department of Psychiatry, UZBrussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Anouk Pierre
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium
| | - Ilse Smolders
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| | - Vincent Van Waes
- Laboratory of Clinical and Integrative Neuroscience, EA481, Université Bourgogne Franche-Comté, 19 rue Ambroise Paré, 25030, Besancon, Cedex, France.
| | - Dimitri De Bundel
- Department of Pharmaceutical Sciences, Research Group Experimental Pharmacology, Center for Neurosciences (C4N), Vrije Universiteit Brussel, Laarbeeklaan 103, 1090, Brussels, Belgium.
| |
Collapse
|
14
|
Ahmad H, Rauf K, Zada W, McCarthy M, Abbas G, Anwar F, Shah AJ. Kaempferol Facilitated Extinction Learning in Contextual Fear Conditioned Rats via Inhibition of Fatty-Acid Amide Hydrolase. Molecules 2020; 25:molecules25204683. [PMID: 33066366 PMCID: PMC7587337 DOI: 10.3390/molecules25204683] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 12/18/2022] Open
Abstract
Background: Fear, stress, and anxiety-like behaviors originate from traumatic events in life. Stress response is managed by endocannabinoids in the body by limiting the uncontrolled retrieval of aversive memories. Pharmacotherapy-modulating endocannabinoids, especially anandamide, presents a promising tool for treating anxiety disorders. Here, we investigated the effect of kaempferol, a flavonoid, in the extinction of fear related memories and associated anxiety-like behavior. Methods: The ability of kaempferol to inhibit fatty-acid amide hydrolase (FAAH, the enzyme that catabolizes anandamide) was assessed in vitro using an enzyme-linked immunosorbent assay (ELISA) kit. For animal studies (in vivo), the extinction learning was evaluated using contextual fear conditioning (CFC, a behavioral paradigm based on ability to learn and remember aversive stimuli). Furthermore, an elevated plus-maze (EPM) model was used for measuring anxiety-like behavior, while serum corticosterone served as a biochemical indicator of anxiety. Lastly, the interaction of kaempferol with FAAH enzyme was also assessed in silico (computational study). Results: Our data showed that kaempferol inhibited the FAAH enzyme with an IC50 value of 1 µM. In CFC, it reduced freezing behavior in rats. EPM data demonstrated anxiolytic activity as exhibited by enhanced number of entries and time spent in the open arm. No change in blood corticosterone levels was noted. Our computational study showed that Kaempferol interacted with the catalytic amino acids (SER241, PHE192, PHE381, and THR377) of FAAH enzyme Conclusion: Our study demonstrate that kaempferol facilitated the extinction of aversive memories along with a reduction of anxiety. The effect is mediated through the augmentation of endocannabinoids via the inhibition of FAAH enzyme.
Collapse
Affiliation(s)
- Hammad Ahmad
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Khyber Pakhtunkhwa 22060, Pakistan; (H.A.); (K.R.); (W.Z.)
| | - Khalid Rauf
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Khyber Pakhtunkhwa 22060, Pakistan; (H.A.); (K.R.); (W.Z.)
| | - Wahid Zada
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Khyber Pakhtunkhwa 22060, Pakistan; (H.A.); (K.R.); (W.Z.)
| | - Margaret McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Ghulam Abbas
- Department of Pharmacology, Faculty of Pharmacy, Ziauddin University, Karachi 75000, Pakistan;
| | - Fareeha Anwar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore 54000, Pakistan;
| | - Abdul Jabbar Shah
- Department of Pharmacy, COMSATS University Islamabad, Abbottabad Campus, Khyber Pakhtunkhwa 22060, Pakistan; (H.A.); (K.R.); (W.Z.)
- Correspondence: ; Tel.: +(92)992-383591-6
| |
Collapse
|
15
|
Leavitt RJ, Acharya MM, Baulch JE, Limoli CL. Extracellular Vesicle-Derived miR-124 Resolves Radiation-Induced Brain Injury. Cancer Res 2020; 80:4266-4277. [PMID: 32816912 PMCID: PMC7541572 DOI: 10.1158/0008-5472.can-20-1599] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/08/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Radiation-induced cognitive dysfunction (RICD) is a progressive and debilitating health issue facing patients following cranial radiotherapy to control central nervous system cancers. There has been some success treating RICD in rodents using human neural stem cell (hNSC) transplantation, but the procedure is invasive, requires immunosuppression, and could cause other complications such as teratoma formation. Extracellular vesicles (EV) are nanoscale membrane-bound structures that contain biological contents including mRNA, miRNA, proteins, and lipids that can be readily isolated from conditioned culture media. It has been previously shown that hNSC-derived EV resolves RICD following cranial irradiation using an immunocompromised rodent model. Here, we use immunocompetent wild-type mice to show that hNSC-derived EV treatment administered either intravenously via retro-orbital vein injection or via intracranial transplantation can ameliorate cognitive deficits following 9 Gy head-only irradiation. Cognitive function assessed on the novel place recognition, novel object recognition, and temporal order tasks was not only improved at early (5 weeks) but also at delayed (6 months) postirradiation times with just a single EV treatment. Improved behavioral outcomes were also associated with reduced neuroinflammation as measured by a reduction in activated microglia. To identify the mechanism of action, analysis of EV cargo implicated miRNA (miR-124) as a potential candidate in the mitigation of RICD. Furthermore, viral vector-mediated overexpression of miR-124 in the irradiated brain ameliorated RICD and reduced microglial activation. Our findings demonstrate for the first time that systemic administration of hNSC-derived EV abrogates RICD and neuroinflammation in cranially irradiated wild-type rodents through a mechanism involving miR-124. SIGNIFICANCE: Radiation-induced neurocognitive decrements in immunocompetent mice can be resolved by systemic delivery of hNSC-derived EVs involving a mechanism dependent on expression of miR-124.
Collapse
Affiliation(s)
- Ron J Leavitt
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine, Irvine, California
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, Irvine, California.
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, Irvine, California.
| |
Collapse
|
16
|
Allen BD, Syage AR, Maroso M, Baddour AAD, Luong V, Minasyan H, Giedzinski E, West BL, Soltesz I, Limoli CL, Baulch JE, Acharya MM. Mitigation of helium irradiation-induced brain injury by microglia depletion. J Neuroinflammation 2020; 17:159. [PMID: 32429943 PMCID: PMC7236926 DOI: 10.1186/s12974-020-01790-9] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 03/26/2020] [Indexed: 12/11/2022] Open
Abstract
Background Cosmic radiation exposures have been found to elicit cognitive impairments involving a wide-range of underlying neuropathology including elevated oxidative stress, neural stem cell loss, and compromised neuronal architecture. Cognitive impairments have also been associated with sustained microglia activation following low dose exposure to helium ions. Space-relevant charged particles elicit neuroinflammation that persists long-term post-irradiation. Here, we investigated the potential neurocognitive benefits of microglia depletion following low dose whole body exposure to helium ions. Methods Adult mice were administered a dietary inhibitor (PLX5622) of colony stimulating factor-1 receptor (CSF1R) to deplete microglia 2 weeks after whole body helium irradiation (4He, 30 cGy, 400 MeV/n). Cohorts of mice maintained on a normal and PLX5622 diet were tested for cognitive function using seven independent behavioral tasks, microglial activation, hippocampal neuronal morphology, spine density, and electrophysiology properties 4–6 weeks later. Results PLX5622 treatment caused a rapid and near complete elimination of microglia in the brain within 3 days of treatment. Irradiated animals on normal diet exhibited a range of behavioral deficits involving the medial pre-frontal cortex and hippocampus and increased microglial activation. Animals on PLX5622 diet exhibited no radiation-induced cognitive deficits, and expression of resting and activated microglia were almost completely abolished, without any effects on the oligodendrocyte progenitors, throughout the brain. While PLX5622 treatment was found to attenuate radiation-induced increases in post-synaptic density protein 95 (PSD-95) puncta and to preserve mushroom type spine densities, other morphologic features of neurons and electrophysiologic measures of intrinsic excitability were relatively unaffected. Conclusions Our data suggest that microglia play a critical role in cosmic radiation-induced cognitive deficits in mice and, that approaches targeting microglial function are poised to provide considerable benefit to the brain exposed to charged particles.
Collapse
Affiliation(s)
- Barrett D Allen
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Amber R Syage
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Mattia Maroso
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Al Anoud D Baddour
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Valerie Luong
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Harutyun Minasyan
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Erich Giedzinski
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | | | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, CA, USA
| | - Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine, CA, USA.
| |
Collapse
|
17
|
Kumari P, Wadhwa M, Chauhan G, Alam S, Roy K, Kumar Jha P, Kishore K, Ray K, Kumar S, Nag TC, Panjwani U. Hypobaric hypoxia induced fear and extinction memory impairment and effect of Ginkgo biloba in its amelioration: Behavioral, neurochemical and molecular correlates. Behav Brain Res 2020; 387:112595. [PMID: 32194184 DOI: 10.1016/j.bbr.2020.112595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/12/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022]
Abstract
Regulated fear and extinction memory is essential for balanced behavioral response. Limbic brain regions are susceptible to hypobaric hypoxia (HH) and are putative target for fear extinction deficit and dysregulation. The present study aimed to examine the effect of HH and Ginkgo biloba extract (GBE) on fear and extinction memory with the underlying mechanism. Adult male Sprague-Dawley rats were evaluated for fear extinction and anxious behavior following GBE administration during HH exposure. Blood and tissue (PFC, hippocampus and amygdala) samples were collected for biochemical, morphological and molecular studies. Results revealed deficit in contextual and cued fear extinction following 3 days of HH exposure. Increased corticosterone, glutamate with decreased GABA level was found with marked pyknosis, decrease in apical dendritic length and number of functional spines. Decline in mRNA expression level of synaptic plasticity genes and immunoreactivity of BDNF, synaptophysin, PSD95, spinophilin was observed following HH exposure. GBE administration during HH exposure improved fear and extinction memory along with decline in anxious behavior. It restored corticosterone, glutamate and GABA levels with an increase in apical dendritic length and number of functional spines with a reduction in pyknosis. It also improved mRNA expression level and immunoreactivity of neurotrophic and synaptic proteins. The present study is the first which demonstrates fear extinction deficit and anxious behavior following HH exposure. GBE administration ameliorated fear and extinction memory dysregulation by restoration of neurotransmitter levels, neuronal pyknosis and synaptic connections along with improved neurotrophic and synaptic protein expressions.
Collapse
Affiliation(s)
- Punita Kumari
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Meetu Wadhwa
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Garima Chauhan
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Shahnawaz Alam
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Koustav Roy
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Prabhash Kumar Jha
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Krishna Kishore
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Koushik Ray
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Sanjeev Kumar
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Tapas Chandra Nag
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| | - Usha Panjwani
- Defence Institute of Physiology & Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
18
|
Immune and Inflammatory Determinants Underlying Alzheimer's Disease Pathology. J Neuroimmune Pharmacol 2020; 15:852-862. [PMID: 32090272 DOI: 10.1007/s11481-020-09908-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/13/2020] [Indexed: 10/24/2022]
Abstract
This study examines the link between peripheral immune changes in perpetuation of the Alzheimer's disease (AD) neuropathology and cognitive deficits. Our research design using human AD patients and rodent model is supported by past evidence from genomic studies. We observed an active immune response against Aβ as indicated by the increased Aβ specific IgG antibody in the serum of AD and patients with mild cognitive impairments as compared to healthy controls. A similar increase in IgG and decrease in IgM antibody against Aβ was also confirmed in the 5xFAD mouse model of AD. More importantly, we observed a negative correlation between reduced IgM levels and cognitive dysfunction that manifested as impaired memory consolidation. Strong peripheral immune activation was supported by increased activation of microglia in the brain and macrophages in the spleen of AD mice compared to wild type control littermates. Furthermore, inflammatory cytokine IL-21 that is involved in antibody class switching was elevated in the plasma of AD patients and correlated positively with the IgG antibody levels. Concurrently, an increase in IL-21 and IL-17 was observed in spleen cells from AD mice. Further investigation revealed that proportions of T follicular helper (Tfh) cells that secrete IL-21 are increased in the spleen of AD mice. In contrast to Tfh, the frequency of B1 cells that produce IgM antibodies was reduced in AD mice. Altogether, these data indicate that in AD the immune tolerance to Aβ is compromised leading to chronic immune/inflammatory responses against Aβ that are detrimental and cause neuropathology. Graphical Abstract Healthy subjects are tolerant to Aβ and usually react weakly to it resulting the in the production of IgM class of antibodies that are efficient at clearing up self-antigens such as Aβ without causing inflammation. In contrast, Alzheimer's disease patients mount a strong immune response against Aβ probably in an effort to clear up excessive Aβ. There is enhanced production of inflammatory cytokines such as IL-21 as well as an increase in Tfh cells that cause antibody class switching form IgM to IgG. The strong immune response is inefficient at clearing up Aβ and instead exacerbates inflammation that causes AD neuropathology and cognitive dysfunction.
Collapse
|
19
|
Baddour AAD, Apodaca LA, Alikhani L, Lu C, Minasyan H, Batra RS, Acharya MM, Baulch JE. Sex-Specific Effects of a Wartime-Like Radiation Exposure on Cognitive Function. Radiat Res 2019; 193:5-15. [DOI: 10.1667/rr15413.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Al Anoud D. Baddour
- Department of Radiation Oncology, University of California Irvine, Irvine, California 92697
| | - Lauren A. Apodaca
- Department of Radiation Oncology, University of California Irvine, Irvine, California 92697
| | - Leila Alikhani
- Department of Radiation Oncology, University of California Irvine, Irvine, California 92697
| | - Celine Lu
- Department of Radiation Oncology, University of California Irvine, Irvine, California 92697
| | - Harutyun Minasyan
- Department of Radiation Oncology, University of California Irvine, Irvine, California 92697
| | - Raja S. Batra
- Department of Radiation Oncology, University of California Irvine, Irvine, California 92697
| | - Munjal M. Acharya
- Department of Radiation Oncology, University of California Irvine, Irvine, California 92697
| | - Janet E. Baulch
- Department of Radiation Oncology, University of California Irvine, Irvine, California 92697
| |
Collapse
|
20
|
Acharya MM, Baulch JE, Klein PM, Baddour AAD, Apodaca LA, Kramár EA, Alikhani L, Garcia C, Angulo MC, Batra RS, Fallgren CM, Borak TB, Stark CEL, Wood MA, Britten RA, Soltesz I, Limoli CL. New Concerns for Neurocognitive Function during Deep Space Exposures to Chronic, Low Dose-Rate, Neutron Radiation. eNeuro 2019; 6:ENEURO.0094-19.2019. [PMID: 31383727 PMCID: PMC6709229 DOI: 10.1523/eneuro.0094-19.2019] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/08/2019] [Accepted: 07/16/2019] [Indexed: 12/12/2022] Open
Abstract
As NASA prepares for a mission to Mars, concerns regarding the health risks associated with deep space radiation exposure have emerged. Until now, the impacts of such exposures have only been studied in animals after acute exposures, using dose rates ∼1.5×105 higher than those actually encountered in space. Using a new, low dose-rate neutron irradiation facility, we have uncovered that realistic, low dose-rate exposures produce serious neurocognitive complications associated with impaired neurotransmission. Chronic (6 month) low-dose (18 cGy) and dose rate (1 mGy/d) exposures of mice to a mixed field of neutrons and photons result in diminished hippocampal neuronal excitability and disrupted hippocampal and cortical long-term potentiation. Furthermore, mice displayed severe impairments in learning and memory, and the emergence of distress behaviors. Behavioral analyses showed an alarming increase in risk associated with these realistic simulations, revealing for the first time, some unexpected potential problems associated with deep space travel on all levels of neurological function.
Collapse
Affiliation(s)
- Munjal M Acharya
- Department of Radiation Oncology, University of California, Irvine, California 92697
| | - Janet E Baulch
- Department of Radiation Oncology, University of California, Irvine, California 92697
| | - Peter M Klein
- Department of Neurosurgery, Stanford University, California 94305
| | - Al Anoud D Baddour
- Department of Radiation Oncology, University of California, Irvine, California 92697
| | - Lauren A Apodaca
- Department of Radiation Oncology, University of California, Irvine, California 92697
| | - Eniko A Kramár
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697
| | - Leila Alikhani
- Department of Radiation Oncology, University of California, Irvine, California 92697
| | - Camillo Garcia
- Department of Radiation Oncology, University of California, Irvine, California 92697
| | - Maria C Angulo
- Department of Radiation Oncology, University of California, Irvine, California 92697
| | - Raja S Batra
- Department of Radiation Oncology, University of California, Irvine, California 92697
| | - Christine M Fallgren
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523
| | - Thomas B Borak
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado 80523
| | - Craig E L Stark
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697
| | - Marcello A Wood
- Department of Neurobiology and Behavior, University of California, Irvine, California 92697
| | - Richard A Britten
- Department of Radiation Oncology, Eastern Virginia Medical School, Norfolk, Virginia 23507
| | - Ivan Soltesz
- Department of Neurosurgery, Stanford University, California 94305
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, California 92697
| |
Collapse
|
21
|
Siddiqui SA, Singh S, Ugale R, Ranjan V, Kanojia R, Saha S, Tripathy S, Kumar S, Mehrotra S, Modi DR, Prakash A. Regulation of HDAC1 and HDAC2 during consolidation and extinction of fear memory. Brain Res Bull 2019; 150:86-101. [PMID: 31108155 DOI: 10.1016/j.brainresbull.2019.05.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/02/2019] [Accepted: 05/15/2019] [Indexed: 01/03/2023]
Abstract
Histone deacetylases (HDACs) regulate gene expression epigenetically through synchronized removal of acetyl groups from histones required towards memory consolidation. Moreover, dysregulated epigenetic machinery during fear or extinction learning may result in altered expression of some of these genes and result in Post Traumatic Stress Disorder (PTSD). In the present study, region-specific expression of Histone deacetylase 1 (HDAC1) and Histone deacetylase 2 (HDAC2) was correlated to the acetylation of histones H3 and H4 and the resultant conditioned response, in rats undergone fear and extinction learning. The neuronal activation, histone acetylation at H3/H4 and expression of HDAC1/HDAC2 in centrolateral amygdala (CeL) and centromedial amygdala (CeM) of central Amygdala (CeA) and prelimbic (PL) and infralimbic (IL) of Prefrontal cortex (PFC) were found to be associated in a differential manner following fear and extinction learning. Moreover in CeM, the main output of the fear circuitry, the level of HDAC1 was down-regulated following conditioning and up-regulated following extinction as opposed to which HDAC2 was down-regulated in CeM following conditioning but not following extinction. Furthermore, in CeL the HDAC1 was upregulated and HDAC2 was downregulated following conditioning and extinction. This has important implications in speculating of the role of HDACs in fear memory consolidation and its extinction.
Collapse
Affiliation(s)
| | - Sanjay Singh
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Rajesh Ugale
- Department of Pharmaceutical Sciences, RTM Nagpur University, Nagpur, India
| | - Vandana Ranjan
- Department of Biochemistry, RML University, Faizabad, India
| | - Rohit Kanojia
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Sudipta Saha
- Department of Pharmaceutical Science, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Sukanya Tripathy
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Shiv Kumar
- Department of Biochemistry, University of Lucknow, Lucknow, India
| | - Sudhir Mehrotra
- Department of Biochemistry, University of Lucknow, Lucknow, India
| | - Dinesh Raj Modi
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Anand Prakash
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India; Department of Biotech, Mahatma Gandhi Central University, Motihari, Bihar, India.
| |
Collapse
|
22
|
Sase AS, Lombroso SI, Santhumayor BA, Wood RR, Lim CJ, Neve RL, Heller EA. Sex-Specific Regulation of Fear Memory by Targeted Epigenetic Editing of Cdk5. Biol Psychiatry 2019; 85:623-634. [PMID: 30661667 DOI: 10.1016/j.biopsych.2018.11.022] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/29/2018] [Accepted: 11/15/2018] [Indexed: 10/27/2022]
Abstract
BACKGROUND Sex differences in the expression and prevalence of trauma- and stress-related disorders have led to a growing interest in the sex-specific molecular and epigenetic mechanisms underlying these diseases. Cyclin-dependent kinase 5 (CDK5) is known to underlie both fear memory and stress behavior in male mice. Given our recent finding that targeted histone acetylation of Cdk5 regulates stress responsivity in male mice, we hypothesized that such a mechanism may be functionally relevant in female mice as well. METHODS We applied epigenetic editing of Cdk5 in the hippocampus and examined the regulation of fear memory retrieval in male and female mice. Viral expression of zinc finger proteins targeting histone acetylation to the Cdk5 promoter was paired with a quantification of learning and memory of contextual fear conditioning, expression of CDK5, and enrichment of histone modifications of the Cdk5 gene. RESULTS We found that male mice exhibit stronger long-term memory retrieval than do female mice, and this finding was associated with male-specific epigenetic activation of hippocampal Cdk5 expression. Sex differences in behavior and epigenetic regulation of Cdk5 occurred after long-term, but not short-term, fear memory retrieval. Finally, targeted histone acetylation of hippocampal Cdk5 promoter attenuated fear memory retrieval and increased tau phosphorylation in female but not male mice. CONCLUSIONS Epigenetic editing uncovered a female-specific role of Cdk5 activation in attenuating fear memory retrieval. This finding may be attributed to CDK5 mediated hyperphosphorylation of tau only in the female hippocampus. Sex-specific epigenetic regulation of Cdk5 may reflect differences in the effect of CDK5 on downstream target proteins that regulate memory.
Collapse
Affiliation(s)
- Ajinkya S Sase
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sonia I Lombroso
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brandon A Santhumayor
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rozalyn R Wood
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Carissa J Lim
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rachael L Neve
- Gene Delivery Technology Core, Massachusetts General Hospital, Boston, Massachusetts
| | - Elizabeth A Heller
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania; Penn Epigenetics Institute, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
23
|
Penha Farias C, Guerino Furini CR, Godfried Nachtigall E, Kielbovicz Behling JA, Silva de Assis Brasil E, Bühler L, Izquierdo I, de Carvalho Myskiw J. Extinction learning with social support depends on protein synthesis in prefrontal cortex but not hippocampus. Proc Natl Acad Sci U S A 2019; 116:1765-1769. [PMID: 30635411 PMCID: PMC6358673 DOI: 10.1073/pnas.1815893116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Extinction of contextual fear conditioning (CFC) in the presence of a familiar nonfearful conspecific (social support), such as that of others tasks, can occur regardless of whether the original memory is retrieved during the extinction training. Extinction with social support is blocked by the protein synthesis inhibitors anisomycin and rapamycin and by the inhibitor of gene expression 5,6-dichloro-1-β-d-ribofuranosylbenzimidazole infused immediately after extinction training into the ventromedial prefrontal cortex (vmPFC) but unlike regular CFC extinction not in the CA1 region of the dorsal hippocampus. So social support generates a form of learning that differs from extinction acquired without social support in terms of the brain structures involved. This finding may lead to a better understanding of the brain mechanisms involved in the social support of memories and in therapies for disorders related to dysfunctional fear memories. Thus, here we show that the consolidation of extinction memory with social support relies on vmPFC rather than hippocampus gene expression and ribosomal- and mammalian target of rapamycin-dependent protein synthesis. These results provide additional knowledge about the cellular mechanisms and brain structures involved on the effect of social support in changing behavior and fear extinction memory.
Collapse
Affiliation(s)
- Clarissa Penha Farias
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
- National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Cristiane Regina Guerino Furini
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
- National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Eduarda Godfried Nachtigall
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
| | - Jonny Anderson Kielbovicz Behling
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
| | - Eduardo Silva de Assis Brasil
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
| | - Letícia Bühler
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil
| | - Ivan Izquierdo
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil;
- National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Jociane de Carvalho Myskiw
- Memory Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, 90610-000 Porto Alegre, RS, Brazil;
- National Institute of Translational Neuroscience (INNT), National Research Council of Brazil, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Goode TD, Maren S. Common neurocircuitry mediating drug and fear relapse in preclinical models. Psychopharmacology (Berl) 2019; 236:415-437. [PMID: 30255379 PMCID: PMC6373193 DOI: 10.1007/s00213-018-5024-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 09/03/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Comorbidity of anxiety disorders, stressor- and trauma-related disorders, and substance use disorders is extremely common. Moreover, therapies that reduce pathological fear and anxiety on the one hand, and drug-seeking on the other, often prove short-lived and are susceptible to relapse. Considerable advances have been made in the study of the neurobiology of both aversive and appetitive extinction, and this work reveals shared neural circuits that contribute to both the suppression and relapse of conditioned responses associated with trauma or drug use. OBJECTIVES The goal of this review is to identify common neural circuits and mechanisms underlying relapse across domains of addiction biology and aversive learning in preclinical animal models. We focus primarily on neural circuits engaged during the expression of relapse. KEY FINDINGS After extinction, brain circuits involving the medial prefrontal cortex and hippocampus come to regulate the expression of conditioned responses by the amygdala, bed nucleus of the stria terminalis, and nucleus accumbens. During relapse, hippocampal projections to the prefrontal cortex inhibit the retrieval of extinction memories resulting in a loss of inhibitory control over fear- and drug-associated conditional responding. CONCLUSIONS The overlapping brain systems for both fear and drug memories may explain the co-occurrence of fear and drug-seeking behaviors.
Collapse
Affiliation(s)
- Travis D Goode
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, 301 Old Main Dr., College Station, TX, 77843-3474, USA
| | - Stephen Maren
- Department of Psychological and Brain Sciences and Institute for Neuroscience, Texas A&M University, 301 Old Main Dr., College Station, TX, 77843-3474, USA.
| |
Collapse
|
25
|
Radecki DZ, Johnson EL, Brown AK, Meshkin NT, Perrine SA, Gow A. Corticohippocampal Dysfunction In The OBiden Mouse Model Of Primary Oligodendrogliopathy. Sci Rep 2018; 8:16116. [PMID: 30382234 PMCID: PMC6208344 DOI: 10.1038/s41598-018-34414-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 09/25/2018] [Indexed: 12/18/2022] Open
Abstract
Despite concerted efforts over decades, the etiology of multiple sclerosis (MS) remains unclear. Autoimmunity, environmental-challenges, molecular mimicry and viral hypotheses have proven equivocal because early-stage disease is typically presymptomatic. Indeed, most animal models of MS also lack defined etiologies. We have developed a novel adult-onset oligodendrogliopathy using a delineated metabolic stress etiology in myelinating cells, and our central question is, “how much of the pathobiology of MS can be recapitulated in this model?” The analyses described herein demonstrate that innate immune activation, glial scarring, cortical and hippocampal damage with accompanying electrophysiological, behavioral and memory deficits naturally emerge from disease progression. Molecular analyses reveal neurofilament changes in normal-appearing gray matter that parallel those in cortical samples from MS patients with progressive disease. Finally, axon initial segments of deep layer pyramidal neurons are perturbed in entorhinal/frontal cortex and hippocampus from OBiden mice, and computational modeling provides insight into vulnerabilities of action potential generation during demyelination and early remyelination. We integrate these findings into a working model of corticohippocampal circuit dysfunction to predict how myelin damage might eventually lead to cognitive decline.
Collapse
Affiliation(s)
- Daniel Z Radecki
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA.,Department of Comparative Biosciences, University of Wisconsin-Madison School of Veterinary Medicine, Madison, WI, 53706, USA
| | - Elizabeth L Johnson
- Institute of Gerontology, Wayne State University, Detroit, MI, 48202, USA.,Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA
| | - Ashley K Brown
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Nicholas T Meshkin
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA.,Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, School of Medicine, Wayne State University, Detroit, MI, 48201, USA
| | - Alexander Gow
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA. .,Carman and Ann Adams Department of Pediatrics, School of Medicine, Wayne State University, Detroit, MI, 48201, USA. .,Department of Neurology, School of Medicine, Wayne State University, Detroit, MI, 48201, USA.
| |
Collapse
|
26
|
Fuentes S, Daviu N, Gagliano H, Belda X, Armario A, Nadal R. Early life stress in rats sex-dependently affects remote endocrine rather than behavioral consequences of adult exposure to contextual fear conditioning. Horm Behav 2018; 103:7-18. [PMID: 29802874 DOI: 10.1016/j.yhbeh.2018.05.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 04/09/2018] [Accepted: 05/22/2018] [Indexed: 01/07/2023]
Abstract
Exposure to electric foot-shocks can induce in rodents contextual fear conditioning, generalization of fear to other contexts and sensitization of the hypothalamic-pituitary-adrenal (HPA) axis to further stressors. All these aspects are relevant for the study of post-traumatic stress disorder. In the present work we evaluated in rats the sex differences and the role of early life stress (ELS) in fear memories, generalization and sensitization. During the first postnatal days subjects were exposed to restriction of nesting material along with exposure to a "substitute" mother. In the adulthood they were exposed to (i) a contextual fear conditioning to evaluate long-term memory and extinction and (ii) to a novel environment to study cognitive fear generalization and HPA axis heterotypic sensitization. ELS did not alter acquisition, expression or extinction of context fear conditioned behavior (freezing) in either sex, but reduced activity in novel environments only in males. Fear conditioning associated hypoactivity in novel environments (cognitive generalization) was greater in males than females but was not specifically affected by ELS. Although overall females showed greater basal and stress-induced levels of ACTH and corticosterone, an interaction between ELS, shock exposure and sex was found regarding HPA hormones. In males, ELS did not affect ACTH response in any situation, whereas in females, ELS reduced both shock-induced sensitization of ACTH and its conditioned response to the shock context. Also, shock-induced sensitization of corticosterone was only observed in males and ELS specifically reduced corticosterone response to stressors in males but not females. In conclusion, ELS seems to have only a minor impact on shock-induced behavioral conditioning, while affecting the unconditioned and conditioned responses of HPA hormones in a sex-dependent manner.
Collapse
Affiliation(s)
- Sílvia Fuentes
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Animal Physiology Unit, School of Biosciences, Universitat Autònoma de Barcelona, Spain
| | - Núria Daviu
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Spain
| | - Humberto Gagliano
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Spain
| | - Xavier Belda
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Spain
| | - Antonio Armario
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Psychobiology Unit, School of Psychology, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Instituto de Salud Carlos III, Spain.
| | - Roser Nadal
- Institut de Neurociències, Universitat Autònoma de Barcelona, Spain; Animal Physiology Unit, School of Biosciences, Universitat Autònoma de Barcelona, Spain; CIBERSAM, Instituto de Salud Carlos III, Spain.
| |
Collapse
|
27
|
Staib JM, Della Valle R, Knox DK. Disruption of medial septum and diagonal bands of Broca cholinergic projections to the ventral hippocampus disrupt auditory fear memory. Neurobiol Learn Mem 2018; 152:71-79. [PMID: 29783059 DOI: 10.1016/j.nlm.2018.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 04/24/2018] [Accepted: 05/17/2018] [Indexed: 01/08/2023]
Abstract
In classical fear conditioning, a neutral conditioned stimulus (CS) is paired with an aversive unconditioned stimulus (US), which leads to a fear memory. If the CS is repeatedly presented without the US after fear conditioning, the formation of an extinction memory occurs, which inhibits fear memory expression. A previous study has demonstrated that selective cholinergic lesions in the medial septum and vertical limb of the diagonal bands of Broca (MS/vDBB) prior to fear and extinction learning disrupt contextual fear memory discrimination and acquisition of extinction memory. MS/vDBB cholinergic neurons project to a number of substrates that are critical for fear and extinction memory. However, it is currently unknown which of these efferent projections are critical for contextual fear memory discrimination and extinction memory. To address this, we induced cholinergic lesions in efferent targets of MS/vDBB cholinergic neurons. These included the dorsal hippocampus (dHipp), ventral hippocampus (vHipp), medial prefrontal cortex (mPFC), and in the mPFC and dHipp combined. None of these lesion groups exhibited deficits in contextual fear memory discrimination or extinction memory. However, vHipp cholinergic lesions disrupted auditory fear memory. Because MS/vDBB cholinergic neurons are the sole source of acetylcholine in the vHipp, these results suggest that MS/vDBB cholinergic input to the vHipp is critical for auditory fear memory. Taken together with previous findings, the results of this study suggest that MS/vDBB cholinergic neurons are critical for fear and extinction memory, though further research is needed to elucidate the role of MS/vDBB cholinergic neurons in these types of emotional memory.
Collapse
Affiliation(s)
- Jennifer M Staib
- University of Delaware, Department of Psychological and Brain Sciences, United States
| | - Rebecca Della Valle
- University of Delaware, Department of Psychological and Brain Sciences, United States
| | - Dayan K Knox
- University of Delaware, Department of Psychological and Brain Sciences, United States.
| |
Collapse
|
28
|
Doherty TS, Blaze J, Keller SM, Roth TL. Phenotypic outcomes in adolescence and adulthood in the scarcity-adversity model of low nesting resources outside the home cage. Dev Psychobiol 2017; 59:703-714. [PMID: 28767135 PMCID: PMC5569321 DOI: 10.1002/dev.21547] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 06/27/2017] [Indexed: 01/10/2023]
Abstract
Early life adversity is known to disrupt behavioral trajectories and many rodent models have been developed to characterize these stress-induced outcomes. One example is the scarcity-adversity model of low nesting resources. This model employs resource scarcity (i.e., low nesting materials) to elicit adverse caregiving conditions (including maltreatment) toward rodent neonates. Our lab utilizes a version of this model wherein caregiving exposures occur outside the home cage during the first postnatal week. The aim of this study was to determine adolescent and adult phenotypic outcomes associated with this model, including assessment of depressive- and anxiety-like behaviors and performance in different cognitive domains. Exposure to adverse caregiving had no effect on adolescent behavioral performance whereas exposure significantly impaired adult behavioral performance. Further, adult behavioral assays revealed substantial differences between sexes. Overall, data demonstrate the ability of repeated exposure to brief bouts of maltreatment outside the home cage in infancy to impact the development of several behavioral domains later in life.
Collapse
Affiliation(s)
- Tiffany S. Doherty
- Department of Psychological and Brain Sciences, University of Delaware,
Newark DE 19716
| | | | | | - Tania L. Roth
- Department of Psychological and Brain Sciences, University of Delaware,
Newark DE 19716
| |
Collapse
|
29
|
Goode TD, Maren S. Role of the bed nucleus of the stria terminalis in aversive learning and memory. Learn Mem 2017; 24:480-491. [PMID: 28814474 PMCID: PMC5580527 DOI: 10.1101/lm.044206.116] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 06/30/2017] [Indexed: 02/06/2023]
Abstract
Surviving threats in the environment requires brain circuits for detecting (or anticipating) danger and for coordinating appropriate defensive responses (e.g., increased cardiac output, stress hormone release, and freezing behavior). The bed nucleus of the stria terminalis (BNST) is a critical interface between the "affective forebrain"-including the amygdala, ventral hippocampus, and medial prefrontal cortex-and the hypothalamic and brainstem areas that have been implicated in neuroendocrine, autonomic, and behavioral responses to actual or anticipated threats. However, the precise contribution of the BNST to defensive behavior is unclear, both in terms of the antecedent stimuli that mobilize BNST activity and the consequent defensive reactions. For example, it is well known that the BNST is essential for contextual fear conditioning, but dispensable for fear conditioning to discrete conditioned stimuli (CSs), at least as indexed by freezing behavior. However, recent evidence suggests that there are circumstances in which contextual freezing may persist independent of the BNST. Furthermore, the BNST is involved in the reinstatement (or relapse) of conditioned freezing to extinguished discrete CSs. As such, there are critical gaps in understanding how the BNST contributes to fundamental processes involved in Pavlovian fear conditioning. Here, we attempt to provide an integrative account of BNST function in fear conditioning. We discuss distinctions between unconditioned stress and conditioned fear and the role of BNST circuits in organizing behaviors associated with these states. We propose that the BNST mediates conditioned defensive responses-not based on the modality or duration of the antecedent threat or the duration of the behavioral response to the threat-but rather as consequence the ability of an antecedent stimulus to predict when an aversive outcome will occur (i.e., its temporal predictability). We argue that the BNST is not uniquely mobilized by sustained threats or uniquely involved in organizing sustained fear responses. In contrast, we argue that the BNST is involved in organizing fear responses to stimuli that poorly predict when danger will occur, no matter the duration, modality, or complexity of those stimuli. The concepts discussed in this review are critical to understanding the contribution of the human BNST to fear and anxiety disorders.
Collapse
Affiliation(s)
- Travis D Goode
- Institute for Neuroscience and the Department of Psychology, Texas A&M University, College Station, Texas 77843-3474, USA
| | - Stephen Maren
- Institute for Neuroscience and the Department of Psychology, Texas A&M University, College Station, Texas 77843-3474, USA
| |
Collapse
|
30
|
Osborne BF, Caulfield JI, Solomotis SA, Schwarz JM. Neonatal infection produces significant changes in immune function with no associated learning deficits in juvenile rats. Dev Neurobiol 2017; 77:1221-1236. [PMID: 28719141 DOI: 10.1002/dneu.22512] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/09/2017] [Accepted: 07/13/2017] [Indexed: 12/30/2022]
Abstract
The current experiments examined the impact of early-life immune activation and a subsequent mild immune challenge with lipopolysaccharide (LPS; 25µg/kg) on hippocampal-dependent learning, proinflammatory cytokine expression in the brain, and peripheral immune function in juvenile male and female rats at P24, an age when hippocampal-dependent learning and memory first emerges. Our results indicate that neonatal infection did not produce learning deficits in the hippocampal-dependent context pre-exposure facilitation effect paradigm in juvenile males and females, contrary to what has been observed in adults. Neonatal infection produced an increase in baseline IL-1β expression in the hippocampus (HP) and medial prefrontal cortex (mPFC) of juvenile rats. Furthermore, neonatally infected rats showed exaggerated IL-1β expression in the HP following LPS treatment as juveniles; and juvenile females, but not males, showed exaggerated IL-1β expression in the mPFC following LPS treatment. Neonatal infection attenuated the production of IL-6 expression following LPS treatment in both the brain and the spleen, and neonatal infection decreased the numbers of circulating white blood cells in juvenile males and females, an effect that was further exacerbated by subsequent LPS treatment. Together, our data indicate that the consequences of neonatal infection are detectable even early in juvenile development, though we found no concomitant hippocampal-dependent learning deficits at this young age. These findings underscore the need to consider age and associated on-going neurodevelopmental processes as important factors contributing to the emergence of cognitive and behavioral disorders linked to early-life immune activation. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 77: 1221-1236, 2017.
Collapse
Affiliation(s)
- Brittany F Osborne
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, 19716
| | - Jasmine I Caulfield
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, 19716
| | - Samantha A Solomotis
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, 19716
| | - Jaclyn M Schwarz
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, 19716
| |
Collapse
|
31
|
Guerra GP, Rubin MA, Mello CF. Modulation of learning and memory by natural polyamines. Pharmacol Res 2016; 112:99-118. [PMID: 27015893 DOI: 10.1016/j.phrs.2016.03.023] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 03/09/2016] [Accepted: 03/11/2016] [Indexed: 01/08/2023]
Abstract
Spermine and spermidine are natural polyamines that are produced mainly via decarboxylation of l-ornithine and the sequential transfer of aminopropyl groups from S-adenosylmethionine to putrescine by spermidine synthase and spermine synthase. Spermine and spermidine interact with intracellular and extracellular acidic residues of different nature, including nucleic acids, phospholipids, acidic proteins, carboxyl- and sulfate-containing polysaccharides. Therefore, multiple actions have been suggested for these polycations, including modulation of the activity of ionic channels, protein synthesis, protein kinases, and cell proliferation/death, within others. In this review we summarize these neurochemical/neurophysiological/morphological findings, particularly those that have been implicated in the improving and deleterious effects of spermine and spermidine on learning and memory of naïve animals in shock-motivated and nonshock-motivated tasks, from a historical perspective. The interaction with the opioid system, the facilitation and disruption of morphine-induced reward and the effect of polyamines and putative polyamine antagonists on animal models of cognitive diseases, such as Alzheimer's, Huntington, acute neuroinflammation and brain trauma are also reviewed and discussed. The increased production of polyamines in Alzheimer's disease and the biphasic nature of the effects of polyamines on memory and on the NMDA receptor are also considered. In light of the current literature on polyamines, which include the description of an inborn error of the metabolism characterized by mild-to moderate mental retardation and polyamine metabolism alterations in suicide completers, we can anticipate that polyamine targets may be important for the development of novel strategies and approaches for understanding the etiopathogenesis of important central disorders and their pharmacological treatment.
Collapse
Affiliation(s)
- Gustavo Petri Guerra
- Department of Food Technology, Federal Technological University of Paraná, Campus Medianeira, Medianeira, PR 85884-000, Brazil
| | - Maribel Antonello Rubin
- Department of Biochemistry, Center of Exact and Natural Sciences, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil.
| | - Carlos Fernando Mello
- Department of Physiology and Pharmacology, Center of Health Sciences, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
32
|
Knox D, Keller SM. Cholinergic neuronal lesions in the medial septum and vertical limb of the diagonal bands of Broca induce contextual fear memory generalization and impair acquisition of fear extinction. Hippocampus 2015; 26:718-26. [PMID: 26606423 DOI: 10.1002/hipo.22553] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2015] [Indexed: 11/06/2022]
Abstract
Previous research has shown that the ventral medial prefrontal cortex (vmPFC) and hippocampus (Hipp) are critical for extinction memory. Basal forebrain (BF) cholinergic input to the vmPFC and Hipp is critical for neural function in these substrates, which suggests BF cholinergic neurons may be critical for extinction memory. In order to test this hypothesis, we applied cholinergic lesions to different regions of the BF and observed the effects these lesions had on extinction memory. Complete BF cholinergic lesions induced contextual fear memory generalization, and this generalized fear was resistant to extinction. Animals with complete BF cholinergic lesions could not acquire cued fear extinction. Restricted cholinergic lesions in the medial septum and vertical diagonal bands of Broca (MS/vDBB) mimicked the effects that BF cholinergic lesions had on contextual fear memory generalization and acquisition of fear extinction. Cholinergic lesions in the horizontal diagonal band of Broca and nucleus basalis (hDBB/NBM) induced a small deficit in extinction of generalized contextual fear memory with no accompanying deficits in cued fear extinction. The results of this study reveal that MS/vDBB cholinergic neurons are critical for inhibition and extinction of generalized contextual fear memory, and via this process, may be critical for acquisition of cued fear extinction. Further studies delineating neural circuits and mechanisms through which MS/vDBB cholinergic neurons facilitate these emotional memory processes are needed. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dayan Knox
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| | - Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware
| |
Collapse
|
33
|
Shang S, Wang C, Guo C, Huang X, Wang L, Zhang C. The formation and extinction of fear memory in tree shrews. Front Behav Neurosci 2015; 9:204. [PMID: 26283941 PMCID: PMC4518201 DOI: 10.3389/fnbeh.2015.00204] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/15/2015] [Indexed: 02/01/2023] Open
Abstract
Fear is an emotion that is well-studied due to its importance for animal survival. Experimental animals, such as rats and mice, have been widely used to model fear. However, higher animals such as nonhuman primates have rarely been used to study fear due to ethical issues and high costs. Tree shrews are small mammals that are closely related to primates; they have been used to model human-related psychosocial conditions such as stress and alcohol tolerance. Here, we describe an experimental paradigm to study the formation and extinction of fear memory in tree shrews. We designed an experimental apparatus of a light/dark box with a voltage foot shock. We found that tree shrews preferred staying in the dark box in the daytime without stimulation and showed avoidance to voltage shocks applied to the footplate in a voltage-dependent manner. Foot shocks applied to the dark box for 5 days (10 min per day) effectively reversed the light–dark preference of the tree shrews, and this memory lasted for more than 50 days without any sign of memory decay (extinction) in the absence of further stimulation. However, this fear memory was reversed with 4 days of reverse training by applying the same stimulus to the light box. When reducing the stimulus intensity during the training period, a memory extinction and subsequently reinstatement effects were observed. Thus, our results describe an efficient method of monitoring fear memory formation and extinction in tree shrews.
Collapse
Affiliation(s)
- Shujiang Shang
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University Beijing, China ; Department of Physiology, Anhui Medical University Hefei, China
| | - Cong Wang
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University Beijing, China ; Department of Physiology, Anhui Medical University Hefei, China
| | - Chengbing Guo
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University Beijing, China ; Department of Physiology, Anhui Medical University Hefei, China
| | - Xu Huang
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University Beijing, China ; Department of Physiology, Anhui Medical University Hefei, China
| | - Liecheng Wang
- Department of Physiology, Anhui Medical University Hefei, China
| | - Chen Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences; PKU-IDG/McGovern Institute for Brain Research, Peking University Beijing, China
| |
Collapse
|
34
|
Reversible Inactivation of the Bed Nucleus of the Stria Terminalis Prevents Reinstatement But Not Renewal of Extinguished Fear. eNeuro 2015; 2:eN-NWR-0037-15. [PMID: 26464990 PMCID: PMC4586936 DOI: 10.1523/eneuro.0037-15.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/02/2015] [Accepted: 06/16/2015] [Indexed: 12/17/2022] Open
Abstract
The extinction of conditioned fear is labile. For example, fear to an extinguished conditioned stimulus (CS) returns after presentation of an aversive stimulus ("reinstatement") or a change in context ("renewal"). Substantial research implicates the bed nucleus of the stria terminalis (BNST) in the stress-induced relapse of extinguished behaviors, such as in instrumental drug seeking, but its role in the relapse of extinguished fear responses is not clear. Here, we explored the role of the BNST in both the reinstatement and renewal of fear, two forms of relapse that are differentially triggered by stress. In Experiment 1, rats received pairings of an auditory CS and footshock unconditioned stimulus (US) followed by an extinction procedure. After extinction, rats received an unsignaled US to reinstate fear to the extinguished CS. Twenty-four hours later, they were infused with either muscimol or vehicle into the BNST immediately prior to a CS retrieval test. In Experiment 2, rats were conditioned and extinguished in two distinct contexts. Twenty-four hours after extinction, the rats were infused with muscimol, NBQX, or vehicle immediately prior to a CS retrieval test in either the extinction context or a different (but familiar) context. In both experiments, freezing behavior served as the index of conditioned fear. The results revealed that BNST inactivation prevented reinstatement (Experiment 1), but not renewal (Experiment 2), of conditioned freezing to the extinguished CS. Hence, the BNST is critical for the reinstatement of extinguished fear in an aversive context, but not for the contextual retrieval processes that mediate fear renewal.
Collapse
|
35
|
Keller SM, Schreiber WB, Stanfield BR, Knox D. Inhibiting corticosterone synthesis during fear memory formation exacerbates cued fear extinction memory deficits within the single prolonged stress model. Behav Brain Res 2015; 287:182-6. [PMID: 25839906 DOI: 10.1016/j.bbr.2015.03.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 03/20/2015] [Accepted: 03/22/2015] [Indexed: 11/16/2022]
Abstract
Using the single prolonged stress (SPS) animal model of post-traumatic stress disorder (PTSD), previous studies suggest that enhanced glucocorticoid receptor (GR) expression leads to cued fear extinction retention deficits. However, it is unknown how the endogenous ligand of GRs, corticosterone (CORT), may contribute to extinction retention deficits in the SPS model. Given that CORT synthesis during fear learning is critical for fear memory consolidation and SPS enhances GR expression, CORT synthesis during fear memory formation could strengthen fear memory in SPS rats by enhancing GR activation during fear learning. In turn, this could lead to cued fear extinction retention deficits. We tested the hypothesis that CORT synthesis during fear learning leads to cued fear extinction retention deficits in SPS rats by administering the CORT synthesis inhibitor metyrapone to SPS and control rats prior to fear conditioning, and observed the effect this had on extinction memory. Inhibiting CORT synthesis during fear memory formation in control rats tended to decrease cued freezing, though this effect never reached statistical significance. Contrary to our hypothesis, inhibiting CORT synthesis during fear memory formation disrupted extinction retention in SPS rats. This finding suggests that even though SPS exposure leads to cued fear extinction memory deficits, CORT synthesis during fear memory formation enhances extinction retention in SPS rats. This suggests that stress-induced CORT synthesis in previously stressed rats can be beneficial.
Collapse
Affiliation(s)
- Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - William B Schreiber
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - Briana R Stanfield
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States
| | - Dayan Knox
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE, United States.
| |
Collapse
|
36
|
Goode TD, Kim JJ, Maren S. Relapse of extinguished fear after exposure to a dangerous context is mitigated by testing in a safe context. Learn Mem 2015; 22:170-8. [PMID: 25691517 PMCID: PMC4340132 DOI: 10.1101/lm.037028.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 01/13/2015] [Indexed: 12/30/2022]
Abstract
Aversive events can trigger relapse of extinguished fear memories, presenting a major challenge to the long-term efficacy of therapeutic interventions. Here, we examined factors regulating the relapse of extinguished fear after exposure of rats to a dangerous context. Rats received unsignaled shock in a distinct context ("dangerous" context) 24 h prior to auditory fear conditioning in another context. Fear to the auditory conditioned stimulus (CS) was subsequently extinguished either in the conditioning context ("ambiguous" context) or in a third novel context ("safe" context). Exposure to the dangerous context 30 min before a CS retention test caused relapse to the CS in the ambiguous and safe test contexts relative to nonextinguished controls. When rats were tested 24 h later (with or without short-term testing), rats tested in the ambiguous context continued to exhibit relapse, whereas rats tested in the safe context did not. Additionally, exposure of rats to the conditioning context--in place of the unsignaled shock context--did not result in relapse of fear to the CS in the safe testing context. Our work highlights the vulnerabilities of extinction recall to interference, and demonstrates the importance of context associations in the relapse of fear after extinction.
Collapse
Affiliation(s)
- Travis D Goode
- Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3473, USA
| | - Janice J Kim
- Department of Psychology, Texas A&M University, College Station, Texas 77843-3473, USA
| | - Stephen Maren
- Institute for Neuroscience, Texas A&M University, College Station, Texas 77843-3473, USA Department of Psychology, Texas A&M University, College Station, Texas 77843-3473, USA
| |
Collapse
|
37
|
High-dose corticosterone after fear conditioning selectively suppresses fear renewal by reducing anxiety-like response. Pharmacol Biochem Behav 2014; 124:188-95. [PMID: 24933336 DOI: 10.1016/j.pbb.2014.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 05/22/2014] [Accepted: 06/07/2014] [Indexed: 11/21/2022]
Abstract
Exposure therapy is widely used to treat anxiety disorders, including posttraumatic stress disorder (PTSD). However, preventing the return of fear is still a major challenge after this behavioral treatment. An increasing number of studies suggest that high-dose glucocorticoid treatment immediately after trauma can alleviate the symptoms of PTSD in humans. Unknown is whether high-dose glucocorticoid treatment following fear conditioning suppresses the return of fear. In the present study, a typical fear renewal paradigm (AAB) was used, in which the fear response to an auditory cue can be restored in a novel context (context B) when both training and extinction occur in the same context (context A). We trained rats for auditory fear conditioning and administered corticosterone (CORT; 5 and 25mg/kg, i.p.) or vehicle with different delays (1 and 24h). Forty-eight hours after drug injection, extinction was conducted with no drug in the training context, followed by a test of tone-induced freezing behavior in the same (AAA) or a shifted (AAB) context. Both immediate and delayed administration of high-dose CORT after fear conditioning reduced fear renewal. To examine the anxiolytic effect of CORT, independent rats were trained for cued or contextual fear conditioning, followed by an injection of CORT (5 and 25mg/kg, i.p.) or vehicle at a 1 or 24h delay. One week later, anxiety-like behavior was assessed in the elevated plus maze (EPM) before and after fear expression. We found that high-dose CORT decreased anxiety-like behavior without changing tone- or context-induced freezing. These findings indicate that a single high-dose CORT administration given after fear conditioning may selectively suppress fear renewal by reducing anxiety-like behavior and not by altering the consolidation, retrieval, or extinction of fear memory.
Collapse
|
38
|
Abstract
Whereas fear memories are rapidly acquired and enduring over time, extinction memories are slow to form and are susceptible to disruption. Consequently, behavioral therapies that involve extinction learning (e.g., exposure therapy) often produce only temporary suppression of fear and anxiety. This review focuses on the factors that are known to influence the relapse of extinguished fear. Several phenomena associated with the return of fear after extinction are discussed, including renewal, spontaneous recovery, reacquisition, and reinstatement. Additionally, this review describes recent work, which has focused on the role of psychological stress in the relapse of extinguished fear. Recent developments in behavioral and pharmacological research are examined in light of treatment of pathological fear in humans.
Collapse
|
39
|
An B, Hong I, Choi S. Long-term neural correlates of reversible fear learning in the lateral amygdala. J Neurosci 2012; 32:16845-56. [PMID: 23175837 PMCID: PMC6621751 DOI: 10.1523/jneurosci.3017-12.2012] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2012] [Revised: 08/31/2012] [Accepted: 09/24/2012] [Indexed: 11/21/2022] Open
Abstract
Fear conditioning and extinction are behavioral models that reflect the association and dissociation of environmental cues to aversive outcomes, both known to involve the lateral amygdala (LA). Accordingly, responses of LA neurons to conditioned stimuli (CS) increase after fear conditioning and decrease partially during extinction. However, the long-term effects of repeated fear conditioning and extinction on LA neuronal firing have not been explored. Here we show, using stable, high signal-to-noise ratio single-unit recordings, that the ensemble activity of all recorded LA neurons correlates tightly with conditioned fear responses of rats in a conditioning/extinction/reconditioning paradigm spanning 3 d. This CS-evoked ensemble activity increased after conditioning, decreased after extinction, and was repotentiated after reconditioning. Cell-by-cell analysis revealed that among the LA neurons that displayed potentiated responses after initial fear conditioning, some exhibited weakened CS responses after extinction (extinction-susceptible), whereas others remained potentiated (extinction-resistant). The majority of extinction-susceptible neurons exhibited strong potentiation after reconditioning, suggesting that this distinct subpopulation (reversible fear neurons) encodes updated CS-unconditioned stimulus (US) association strength. Interestingly, these reversible fear neurons displayed larger, more rapid potentiation during reconditioning compared with the initial conditioning, providing a neural correlate of savings after extinction. In contrast, the extinction-resistant fear neurons did not show further increases after reconditioning, suggesting that this subpopulation encodes persistent fear memory representing the original CS-US association. This longitudinal report on LA neuronal activity during reversible fear learning suggests the existence of distinct populations encoding various facets of fear memory and provides insight into the neuronal mechanisms of fear memory modulation.
Collapse
Affiliation(s)
- Bobae An
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Ingie Hong
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| | - Sukwoo Choi
- School of Biological Sciences, College of Natural Sciences, Seoul National University, Seoul 151-742, Republic of Korea
| |
Collapse
|
40
|
Na ES, Nelson ED, Adachi M, Autry AE, Mahgoub MA, Kavalali ET, Monteggia LM. A mouse model for MeCP2 duplication syndrome: MeCP2 overexpression impairs learning and memory and synaptic transmission. J Neurosci 2012; 32:3109-17. [PMID: 22378884 PMCID: PMC3835557 DOI: 10.1523/jneurosci.6000-11.2012] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2011] [Revised: 12/30/2011] [Accepted: 01/06/2012] [Indexed: 01/01/2023] Open
Abstract
Rett syndrome and MECP2 duplication syndrome are neurodevelopmental disorders that arise from loss-of-function and gain-of-function alterations in methyl-CpG binding protein 2 (MeCP2) expression, respectively. Although there have been studies examining MeCP2 loss of function in animal models, there is limited information on MeCP2 overexpression in animal models. Here, we characterize a mouse line with MeCP2 overexpression restricted to neurons (Tau-Mecp2). This MeCP2 overexpression line shows motor coordination deficits, heightened anxiety, and impairments in learning and memory that are accompanied by deficits in long-term potentiation and short-term synaptic plasticity. Whole-cell voltage-clamp recordings of cultured hippocampal neurons from Tau-Mecp2 mice reveal augmented frequency of miniature EPSCs with no change in miniature IPSCs, indicating that overexpression of MeCP2 selectively impacts excitatory synapse function. Moreover, we show that alterations in transcriptional repression mechanisms underlie the synaptic phenotypes in hippocampal neurons from the Tau-Mecp2 mice. These results demonstrate that the Tau-Mecp2 mouse line recapitulates many key phenotypes of MECP2 duplication syndrome and support the use of these mice to further study this devastating disorder.
Collapse
Affiliation(s)
| | | | | | | | | | - Ege T. Kavalali
- Neuroscience, The University of Texas Southwestern Medical Center, Dallas, Texas 75390-9070
| | | |
Collapse
|
41
|
Knox D, George SA, Fitzpatrick CJ, Rabinak CA, Maren S, Liberzon I. Single prolonged stress disrupts retention of extinguished fear in rats. Learn Mem 2012; 19:43-9. [PMID: 22240323 DOI: 10.1101/lm.024356.111] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Clinical research has linked post-traumatic stress disorder (PTSD) with deficits in fear extinction. However, it is not clear whether these deficits result from stress-related changes in the acquisition or retention of extinction or in the regulation of extinction memories by context, for example. In this study, we used the single prolonged stress (SPS) animal model of PTSD and fear conditioning procedures to examine the effects of prior traumatic stress on the acquisition, retention, and context-specificity of extinction. SPS administered one week prior to fear conditioning had no effect on the acquisition of fear conditioning or extinction but disrupted the retention of extinction memories for both contextual and cued fear. This SPS effect required a post-stress incubation period to manifest. The results demonstrate that SPS disrupts extinction retention, leading to enhanced fear renewal; further research is needed to identify the neurobiological processes through which SPS induces these effects.
Collapse
Affiliation(s)
- Dayan Knox
- Department of Psychiatry, University of Michigan, Ann Arbor, Michigan 48109, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Silva L, Zhao N, Popp S, Dow-Edwards D. Prenatal tetrahydrocannabinol (THC) alters cognitive function and amphetamine response from weaning to adulthood in the rat. Neurotoxicol Teratol 2011; 34:63-71. [PMID: 22080840 DOI: 10.1016/j.ntt.2011.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 10/19/2011] [Accepted: 10/19/2011] [Indexed: 12/14/2022]
Abstract
Research suggests that not only is marijuana use prevalent among women of reproductive age, but a significant number of women continue to use marijuana and its derivatives throughout pregnancy. Many studies have shown, in both humans and animals, that marijuana exposure during adolescence and adulthood is detrimental to normal cognition and memory. In this study, we examined the effects of daily intravenous injections of 0.15 mg/kg Δ(9)-tetrahydrocannabinol (THC), given to pregnant dams throughout gestation, on cognitive function in the offspring. Offspring were exposed to three tests: a passive avoidance test at postnatal day (PND) 22, an active place avoidance test at PND 45, and an attention task at PND 60, which assessed learning and long-term memory, spatial working memory and prediction, and attention, respectively. Other offspring were also given a 1mg/kg amphetamine challenge at PND 60. Passive avoidance testing showed that prenatal THC had no effect on acquisition but interfered with consolidation during retention testing. The active place avoidance task showed no treatment-related effects on acquisition but a significant treatment effect was observed in reversal performance in males. The attention task showed that a smaller percentage of THC-exposed rats completed the test, although the failure rate of both groups was quite high. Finally, THC exposed animals, both male and female, showed a dampened locomotor response to amphetamine, but females were more active than males overall. These results suggest that prenatal THC exposure has effects on certain aspects of cognitive function in rats from weaning to adulthood. These effects suggest that prenatal marijuana exposure could also alter cognitive function in humans and therefore have an impact on school performance and dampen responses to psychostimulants as well.
Collapse
Affiliation(s)
- Lindsay Silva
- State University of New York, Downstate Medical Center, School of Graduate Studies, Program in Neural and Behavioral Science, NY 11203, USA.
| | | | | | | |
Collapse
|
43
|
Brakel LAW. Extinction phenomena: a biologic perspective on how and why psychoanalysis works. Front Psychol 2011; 2:223. [PMID: 21927610 PMCID: PMC3169790 DOI: 10.3389/fpsyg.2011.00223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2011] [Accepted: 08/23/2011] [Indexed: 11/13/2022] Open
Abstract
This article presents the view that much of the success of classical psychoanalysis is centrally predicated on its biological potency; focusing not on neuropsychology, but on the biology of conditioning. The argument suggests that features of classic psychoanalytic technique - the couch, meetings several times per week with both parties present, and free association - uniquely facilitate intense transferences of various sorts, and that these in turn constitute the multiple and diverse extinction trials necessary to best approximate extinction.
Collapse
Affiliation(s)
- Linda A. W. Brakel
- Department of Psychiatry, University of MichiganAnn Arbor, MI, USA
- Department of Philosophy, University of MichiganAnn Arbor, MI, USA
| |
Collapse
|
44
|
Abstract
This review summarizes the major discussion points of a symposium on stress modulation of cognitive and affective processes, which was held during the 2010 workshop on the neurobiology of stress (Boulder, CO, USA). The four discussants addressed a number of specific cognitive and affective factors that are modulated by exposure to acute or repeated stress. Dr David Morilak discussed the effects of various repeated stress situations on cognitive flexibility, as assessed with a rodent model of attentional set-shifting task, and how performance on slightly different aspects of this test is modulated by different prefrontal regions through monoaminergic neurotransmission. Dr Serge Campeau summarized the findings of several studies exploring a number of factors and brain regions that regulate habituation of various autonomic and neuroendocrine responses to repeated audiogenic stress exposures. Dr Kerry Ressler discussed a body of work exploring the modulation and extinction of fear memories in rodents and humans, especially focusing on the role of key neurotransmitter systems including excitatory amino acids and brain-derived neurotrophic factor. Dr Israel Liberzon presented recent results on human decision-making processes in response to exogenous glucocorticoid hormone administration. Overall, these discussions are casting a wider framework on the cognitive/affective processes that are distinctly regulated by the experience of stress and some of the brain regions and neurotransmitter systems associated with these effects.
Collapse
Affiliation(s)
- Serge Campeau
- Department of Psychology and Neuroscience, University of Colorado at Boulder, Boulder, CO, USA.
| | | | | | | |
Collapse
|
45
|
Schreurs BG, Smith-Bell CA, Burhans LB. Unpaired extinction: implications for treating post-traumatic stress disorder. J Psychiatr Res 2011; 45:638-49. [PMID: 21074779 PMCID: PMC3081978 DOI: 10.1016/j.jpsychires.2010.10.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 10/14/2010] [Accepted: 10/19/2010] [Indexed: 11/19/2022]
Abstract
Extinction of fear is important for treating stress-related conditions particularly post-traumatic stress disorder (PTSD). Although traditional extinction presents the feared stimulus by itself, there is evidence from both clinical and basic research that repeatedly presenting the feared stimulus by itself does not prevent fear from returning. This renewal or relapse can be "thwarted" by unpaired extinction-presentations of the feared stimulus and the event producing the fear. However, no matter how effective standard unpaired extinction may be in the laboratory, repeated presentation of a traumatic event is untenable. To make an unpaired extinction procedure more clinically relevant, we classically conditioned the rabbit nictitating membrane response using electrical stimulation or air puff as the unconditioned stimulus and then during unpaired extinction reduced both the intensity of the unconditioned stimulus and the days of unpaired stimulus presentations. We found unpaired extinction reduced conditioned and exaggerated unconditioned responding (an animal analog of PTSD called conditioning-specific reflex modification) and could be accomplished with a weak unconditioned stimulus as long as extended presentations were used. Surprisingly, brief presentations of a weak unconditioned stimulus or extended presentations of a strong one made the exaggerated responses stronger. One implication is that brief treatment may not just be ineffectual; it may heighten the symptoms of PTSD. Another implication is that using strong stimuli may also heighten those symptoms.
Collapse
Affiliation(s)
- Bernard G Schreurs
- Blanchette Rockefeller Neurosciences Institute and Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV, USA.
| | | | | |
Collapse
|
46
|
Gomes GM, Mello CF, Rosa MMD, Bochi GV, Ferreira J, Barron S, Rubin MA. Polyaminergic agents modulate contextual fear extinction in rats. Neurobiol Learn Mem 2010; 93:589-95. [DOI: 10.1016/j.nlm.2010.02.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 01/21/2010] [Accepted: 02/26/2010] [Indexed: 10/19/2022]
|