1
|
Huang LY, Liu YN, Chen J, Zhu HX, Li LL, Liang ZY, Song JX, Li YJ, Hu ZL, Demon D, Wullaert A, Wang W, Qi SH. Caspase-12 is Expressed in Purkinje Neurons and Prevents Psychiatric-Like Behavior in Mice. Mol Neurobiol 2025; 62:1705-1719. [PMID: 39023795 DOI: 10.1007/s12035-024-04356-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
Caspase-12 is a caspase family member for which functions in regulating cell death and inflammation have previously been suggested. In this study, we used caspase-12 lacZ reporter mice to elucidate the expression pattern of caspase-12 in order to obtain an idea about its possible in vivo function. Strikingly, these reporter mice showed that caspase-12 is expressed explicitly in Purkinje neurons of the cerebellum. As this observation suggested a function for caspase-12 in Purkinje neurons, we analyzed the brain and behavior of caspase-12 deficient mice in detail. Extensive histological analyses showed that caspase-12 was not crucial for establishing cerebellum structure or for maintaining Purkinje cell numbers. We then performed behavioral tests to investigate whether caspase-12 deficiency affects memory, motor, and psychiatric functions in mice. Interestingly, while the absence of caspase-12 did not affect memory and motor function, caspase-12 deficient mice showed depression and hyperactivity tendencies, together resembling manic behavior. Next, suggesting a possible molecular mechanistic explanation, we showed that caspase-12 deficient cerebella harbored diminished signaling through the brain-derived neurotrophic factor/tyrosine kinase receptor B/cyclic-AMP response binding protein axis, as well as strongly enhanced expression of the neuronal activity marker c-Fos. Thus, our study establishes caspase-12 expression in mouse Purkinje neurons and opens novel avenues of research to investigate the role of caspase-12 in regulating psychiatric behavior.
Collapse
Affiliation(s)
- Lin-Yan Huang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
- VIB Center for Inflammation Research, Zwijnaarde, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Yi-Ning Liu
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Jie Chen
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
- Department of Laboratory Medicine, The Affiliated Hospital of Xuzhou Medical University, Huaihai West Road 99, Xuzhou, 221002, China
| | - Hai-Xue Zhu
- Department of Ophthalmology, The Affiliated Huai'an Hospital of Xuzhou Medical University, No.62 Huaihai South Road, Huai'an, 223001, Jiangsu, People's Republic of China
| | - Li-Li Li
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Zhi-Yan Liang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Jin-Xiu Song
- Department of Pharmacology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Yu-Jie Li
- Department of Clinical Laboratory, Kunshan First People's Hospital, Kunshan, Jiangsu, 215300, People's Republic of China
| | - Zhao-Li Hu
- Research Center for Biochemistry and Molecular Biology and Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Dieter Demon
- VIB Center for Inflammation Research, Zwijnaarde, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Andy Wullaert
- VIB Center for Inflammation Research, Zwijnaarde, Belgium
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
- Cell Death Signaling Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Wan Wang
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Su-Hua Qi
- School of Medical Technology, Xuzhou Key Laboratory of Laboratory Diagnostics, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China.
| |
Collapse
|
2
|
Wang H, He Y, Tang J, Liu Y, Wu C, Li C, Sun H, Sun L. (2R, 6R)-hydroxynorketamine ameliorates PTSD-like behaviors during the reconsolidation phase of fear memory in rats by modulating the VGF/BDNF/GluA1 signaling pathway in the hippocampus. Behav Brain Res 2025; 476:115273. [PMID: 39326635 DOI: 10.1016/j.bbr.2024.115273] [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: 06/04/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 09/28/2024]
Abstract
RATIONALE Fear memory, a fundamental symptom of post-traumatic stress disorder (PTSD), is improved by (2R, 6R)-hydroxynorketamine ((2R, 6R)-HNK) administration. However, the phase of fear memory in which the injected drug is the most effective at mitigating PTSD-like effects remains unknown. OBJECTIVE This study aimed to explore the effects of (2 R, 6 R)-HNK administration during three phases [acquisition (AP), reconsolidation (RP), and extinction (EP)] on PTSD-like behaviors in single prolonged stress (SPS) and contextual fear conditioning (CFC) rat models. The effects of VGF-inducible type of nerve growth factor (VGF), brain-derived neurotrophic factor (BDNF), and GluA1 on hippocampus (HIP) expression were also explored. METHODS SPS and CFC (SPSC) were used to establish a PTSD rat model. After lateral ventricle injection of 5 μL (2 R, 6 R)-HNK (0.5 nmol). Anxiety-depression-like behaviors were assessed in rats by the open field test (OFT) and elevated plus maze test (EPMT). Situational fear responses were evaluated in rodents by freezing behavior test (FBT) test. In addition, GluA1, VGF, and BDNF were assessed in the hippocampus by Western blot assay (WB) and Immunohistochemistry assay (IF). RESULTS SPSC procedure induced PTSD-like behaviors. The SPSC group had decreased spontaneous exploratory behavior and increased fear response. The (2R, 6R)-HNK group showed improved SPSC-induced reduction in GluA1, VGF, and BDNF levels in the HIP. During RP, anxiety and fear avoidance behaviors were alleviated, and the protein levels of GluA1, VGF, and BDNF in the HIP were restored. In contrast, no significant improvement was noted during AP and EP. CONCLUSIONS (2R,6R)-HNK modulates the VGF/BDNF/GluA1 signaling pathway in the hippocampus and improves PTSD-like behaviors during the reconsolidation phase of fear memory in rats, which may provide a new target for the clinical treatment and prevention of fear-related disorders such as PTSD.
Collapse
Affiliation(s)
- Han Wang
- School of Mental Health, Jining Medical University, Jining, Shandong 272067, China; School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yuxuan He
- Department of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Jiahao Tang
- Department of Clinical Medicine, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Yang Liu
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Chunyan Wu
- Department of Neurology, Affiliated Hospital of Shandong Second Medical University, Weifang 261031, China
| | - Changjiang Li
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Hongwei Sun
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China
| | - Lin Sun
- School of Psychology, Shandong Second Medical University, 7166# Baotong West Street, Weifang, Shandong 261053, PR China.
| |
Collapse
|
3
|
Lucon-Xiccato T, Savaşçı BB, Merola C, Benedetti E, Caioni G, Aliko V, Bertolucci C, Perugini M. Environmentally relevant concentrations of triclocarban affect behaviour, learning, and brain gene expression in fish. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 903:166717. [PMID: 37657536 DOI: 10.1016/j.scitotenv.2023.166717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
Many chemicals spilled in aquatic ecosystems can interfere with cognitive abilities and brain functions that control fitness-related behaviour. Hence, their harmful potential may be substantially underestimated. Triclocarban (TCC), one of the most common aquatic contaminants, is known to disrupt hormonal activity, but the consequences of this action on behaviour and its underlying cognitive mechanisms are unclear. We tried to fill this knowledge gap by analysing behaviour, cognitive abilities, and brain gene expression in zebrafish larvae exposed to TCC sublethal concentrations. TCC exposure substantially decreased exploratory behaviour and response to stimulation, while it increased sociability. Additionally, TCC reduced the cognitive performance of zebrafish in a habituation learning task. In the brain of TCC-exposed zebrafish, we found upregulation of c-fos, a gene involved in neural activity, and downregulation of bdnf, a gene that influences behavioural and cognitive traits such as activity, learning, and memory. Overall, our experiments highlight consistent effects of non-lethal TCC concentrations on behaviour, cognitive abilities, and brain functioning in a teleost fish, suggesting critical fitness consequences of these compounds in aquatic ecosystems as well as the potential to affect human health.
Collapse
Affiliation(s)
- Tyrone Lucon-Xiccato
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.
| | - Beste Başak Savaşçı
- Unit of Evolutionary Biology/Systematic Zoology, Institute for Biochemistry and Biology, University of Potsdam, Potsdam, Germany; Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Carmine Merola
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Giulia Caioni
- Department of Life, Health and Environmental Sciences, University of L'Aquila, L'Aquila, Italy
| | - Valbona Aliko
- Department of Biology, Faculty of Natural Sciences, University of Tirana, Tirana, Albania
| | - Cristiano Bertolucci
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Monia Perugini
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Teramo, Italy
| |
Collapse
|
4
|
Zhao H, Li Y, Luo T, Chou W, Sun T, Liu H, Qiu H, Zhu D, Chen D, Gu Y. Preventing Post-Traumatic Stress Disorder (PTSD) in rats with pulsed 810 nm laser transcranial phototherapy. Transl Psychiatry 2023; 13:281. [PMID: 37580354 PMCID: PMC10425462 DOI: 10.1038/s41398-023-02583-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 08/16/2023] Open
Abstract
Post-traumatic stress disorder (PTSD) is a debilitating condition that occurs following exposure to traumatic events. Current treatments, such as psychological debriefing and pharmacotherapy, often have limited efficacy and may result in unwanted side effects, making early intervention is a more desirable strategy. In this study, we investigated the efficacy of a single dose of pulsed (10 Hz) 810 nm laser-phototherapy (P-PT) as an early intervention for preventing PTSD-like comorbidities in rats induced by single inescapable electric foot shock following the single prolonged stress (SPS&S). As indicated by the results of the open filed test, elevated plus maze test, and contextual fear conditioning test, P-PT prevented the development of anxiety and freezing behaviors in rats exposed to the SPS&S. We also compared the effects of P-PT and continuous wave 810 nm laser-phototherapy (CW-PT) in preventing PTSD-like comorbidities in rats. The results revealed that P-PT was effective in preventing both freezing and anxiety behavior in stressed rats. In contrast, CW-PT only had a preventive effect on freezing behavior but not anxiety. Additionally, P-PT significantly reduced the c-fos expression in cingulate cortex area 1(Cg1) and infralimbic cortex (IL) of stressed rats, while CW-PT had no significant effects on c-fos expression. Taken together, our results demonstrate that P-PT is a highly effective strategy for preventing the occurrence of PTSD-like comorbidities in rats.
Collapse
Affiliation(s)
- Hongyou Zhao
- School of Medical Technology, Beijing Institute of Technology, Beijing, China.
| | - Yi Li
- Department of Laser Medicine, the First Medical Center of the PLA General Hospital, Beijing, China
| | - Ting Luo
- Moores Cancer Center, University of California San Diego, San Diego, USA
| | - Wenxin Chou
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Tianzhen Sun
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Haolin Liu
- No.965 Hospital, Joint Logistics Support Force of Chinese PLA, Jilin, China
| | - Haixia Qiu
- Department of Laser Medicine, the First Medical Center of the PLA General Hospital, Beijing, China
| | - Dan Zhu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| | - Defu Chen
- School of Medical Technology, Beijing Institute of Technology, Beijing, China.
| | - Ying Gu
- Department of Laser Medicine, the First Medical Center of the PLA General Hospital, Beijing, China.
| |
Collapse
|
5
|
Boutros SW, Zimmerman B, Nagy SC, Unni VK, Raber J. Age, sex, and apolipoprotein E isoform alter contextual fear learning, neuronal activation, and baseline DNA damage in the hippocampus. Mol Psychiatry 2023; 28:3343-3354. [PMID: 36732588 PMCID: PMC10618101 DOI: 10.1038/s41380-023-01966-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 01/06/2023] [Accepted: 01/16/2023] [Indexed: 02/04/2023]
Abstract
Age, female sex, and apolipoprotein E4 (E4) are risk factors to develop Alzheimer's disease (AD). There are three major human apoE isoforms: E2, E3, and E4. Compared to E3, E4 increases while E2 decreases AD risk. However, E2 is associated with increased risk and severity of post-traumatic stress disorder (PTSD). In cognitively healthy adults, E4 carriers have greater brain activation during learning and memory tasks in the absence of behavioral differences. Human apoE targeted replacement (TR) mice display differences in fear extinction that parallel human data: E2 mice show impaired extinction, mirroring heightened PTSD symptoms in E2 combat veterans. Recently, an adaptive role of DNA double strand breaks (DSBs) in immediate early gene expression (IEG) has been described. Age and disease synergistically increase DNA damage and decrease DNA repair. As the mechanisms underlying the relative risks of apoE, sex, and their interactions in aging are unclear, we used young (3 months) and middle-aged (12 months) male and female TR mice to investigate the influence of these factors on DSBs and IEGs at baseline and following contextual fear conditioning. We assessed brain-wide changes in neural activation following fear conditioning using whole-brain cFos imaging in young female TR mice. E4 mice froze more during fear conditioning and had lower cFos immunoreactivity across regions important for somatosensation and contextual encoding compared to E2 mice. E4 mice also showed altered co-activation compared to E3 mice, corresponding to human MRI and cognitive data, and indicating that there are differences in brain activity and connectivity at young ages independent of fear learning. There were increased DSB markers in middle-aged animals and alterations to cFos levels dependent on sex and isoform, as well. The increase in hippocampal DSB markers in middle-aged animals and female E4 mice may play a role in the risk for developing AD.
Collapse
Affiliation(s)
- Sydney Weber Boutros
- Department of Behavioral Neuroscience, OHSU, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Department of Psychological Sciences, Boise State University, 2133 W Cesar Chavez Ln, Boise, ID, 83725, USA
| | - Benjamin Zimmerman
- Department of Behavioral Neuroscience, OHSU, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Advanced Imaging Research Center, OHSU, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Helfgott Research Institute, NUNM, 2201 SW First Avenue, Portland, OR, 97201, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, 405 N, Matthews Avenue, Urbana, IL 61801, USA
| | - Sydney C Nagy
- Department of Behavioral Neuroscience, OHSU, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Vivek K Unni
- Department of Neurology, OHSU, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
- Jungers Center for Neurosciences Research, OHSU; and OHSU Parkinson Center, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, OHSU, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
- Department of Neurology, OHSU, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
- Departments of Psychiatry and Radiation Medicine, OHSU, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
- Division of Neuroscience, ONPRC, 505 NW 185th Ave, Beaverton, OR, 97006, USA.
| |
Collapse
|
6
|
Wu S, Ning K, Wang Y, Zhang L, Liu J. Up-regulation of BDNF/TrkB signaling by δ opioid receptor agonist SNC80 modulates depressive-like behaviors in chronic restraint-stressed mice. Eur J Pharmacol 2023; 942:175532. [PMID: 36708979 DOI: 10.1016/j.ejphar.2023.175532] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/18/2023] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
Depressive disorder is a psychiatric disease characterized by its main symptoms of low mood and anhedonia. Due to its complex etiology, current clinical treatments for depressive disorder are limited. In this study, we assessed the role of the δ opioid receptor (δOR) system in the development of chronic-restraint-stressed (CRS)-induced depressive behaviors. We employed a 21-day CRS model and detected the c-fos activation and protein levels' changes in enkephalin (ENK)/δOR. It was found that the hippocampus and amygdala were involved in CRS-induced depression. The expression of pro-enkephalin (PENK), the precursors of the endogenous ligand for δOR, was significantly decreased in the hippocampus and amygdala following CRS. We then treated the mice with SNC80, a specific δOR agonist, to examine its anti-depressant effects in the tail suspension test (TST), forced swimming test (FST), and sucrose preference test (SPT). SNC80 administration significantly reversed depressive-like behaviors, and this antidepressant effect could be blocked by a TrkB inhibitor: ANA-12. Although ANA-12 treatment had no significant effect on the expression of ENK/δOR, it blocked the promoting effects of brain-derived neurotrophic factor (BDNF)/tyrosine kinase B(TrkB) signaling by SNC80 in the hippocampus and amygdala. Therefore, the present study demonstrates that SNC80 exerts anti-depressant effects by up-regulating the BDNF/TrkB signaling pathway in the hippocampus and amygdala in CRS-induced depression and provides evidence that δOR's agonists may be potential anti-depressant therapeutic agents.
Collapse
Affiliation(s)
- Shuo Wu
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China; Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Kuan Ning
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yujun Wang
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Lesha Zhang
- Department of Physiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Jinggen Liu
- Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| |
Collapse
|
7
|
The Medial Prefrontal Cortex, Nucleus Accumbens, Basolateral Amygdala, and Hippocampus Regulate the Amelioration of Environmental Enrichment and Cue in Fear Behavior in the Animal Model of PTSD. Behav Neurol 2022; 2022:7331714. [PMID: 35178125 PMCID: PMC8843982 DOI: 10.1155/2022/7331714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/07/2022] [Accepted: 01/27/2022] [Indexed: 11/21/2022] Open
Abstract
A growing body of evidence showed that environmental enrichment (EE) ameliorated footshock-induced fear behavior of posttraumatic stress disorder (PTSD). However, no research comprehensively tested the effect of EE, cue, and the combination of EE and cue in footshock-induced fear behavior of PTSD symptoms. The present study addressed this issue and examined whether the medial prefrontal cortex (mPFC, including the cingulate cortex 1 (Cg1), prelimbic cortex (PrL), and infralimbic cortex (IL)), the nucleus accumbens (NAc), the basolateral amygdala (BLA), and the hippocampus (e.g., CA1, CA3, and dentate gyrus (DG)) regulated the amelioration of the EE, cue, or the combination of EE and cue. The results showed that EE or cue could reduce fear behavior. The combination of EE and cue revealed a stronger decrease in fear behavior. The cue stimulus may play an occasion setting or a conditioned stimulus to modulate the reduction in fear behavior induced by footshock. Regarding the reduction of the EE in fear behavior, the Cg1 and IL of the mPFC and the NAc upregulated the c-Fos expression; however, the BLA downregulated the c-Fos expression. The mPFC (i.e., the Cg1, PrL, and IL) and the hippocampus (i.e., the CA1, CA3, and DG) downregulated the c-Fos expression in the suppression of the cue in fear behavior. The interaction of EE and cue in reduction of fear behavior occurred in the Cg1 and NAc for the c-Fos expression. The data of c-Fos mRNA were similar to the findings of the c-Fos protein expression. These findings related to the EE and cue modulations in fear behavior may develop a novel nonpharmacological treatment in PTSD.
Collapse
|
8
|
Ko MJ, Chiang T, Mukadam AA, Mulia GE, Gutridge AM, Lin A, Chester JA, van Rijn RM. β-Arrestin-dependent ERK signaling reduces anxiety-like and conditioned fear-related behaviors in mice. Sci Signal 2021; 14:14/694/eaba0245. [PMID: 34344831 DOI: 10.1126/scisignal.aba0245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
G protein-coupled receptors (GPCRs) are implicated in the regulation of fear and anxiety. GPCR signaling involves canonical G protein pathways but can also engage downstream kinases and effectors through scaffolding interactions mediated by β-arrestin. Here, we investigated whether β-arrestin signaling regulates anxiety-like and fear-related behavior in mice in response to activation of the GPCR δ-opioid receptor (δOR or DOR). Administration of β-arrestin-biased δOR agonists to male C57BL/6 mice revealed β-arrestin 2-dependent activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in the dorsal hippocampus and amygdala and β-arrestin 1-dependent activation of ERK1/2 in the nucleus accumbens. In mice, β-arrestin-biased agonist treatment was associated with reduced anxiety-like and fear-related behaviors, with some overlapping and isoform-specific input. In contrast, applying a G protein-biased δOR agonist decreased ERK1/2 activity in all three regions as well as the dorsal striatum and was associated with increased fear-related behavior without effects on baseline anxiety. Our results indicate a complex picture of δOR neuromodulation in which β-arrestin 1- and 2-dependent ERK signaling in specific brain subregions suppresses behaviors associated with anxiety and fear and opposes the effects of G protein-biased signaling. Overall, our findings highlight the importance of noncanonical β-arrestin-dependent GPCR signaling in the regulation of these interrelated emotions.
Collapse
Affiliation(s)
- Mee Jung Ko
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA
| | - Terrance Chiang
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Arbaaz A Mukadam
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Department of Psychological Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Grace E Mulia
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA
| | - Anna M Gutridge
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA.,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA
| | - Angel Lin
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Julia A Chester
- Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA.,Department of Psychological Sciences, College of Health and Human Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA. .,Purdue Institute for Integrative Neuroscience, West Lafayette, IN 47907, USA.,Purdue Interdisciplinary Life Sciences Graduate Program, West Lafayette, IN 47907, USA.,Purdue Institute for Drug Discovery, West Lafayette, IN 47907, USA
| |
Collapse
|
9
|
Gupta A, Gullapalli S, Pan H, Ramos-Ortolaza DL, Hayward MD, Low MJ, Pintar JE, Devi LA, Gomes I. Regulation of Opioid Receptors by Their Endogenous Opioid Peptides. Cell Mol Neurobiol 2021; 41:1103-1118. [PMID: 33389463 PMCID: PMC8277103 DOI: 10.1007/s10571-020-01015-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022]
Abstract
Activation of μ, δ, and κ opioid receptors by endogenous opioid peptides leads to the regulation of many emotional and physiological responses. The three major endogenous opioid peptides, β-endorphin, enkephalins, and dynorphins result from the processing of three main precursors: proopiomelanocortin, proenkephalin, and prodynorphin. Using a knockout approach, we sought to determine whether the absence of endogenous opioid peptides would affect the expression or activity of opioid receptors in mice lacking either proenkephalin, β-endorphin, or both. Since gene knockout can lead to changes in the levels of peptides generated from related precursors by compensatory mechanisms, we directly measured the levels of Leu-enkephalin and dynorphin-derived peptides in the brain of animals lacking proenkephalin, β-endorphin, or both. We find that whereas the levels of dynorphin-derived peptides were relatively unaltered, the levels of Leu-enkephalin were substantially decreased compared to wild-type mice suggesting that preproenkephalin is the major source of Leu-enkephalin. This data also suggests that the lack of β-endorphin and/or proenkephalin does not lead to a compensatory change in prodynorphin processing. Next, we examined the effect of loss of the endogenous peptides on the regulation of opioid receptor levels and activity in specific regions of the brain. We also compared the receptor levels and activity in males and females and show that the lack of β-endorphin and/or proenkephalin leads to differential modulation of the three opioid receptors in a region- and gender-specific manner. These results suggest that endogenous opioid peptides are important modulators of the expression and activity of opioid receptors in the brain.
Collapse
Affiliation(s)
- Achla Gupta
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA
| | - Srinivas Gullapalli
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA
- Emcure Pharmaceuticals, Mumbai, India
| | - Hui Pan
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA
- University of Southern California Medical Center, Los Angeles, CA, USA
| | - Dinah L Ramos-Ortolaza
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA
- Pontifico Catholic Univ. Puerto Rico, Ponce, Puerto Rico
| | - Michael D Hayward
- Vollum Institute, Oregon Health Sciences University, Portland, OR, 97201, USA
- Invivotek, Trenton, NJ, USA
| | - Malcom J Low
- Vollum Institute, Oregon Health Sciences University, Portland, OR, 97201, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - John E Pintar
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - Lakshmi A Devi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA.
| | - Ivone Gomes
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY, 10029, USA.
| |
Collapse
|
10
|
Chuang HG, Aziz NHA, Wong JH, Mustapha M, Abdullah JM, Idris Z, Abdullah Z, Alrafiah A, Muthuraju S. Role of toll-like receptor 4 antagonist Lipopolysaccharide-Rhodobacter sphaeroides on acute stress-induced voluntary ethanol preference and drinking behaviour: In vivo Swiss Albino mouse model. Eur Neuropsychopharmacol 2021; 45:59-72. [PMID: 32014377 DOI: 10.1016/j.euroneuro.2019.12.121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 06/06/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022]
Abstract
The present study focused on investigating the effect of toll-like receptor 4 (TLR4) antagonist Lipopolysaccharide-Rhodobacter sphaeroides(LPS-RS) on acute, stress-induced voluntary ethanol preference and drinking behaviour, neuronal components activation, and gene expression associated with stress and addictive behaviour. This study involved the exposure of restraint stress and social isolation using Swiss Albino mice. Two-bottle choice ethanol preference analysis was used in the evaluation of voluntary ethanol seeking and drinking behaviour. Several behavioural assessments were carried out to assess fear and anxiety-like behaviour, neuromuscular ability, motor coordination and locomotion. Morphological and immunoreactivity analysis and gene expression analysis were done after the completion of behavioural assessments. TLR4 antagonist LPS-RS treated stressed-mice showed a significant decrease in ethanol drinking compared with stressed mice. Behavioural results showed that stress exposure induced fear and anxiety-like behaviour; however; no significant deficit was found on motor coordination, neuromuscular ability, locomotion and exploratory behaviour among groups. Morphological analysis showed no significant change in the prefrontal cortex and hippocampus among all groups, while immunoreactivity analysis showed higher expression of c-Fos in prefrontal cortex and hippocampus, higher TLR4 expression in the prefrontal cortex and glial fibrillary acidic protein (GFAP) in hippocampus among stressed-animals. Stressed-mice also showed significant increase in TLR4, Nuclear Factor-Kappa B (NF-kB), inducible nitric oxide synthase (iNOS), dopamine receptor D2 (DRD2), cyclic adenosine monophosphate (cAMP) response element binding protein-1 (CREB-1) and opioid receptor MU-1 (OPRM-1) genes expression compared with control and LPS-RS treated stressed-mice. As a conclusion, the antagonism of TLR4 could provide therapeutic value in the treatment of stress-induced addiction.
Collapse
Affiliation(s)
- Huei Gau Chuang
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - NurNaznee Hirni Abd Aziz
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Jia Hui Wong
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Muzaimi Mustapha
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Jafri Malin Abdullah
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Zamzuri Idris
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Zuraidah Abdullah
- Biomedicine Program, School of Health Science, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia
| | - Aziza Alrafiah
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King AbdulAziz University, Jeddah, Saudi Arabia
| | - Sangu Muthuraju
- Integrated Neuroscience Program (INP), Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Neurosciences, Brain and Behaviour Cluster, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kubang Kerian, Kota Bharu, Kelantan 16150, Malaysia; Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston 77240, TX, USA.
| |
Collapse
|
11
|
BDNF Protein and BDNF mRNA Expression of the Medial Prefrontal Cortex, Amygdala, and Hippocampus during Situational Reminder in the PTSD Animal Model. Behav Neurol 2021; 2021:6657716. [PMID: 33763156 PMCID: PMC7964114 DOI: 10.1155/2021/6657716] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 11/17/2022] Open
Abstract
Whether BDNF protein and BDNF mRNA expression of the medial prefrontal cortex (mPFC; cingulated cortex area 1 (Cg1), prelimbic cortex (PrL), and infralimbic cortex (IL)), amygdala, and hippocampus (CA1, CA2, CA3, and dentate gyrus (DG)) was involved in fear of posttraumatic stress disorder (PTSD) during the situational reminder of traumatic memory remains uncertain. Footshock rats experienced an inescapable footshock (3 mA, 10 s), and later we have measured fear behavior for 2 min in the footshock environment on the situational reminder phase. In the final retrieval of situational reminder, BDNF protein and mRNA levels were measured. The results showed that higher BDNF expression occurred in the Cg1, PrL, and amygdala. Lower BDNF expression occurred in the IL, CA1, CA2, CA3, and DG. BDNF mRNA levels were higher in the mPFC and amygdala but lower in the hippocampus. The neural connection analysis showed that BDNF protein and BDNF mRNA exhibited weak connections among the mPFC, amygdala, and hippocampus during situational reminders. The present data did not support the previous viewpoint in neuroimaging research that the mPFC and hippocampus revealed hypoactivity and the amygdala exhibited hyperactivity for PTSD symptoms. These findings should be discussed with the previous evidence and provide clinical implications for PTSD.
Collapse
|
12
|
Yi Y, Liu Y, Wu K, Wu W, Zhang W. The core genes involved in the promotion of depression in patients with ovarian cancer. Oncol Lett 2019; 18:5995-6007. [PMID: 31788074 PMCID: PMC6865084 DOI: 10.3892/ol.2019.10934] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 08/08/2019] [Indexed: 12/09/2022] Open
Abstract
The present study aimed to identify the core genes and pathways involved in depression in patients with ovarian cancer (OC) who suffer from high or low-grade depression. The dataset GSE9116 from Gene Expression Omnibus database was analyzed to identify differentially expressed genes (DEGs) in these patients. To elucidate how certain genes could promote depression in patients with OC, pathway crosstalk, protein-protein interaction (PPI) and comprehensive gene-pathway analyses were determined using WebGestalt, ToppGene and Search Tool for the Retrieval of Interacting Genes and gene ontology analysis. Key genes and pathways were extracted from the gene-pathway network, and gene expression and survival analysis were evaluated. A total of 93 DEGs were identified from GSE9116 dataset, including 84 upregulated genes and nine downregulated genes. The PPI, pathway crosstalk and comprehensive gene-pathway analyses highlighted C-C motif chemokine ligand 2 (CCL2), Fos proto-oncogene, AP-1 transcription factor subunit (FOS), serpin family E member 1 (SERPINE1) and serpin family G member 1 (SERPING1) as core genes involved in the promotion of depression in patients with OC. These core genes were involved in the following four pathways 'Ensemble of genes encoding ECM-associated proteins including ECM-affiliated proteins', 'ECM regulators and secreted factors', 'Ensemble of genes encoding extracellular matrix and extracellular matrix-associated proteins' and 'MAPK signaling pathway and IL-17 signaling pathway'. The results from gene expression and survival analysis demonstrated that these four key genes were upregulated in patients with OC and high-grade depression and could worsen patients' survival. These results suggested that CCL2, FOS, SERPINE1 and SERPING1 may serve a crucial role in the promotion of depression in patients with OC. This finding may provide novel markers for predicting and treating depression in patients with OC; however, the underlying mechanisms remain unknown and require further investigation.
Collapse
Affiliation(s)
- Yuexiong Yi
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Yanyan Liu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Kejia Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wanrong Wu
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
13
|
Yun H, Park ES, Choi S, Shin B, Yu J, Yu J, Amarasekara DS, Kim S, Lee N, Choi JS, Choi Y, Rho J. TDAG51 is a crucial regulator of maternal care and depressive-like behavior after parturition. PLoS Genet 2019; 15:e1008214. [PMID: 31251738 PMCID: PMC6599150 DOI: 10.1371/journal.pgen.1008214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 05/27/2019] [Indexed: 12/11/2022] Open
Abstract
Postpartum depression is a severe emotional and mental disorder that involves maternal care defects and psychiatric illness. Postpartum depression is closely associated with a combination of physical changes and physiological stress during pregnancy or after parturition in stress-sensitive women. Although postpartum depression is relatively well known to have deleterious effects on the developing fetus, the influence of genetic risk factors on the development of postpartum depression remains unclear. In this study, we discovered a novel function of T cell death-associated gene 51 (TDAG51/PHLDA1) in the regulation of maternal and depressive-like behavior. After parturition, TDAG51-deficient dams showed impaired maternal behavior in pup retrieving, nursing and nest building tests. In contrast to the normal dams, the TDAG51-deficient dams also exhibited more sensitive depressive-like behaviors after parturition. Furthermore, changes in the expression levels of various maternal and depressive-like behavior-associated genes regulating neuroendocrine factor and monoamine neurotransmitter levels were observed in TDAG51-deficient postpartum brain tissues. These findings indicate that TDAG51 plays a protective role against maternal care defects and depressive-like behavior after parturition. Thus, TDAG51 is a maternal care-associated gene that functions as a crucial regulator of maternal and depressive-like behavior after parturition. Postpartum depression is a severe emotional and mental disease that can affect women typically after parturition. However, the genetic risk factors associated with the development of postpartum depression are still largely unknown. We discovered a novel function of T cell death-associated gene 51 (TDAG51) in the regulation of maternal behavior and postpartum depression. We report that TDAG51 deficiency induces depressive-like and abnormal maternal behavior after parturition. The loss of TDAG51 in postpartum brain tissues induces changes in the expression levels of various maternal and depressive-like behavior-associated genes that regulate the levels of neuroendocrine factors and monoamine neurotransmitters. TDAG51 is a maternal care-associated gene that functions as a crucial regulator of maternal and depressive-like behavior after parturition.
Collapse
Affiliation(s)
- Hyeongseok Yun
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Eui-Soon Park
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Seunga Choi
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Bongjin Shin
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jungeun Yu
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jiyeon Yu
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | | | - Sumi Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Nari Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
| | - Jong-Soon Choi
- Division of Life Science, Korea Basic Science Institute, Daejeon, Korea
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Korea
- * E-mail:
| |
Collapse
|
14
|
Huang CC, Kuo SC, Yeh TC, Yeh YW, Chen CY, Liang CS, Tsou CC, Lin CL, Ho PS, Huang SY. OPRD1 gene affects disease vulnerability and environmental stress in patients with heroin dependence in Han Chinese. Prog Neuropsychopharmacol Biol Psychiatry 2019; 89:109-116. [PMID: 30171993 DOI: 10.1016/j.pnpbp.2018.08.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/24/2018] [Accepted: 08/25/2018] [Indexed: 12/24/2022]
Abstract
Exposure to stress not only increases the vulnerability to heroin dependence (HD) but also provokes relapse. The etiology of HD and the role of life stress remain unclear, but prior studies suggested that both genetic and environmental factors are important. Opioid related genes, including OPRM1, OPRD1, OPRK1, and POMC, are obvious candidates for HD. Therefore, this study was conducted to explore whether the genetic polymorphisms of the candidates could affect vulnerability to HD and response to life stress in patients with HD. Ten polymorphisms of the opioid related genes were analyzed in 801 patients and 530 controls. The Life Event Questionnaire was used to assess the perspective and response to life stress in the past year. The genotype distribution and allelic frequency analyses showed that the minor C allele of rs2234918 in OPRD1 is over-represented in the HD group (P = .006 and P = .002, respectively). This finding was further confirmed by logistic regression analysis, showing that C allele carriers have a 1.42 times greater risk for HD compared to T/T homozygotes. A subgroup of 421 patients and 135 controls were eligible for life stress assessment. Patients with HD have a higher occurrence of negative events (No), negative events score (Ns), and average negative event score (Na) than those of controls (all P < .001), but there was no difference regarding positive recent events between the two groups. Gene-stress assessment in the HD group showed that T/T homozygotes of OPRD1 rs2236857 have more severe stress than C allele carriers (Ns, P = .004 and Na, P = .047). Our results indicate that the OPRD1 gene may not only play a role in the pathogenesis of HD but also affect the response to life stress among patients with HD in our Han Chinese population. Patients with the risk genotype may need additional psychosocial intervention for relapse prevention.
Collapse
Affiliation(s)
- Chang-Chih Huang
- Department of Psychiatry, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Shin-Chang Kuo
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan
| | - Ta-Chuan Yeh
- Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan
| | - Yi-Wei Yeh
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan
| | - Chun-Yen Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan
| | - Chih-Sung Liang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, Taipei, Taiwan
| | - Chang-Chih Tsou
- Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan
| | - Chun-Long Lin
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Department of Psychiatry, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan
| | - Pei-Shen Ho
- Department of Psychiatry, Beitou Branch, Tri-Service General Hospital, Taipei, Taiwan
| | - San-Yuan Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan; Department of Psychiatry, Tri-Service General Hospital, Taipei, Taiwan.
| |
Collapse
|
15
|
Endocannabinoid interactions in the regulation of acquisition of contextual conditioned fear. Prog Neuropsychopharmacol Biol Psychiatry 2019; 90:84-91. [PMID: 30458201 DOI: 10.1016/j.pnpbp.2018.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 11/14/2018] [Accepted: 11/15/2018] [Indexed: 11/23/2022]
Abstract
Endocannabinoids (eCBs) anandamide (AEA) and 2-arachidonoylglycerol (2-AG) were shown to be involved in the basis of trauma-induced behavioral changes, particularly contextual conditioned fear, however, their ligand-specific effects and possible interactions are poorly understood. Here we assessed specific eCB effects and interactions on acquisition of contextual conditioned fear employing electric footshocks in a rat model. We selectively increased eCB levels by pharmacological blockade of the degrading enzymes of AEA by URB597 and 2-AG by JZL184 before traumatization either systemically or locally in relevant brain areas, the prelimbic cortex (PrL), ventral hippocampus (vHC) and basolateral amygdala (BLA). Following traumatization, a series of contextual reminders were conducted during which conditioned fear was assessed. While systemic URB597-treatment during traumatization only slightly enhanced the acquisition of contextual conditioned fear, administration of the compound in the PrL and vHC led to the acquisition of stable, lasting conditioned fear, resistant to extinction. These effects of URB597 were blocked by simultaneous administration of JZL184. Similar treatment effects did not occur in the BLA. Treatment effects were not secondary to alterations in locomotor activity or nociception. Our findings suggest that AEA and 2-AG functionally interact in the regulation of acquisition of contextual conditioned fear. AEA signaling in the PrL and vHC is a crucial promoter of fear acquisition while 2-AG potentially modulates this effect. The lack of eCB effects in the BLA suggests functional specificity of eCBs at distinct brain sites.
Collapse
|
16
|
Wu YP, Gao HY, Ouyang SH, Kurihara H, He RR, Li YF. Predator stress-induced depression is associated with inhibition of hippocampal neurogenesis in adult male mice. Neural Regen Res 2019; 14:298-305. [PMID: 30531013 PMCID: PMC6301170 DOI: 10.4103/1673-5374.244792] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Stress has been suggested to disturb the 5-hydroxytryptamine system and decrease neurogenesis, which contribute to the development of depression. Few studies have investigated the effect of predator stress, a type of psychological stress, on depression and hippocampal neurogenesis in adult mice; we therefore investigated this in the present study. A total of 35 adult male Kunming mice were allocated to a cat stress group, cat odor stress group, cat stress + fluoxetine group, cat odor stress + fluoxetine group, or a control group (no stress/treatment). After 12 days of cat stress or cat odor stress, behavioral correlates of depression were measured using the open field test, elevated plus maze test, and dark-avoidance test. The concentrations of hippocampal 5-hydroxytryptamine and 5-hydroxyindoleacetic acid were measured using high-performance liquid chromatography-electrochemical detection. Neurogenesis was also analyzed using a bromodeoxyuridine and doublecortin double-immunostaining method. Cat stress and cat odor stress induced depression-like behaviors; this effect was stronger in the cat stress model. Furthermore, compared with the control group, cat stress mice exhibited lower 5-hydroxytryptamine concentrations, higher 5-hydroxyindoleacetic acid concentrations, and significantly fewer bromodeoxyuridine+/doublecortin+-labeled cells in the dentate gyrus, which was indicative of less neurogenesis. The changes observed in the cat stress group were not seen in the cat stress + fluoxetine group, which suggests that the effects of predator stress on depression and neurogenesis were reversed by fluoxetine. Taken together, our results indicate that depression-like behaviors induced by predator stress are associated with the inhibition of hippocampal neurogenesis.
Collapse
Affiliation(s)
- Yan-Ping Wu
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Hua-Ying Gao
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Shu-Hua Ouyang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Hiroshi Kurihara
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Rong-Rong He
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| | - Yi-Fang Li
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research; Anti-Stress and Health Research Center, College of Pharmacy, Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
17
|
Seo JH. Treadmill exercise alleviates stress-induced anxiety-like behaviors in rats. J Exerc Rehabil 2018; 14:724-730. [PMID: 30443516 PMCID: PMC6222149 DOI: 10.12965/jer.1836442.221] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 10/08/2018] [Indexed: 11/23/2022] Open
Abstract
Stress is the physiological responses of organisms to harmful or threatening stimuli that allow appropriate behavioral responses to the stressor. In the present study, the effect of treadmill exercise on stress-induced anxiety was evaluated using rats. To induce stress, the rats were exposed to an inescapable electric foot shock. Exposure of rats to the electric foot shock was performed for 7 days. The rats in the exercise groups were made to run on a motorized treadmill for 30 min once a day for 4 weeks stating one day after last electric food shock. Anxiety-like behaviors were determined by open field test and elevated plus-maze test. The expressions of c-Fos and neuronal nitric oxide synthase (nNOS) in the hypothalamus and locus coeruleus were detected by immunohistochemistry. In the present results, locomotor activity in the center of the open field test and the number of entries and time in the open arms of the elevated plus-maze test were reduced in the rats with stress-induced anxiety. Treadmill running enhanced these locomotor activities, the number of entries and time in the stress-induced anxiety rats. c-Fos and nNOS expressions in the hypothalamus and locus coeruleus were increased in the stress-induced rats. Treadmill exercise reduced c-Fos and nNOS overexpressions in the stress-induced rats. In the present study, treadmill exercise ameliorated anxiety-like behaviors in the stress-induced rats. The improving effect of treadmill exercise on anxiety-like behaviors might be ascribed to the suppressing effect of exercise on c-Fos and nNOS expressions.
Collapse
Affiliation(s)
- Jin-Hee Seo
- Department of Adaptive Physical Education, Baekseok University, Cheonan, Korea
| |
Collapse
|
18
|
Wang YC, Yu YH, Tsai ML, Huang ACW. Motor function in an animal model with ouabain-induced bipolar disorder and comorbid anxiety behavior. Psychiatry Res 2018; 268:508-513. [PMID: 30165326 DOI: 10.1016/j.psychres.2018.07.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 04/10/2018] [Accepted: 07/18/2018] [Indexed: 12/26/2022]
Abstract
In a clinical setting, anxiety disorder is highly correlated with bipolar I disorder in humans. However, the comorbidity of anxiety behavior and bipolar disorder still remains unclear in an animal model. This study utilized an ouabain-induced animal mode to examine anxiety and mania in an open field test. In the present study, 5 µl of artificial cerebrospinal fluid (aCSF) or ouabain (10-5, 10-4, and 10-3 M) were administered into the left ventricle. The animals' motor functions and anxiety behaviors were measured for 15 min. The results showed that 10-3 M ouabain significantly increased the animal's total distance traveled, average speed, and maximum speed compared to the control group. The time spent inside (i.e., how much time rats spent in the center of the square) and the inside-outside times of the central square (i.e., how many times rats ran across the center square) of the higher-concentration groups (10-4 M and 10-3 M) were significantly decreased. Therefore, a high concentration of ouabain may induce hyperactivity. The 10-4 M and 10-3 M ouabain groups exhibited more anxiety behaviors. The study is the first model to examine comorbid anxiety behaviors and bipolar disorder in an animal model. The study provides some insights for comorbid anxiety and bipolar disorder in clinics.
Collapse
Affiliation(s)
- Ying-Chou Wang
- Department of Clinical Psychology, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Ying Hao Yu
- Department of Psychology, Fo Guang University, Yilan County 26247, Taiwan
| | - Meng-Li Tsai
- Department of Biomechatronic Engineering, National Ilan University, Ilan, Taiwan
| | | |
Collapse
|
19
|
Rouine J, Callaghan CK, O'Mara SM. Opioid modulation of depression: A focus on imaging studies. PROGRESS IN BRAIN RESEARCH 2018; 239:229-252. [PMID: 30314568 DOI: 10.1016/bs.pbr.2018.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Depression is the leading cause of disability worldwide, with over 300 million people affected. Almost all currently available antidepressant treatments target monoamine neurotransmitter systems and have a delayed onset of action up to several weeks that can be associated with low rates of treatment response. The endogenous opioid system has been identified as a potential target for the development of novel antidepressants due to its high opioid receptor concentrations in central limbic areas that are also implicated in physiological processes including regulation of mood and emotion. Genetic depletion, pharmacological manipulation, and preclinical models have been widely used to characterize the role of opioid transmission in depressive states. Neuroimaging studies have been carried out in clinical populations to investigate opioid transmission in mood and emotion in an attempt to identify those regional anatomical and functional brain changes that are associated with depression. Great insight has been provided into the cerebral structural and functional changes associated with depression but there remains a need to tie the functional theories of depression to anatomical localization and further neuroimaging studies are best placed to do this.
Collapse
Affiliation(s)
- Jennifer Rouine
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland.
| | - Charlotte K Callaghan
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| | - Shane M O'Mara
- Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
20
|
Okhovatian F, Rezaei Tavirani M, Rostami-Nejad M, Rezaei Tavirani S. Protein-Protein Interaction Network Analysis Revealed a New Prospective of Posttraumatic Stress Disorder. Galen Med J 2018; 7:e1137. [PMID: 34466439 PMCID: PMC8344167 DOI: 10.22086/gmj.v0i0.1137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/25/2018] [Accepted: 05/08/2018] [Indexed: 11/30/2022] Open
Abstract
Background Posttraumatic stress disorder (PTSD) is known by a number of mental disorders, including recurring memories of trauma, mental appalling, and escaping of sign that make them recall the trauma in question. Clinical interviews serve as the main diagnostic tool for PTSD. With respect to treatment, either pharmacotherapy or psychotherapy or a combination of both is used as a therapeutic method for PTSD. In this study, a number of crucial genes related to PTSD, which can be considered as biomarker candidates, were represented. Materials and Methods The genes related to PTSD were extracted from the STRING database and organized in a protein-protein interaction network with the help of Cytoscape software version 3.6.0. The network was analyzed, and the important genes were introduced based on central indices. The biological processes related to the crucial genes were enriched via gene ontology using ClueGO. Results From a total of 100 genes, 63 genes were extracted that formed the main connected component, and of these, 12 crucial genes-POMC, BDNF, FOS, NR3C1, CRH, IL6, NPS, HTR1A, NPY, CREB1, CRHR1, and TAC1-were introduced. Biological processes were classified into the regulation of corticosterone, regulation of behavior, response to fungus, multicellular organism response to stress, and associative learning. Conclusion The introduced 12 crucial genes can be used as a biomarker panel related to PTSD and can be considered as a diagnostic reagent or drug target; however, more investigations are needed to use these genes as biomarkers.
Collapse
Affiliation(s)
- Farshad Okhovatian
- Physiotherapy Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sina Rezaei Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Li W, Papilloud A, Lozano-Montes L, Zhao N, Ye X, Zhang X, Sandi C, Rainer G. Stress Impacts the Regulation Neuropeptides in the Rat Hippocampus and Prefrontal Cortex. Proteomics 2018; 18:e1700408. [PMID: 29406625 DOI: 10.1002/pmic.201700408] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/10/2018] [Indexed: 11/05/2022]
Abstract
Adverse life experiences increase the lifetime risk to several stress-related psychopathologies, such as anxiety or depressive-like symptoms following stress in adulthood. However, the neurochemical modulations triggered by stress have not been fully characterized. Neuropeptides play an important role as signaling molecules that contribute to physiological regulation and have been linked to neurological and psychiatric diseases. However, little is known about the influence of stress on neuropeptide regulation in the brain. Here, we have performed an exploratory study of how neuropeptide expression at adulthood is modulated by experiencing a period of multiple stressful experiences. We have targeted hippocampus and prefrontal cortex (PFC) brain areas, which have previously been shown to be modulated by stressors, employing a targeted liquid chromatography-mass spectrometry (LC-MS) based approach that permits broad peptide coverage with high sensitivity. We found that in the hippocampus, Met-enkephalin, Met-enkephalin-Arg-Phe, and Met-enkephalin-Arg-Gly-Leu were upregulated, while Leu-enkephalin and Little SAAS were downregulated after stress. In the PFC area, Met-enkephalin-Arg-Phe, Met-enkephalin-Arg-Gly-Leu, peptide PHI-27, somatostatin-28 (AA1-12), and Little SAAS were all downregulated. This systematic evaluation of neuropeptide alterations in the hippocampus and PFC suggests that stressors impact neuropeptides and that neuropeptide regulation is brain-area specific. These findings suggest several potential peptide candidates, which warrant further investigations in terms of correlation with depression-associated behaviors.
Collapse
Affiliation(s)
- Wenxue Li
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Aurelie Papilloud
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Science, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | | | - Nan Zhao
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Xueting Ye
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Xiaozhe Zhang
- Division of Biological Technology, Chinese Academy of Science, Dalian Institute of Chemical Physics, Dalian, P. R. China
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, School of Life Science, Ecole Polytechnique Federale de Lausanne, Lausanne, Switzerland
| | - Gregor Rainer
- Department of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
22
|
Song H, Zheng Y, Cai F, Ma Y, Yang J, Wu Y. c-Fos downregulation positively regulates EphA5 expression in a congenital hypothyroidism rat model. J Mol Histol 2018; 49:147-155. [PMID: 29330744 DOI: 10.1007/s10735-018-9754-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 01/05/2018] [Indexed: 12/16/2022]
Abstract
The EphA5 receptor is well established as an axon guidance molecule during neural system development and plays an important role in dendritic spine formation and synaptogenesis. Our previous study has showed that EphA5 is decreased in the developing brain of congenital hypothyroidism (CH) and the EphA5 promoter methylation modification participates in its decrease. c-Fos, a well-kown transcription factor, has been considered in association with brain development. Bioinformatics analysis showed that the EphA5 promoter region contained five putative c-fos binding sites. The chromatin immunoprecipitation (ChIP) assays were used to assess the direct binding of c-fos to the EphA5 promoter. Furthermore, dual-luciferase assays showed that these three c-fos protein binding sites were positive regulatory elements for EphA5 expression in PC12 cells. Moreover, We verified c-fos positively regulation for EphA5 expression in CH model. Q-PCR and Western blot showed that c-fos overexpression could upregulate EphA5 expression in hippocampal neurons of rats with CH. Our results suggest that c-fos positively regulates EphA5 expression in CH rat model.
Collapse
Affiliation(s)
- Honghua Song
- Department of Pediatrics, Affiliated Hospital of Nantong University, 20 Xi Si Road, Nantong, 226001, Jiangsu Province, China
| | - Yuqin Zheng
- Department of Pediatrics, Affiliated Hospital of Nantong University, 20 Xi Si Road, Nantong, 226001, Jiangsu Province, China
| | - Fuying Cai
- Department of Pediatrics, Yin Shan Lake Hospital of Wuzhong District, Suzhou, 215100, Jiangsu Province, China
| | - Yanyan Ma
- Department of Pediatrics, Affiliated Hospital of Nantong University, 20 Xi Si Road, Nantong, 226001, Jiangsu Province, China
| | - Jingyue Yang
- Department of Pediatrics, Affiliated Hospital of Nantong University, 20 Xi Si Road, Nantong, 226001, Jiangsu Province, China
| | - Youjia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, 20 Xi Si Road, Nantong, 226001, Jiangsu Province, China.
| |
Collapse
|
23
|
Maldonado R, Baños JE, Cabañero D. Usefulness of knockout mice to clarify the role of the opioid system in chronic pain. Br J Pharmacol 2018; 175:2791-2808. [PMID: 29124744 DOI: 10.1111/bph.14088] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/13/2017] [Accepted: 10/17/2017] [Indexed: 12/29/2022] Open
Abstract
Several lines of knockout mice deficient in the genes encoding each component of the endogenous opioid system have been used for decades to clarify the specific role of the different opioid receptors and peptide precursors in many physiopathological conditions. The use of these genetically modified mice has improved our knowledge of the specific involvement of each endogenous opioid component in nociceptive transmission during acute and chronic pain conditions. The present review summarizes the recent advances obtained using these genetic tools in understanding the role of the opioid system in the pathophysiological mechanisms underlying chronic pain. Behavioural data obtained in these chronic pain models are discussed considering the peculiarities of the behavioural phenotype of each line of knockout mice. These studies have identified the crucial role of specific components of the opioid system in different manifestations of chronic pain and have also opened new possible therapeutic approaches, such as the development of opioid compounds simultaneously targeting several opioid receptors. However, several questions still remain open and require further experimental effort to be clarified. The novel genetic tools now available to manipulate specific neuronal populations and precise genome editing in mice will facilitate in a near future the elucidation of the role of each component of the endogenous opioid system in chronic pain. LINKED ARTICLES This article is part of a themed section on Emerging Areas of Opioid Pharmacology. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.14/issuetoc.
Collapse
Affiliation(s)
- Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Josep Eladi Baños
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain.,IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - David Cabañero
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
24
|
Enkephalins: Endogenous Analgesics with an Emerging Role in Stress Resilience. Neural Plast 2017; 2017:1546125. [PMID: 28781901 PMCID: PMC5525068 DOI: 10.1155/2017/1546125] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/15/2017] [Accepted: 05/24/2017] [Indexed: 12/20/2022] Open
Abstract
Psychological stress is a state of mental or emotional strain or tension that results from adverse or demanding circumstances. Chronic stress is well known to induce anxiety disorders and major depression; it is also considered a risk factor for Alzheimer's disease. Stress resilience is a positive outcome that is associated with preserved cognition and healthy aging. Resilience presents psychological and biological characteristics intrinsic to an individual conferring protection against the development of psychopathologies in the face of adversity. How can we promote or improve resilience to chronic stress? Numerous studies have proposed mechanisms that could trigger this desirable process. The roles of enkephalin transmission in the control of pain, physiological functions, like respiration, and affective disorders have been studied for more than 30 years. However, their role in the resilience to chronic stress has received much less attention. This review presents the evidence for an emerging involvement of enkephalin signaling through its two associated opioid receptors, μ opioid peptide receptor and δ opioid peptide receptor, in the natural adaptation to stressful lifestyles.
Collapse
|
25
|
Interaction of Depression and Anxiety in the Development of Mixed Anxiety/Depression Disorder. Experimental Studies of the Mechanisms of Comorbidity (review). ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s11055-017-0458-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Vicario A, Mendoza E, Abellán A, Scharff C, Medina L. Genoarchitecture of the extended amygdala in zebra finch, and expression of FoxP2 in cell corridors of different genetic profile. Brain Struct Funct 2016; 222:481-514. [PMID: 27160258 PMCID: PMC5225162 DOI: 10.1007/s00429-016-1229-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 04/21/2016] [Indexed: 02/01/2023]
Abstract
We used a battery of genes encoding transcription factors (Pax6, Islet1, Nkx2.1, Lhx6, Lhx5, Lhx9, FoxP2) and neuropeptides to study the extended amygdala in developing zebra finches. We identified different components of the central extended amygdala comparable to those found in mice and chickens, including the intercalated amygdalar cells, the central amygdala, and the lateral bed nucleus of the stria terminalis. Many cells likely originate in the dorsal striatal domain, ventral striatal domain, or the pallidal domain, as is the case in mice and chickens. Moreover, a cell subpopulation of the central extended amygdala appears to originate in the prethalamic eminence. As a general principle, these different cells with specific genetic profiles and embryonic origin form separate or partially intermingled cell corridors along the extended amygdala, which may be involved in different functional pathways. In addition, we identified the medial amygdala of the zebra finch. Like in the chickens and mice, it is located in the subpallium and is rich in cells of pallido-preoptic origin, containing minor subpopulations of immigrant cells from the ventral pallium, alar hypothalamus and prethalamic eminence. We also proposed that the medial bed nucleus of the stria terminalis is composed of several parallel cell corridors with different genetic profile and embryonic origin: preoptic, pallidal, hypothalamic, and prethalamic. Several of these cell corridors with distinct origin express FoxP2, a transcription factor implicated in synaptic plasticity. Our results pave the way for studies using zebra finches to understand the neural basis of social behavior, in which the extended amygdala is involved.
Collapse
Affiliation(s)
- Alba Vicario
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain
| | | | - Antonio Abellán
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain
| | | | - Loreta Medina
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain.
| |
Collapse
|
27
|
Roques BP. Contribution of Delta-Opioid Receptors to Pathophysiological Events Explored by Endogenous Enkephalins. Handb Exp Pharmacol 2016; 247:53-70. [PMID: 27417433 DOI: 10.1007/164_2016_17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023]
Abstract
Very few discoveries in the neurosciences have triggered clinical speculation and experimentation regarding the etiology of psychiatric illness to the same extent as that following identification of the opiate receptor(s) and subsequent isolation of endogenous morphine-like peptides. There is overwhelming evidence in animals and in human that opioids are involved in behaviorally relevant issues such as the modulation of pain, the response to stress, motivation, addiction, sexuality, food intake, etc., but our knowledge on the possible relation between opioids and mental illness is still very limited.These responses could be explored eitheir by using higlhy selective delta agonist or by emphasizing the effects of phasically secreted endogenous opioid peptides, enkephalin. Both approaches were investigated in particular through protection of enkephalin degradation by dual enkephalinase ihibitors DENKIs such as RB101, PL37 or PL265.
Collapse
Affiliation(s)
- Bernard P Roques
- Membre de l'Académie des Sciences (France et Europe), U1022 INSERM/UMR 8258 CNRS, Université Paris-Descartes (Paris V), CSO Pharmaleads SAS, 4 Avenue de l'Observatoire, 75006, Paris, France.
| |
Collapse
|
28
|
Ahn JY, Tae HJ, Cho JH, Kim IH, Ahn JH, Park JH, Kim DW, Cho JH, Won MH, Hong S, Lee JC, Seo JY. Activation of immediate-early response gene c-Fos protein in the rat paralimbic cortices after myocardial infarction. Neural Regen Res 2015; 10:1251-7. [PMID: 26487852 PMCID: PMC4590237 DOI: 10.4103/1673-5374.162757] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
c-Fos is a good biological marker for detecting the pathogenesis of central nervous system disorders. Few studies are reported on the change in myocardial infarction-induced c-Fos expression in the paralimbic regions. Thus, in this study, we investigated the changes in c-Fos expression in the rat cingulate and piriform cortices after myocardial infarction. Neuronal degeneration in cingulate and piriform cortices after myocardial infarction was detected using cresyl violet staining, NeuN immunohistochemistry and Fluoro-Jade B histofluorescence staining. c-Fos-immunoreactive cells were observed in cingulate and piriform cortices at 3 days after myocardial infarction and peaked at 7 and 14 days after myocardial infarction. But they were hardly observed at 56 days after myocardial infarction. The chronological change of c-Fos expression determined by western blot analysis was basically the same as that of c-Fos immunoreactivity. These results indicate that myocardial infarction can cause the chronological change of immediate-early response gene c-Fos protein expression, which might be associated with the neural activity induced by myocardial infarction.
Collapse
Affiliation(s)
- Ji Yun Ahn
- Department of Emergency Medicine, Sacred Heart Hospital, College of Medicine, Hallym University, Anyang, South Korea ; Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Hyun-Jin Tae
- Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon, South Korea
| | - Jeong-Hwi Cho
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Ji Hyeon Ahn
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Dong Won Kim
- Department of Emergency Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Seongkweon Hong
- Department of Surgery, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Jeong Yeol Seo
- Department of Emergency Medicine, Chuncheon Sacred Heart Hospital, College of Medicine, Hallym University, Chuncheon, South Korea
| |
Collapse
|
29
|
Viggiano A, Cacciola G, Widmer DAJ, Viggiano D. Anxiety as a neurodevelopmental disorder in a neuronal subpopulation: Evidence from gene expression data. Psychiatry Res 2015; 228:729-40. [PMID: 26089015 DOI: 10.1016/j.psychres.2015.05.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 05/14/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022]
Abstract
The relationship between genes and anxious behavior, is nor linear nor monotonic. To address this problem, we analyzed with a meta-analytic method the literature data of the behavior of knockout mice, retrieving 33 genes whose deletion was accompanied by increased anxious behavior, 34 genes related to decreased anxious behavior and 48 genes not involved in anxiety. We correlated the anxious behavior resulting from the deletion of these genes to their brain expression, using the Allen Brain Atlas and Gene Expression Omnibus (GEO) database. The main finding is that the genes accompanied, after deletion, by a modification of the anxious behavior, have lower expression in the cerebral cortex, the amygdala and the ventral striatum. The lower expression level was putatively due to their selective presence in a neuronal subpopulation. This difference was replicated also using a database of human gene expression, further showing that the differential expression pertained, in humans, a temporal window of young postnatal age (4 months up to 4 years) but was not evident at fetal or adult human stages. Finally, using gene enrichment analysis we also show that presynaptic genes are involved in the emergence of anxiety and postsynaptic genes in the reduction of anxiety after gene deletion.
Collapse
Affiliation(s)
- Adela Viggiano
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy
| | - Giovanna Cacciola
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy
| | | | - Davide Viggiano
- Department of Health Sciences, University of Molise, Campobasso 86100, Italy; Department of Cardio-Thoracic and Respiratory Science, Second University of Naples, Naples, Italy.
| |
Collapse
|
30
|
Victoria NC, Murphy AZ. The long-term impact of early life pain on adult responses to anxiety and stress: Historical perspectives and empirical evidence. Exp Neurol 2015. [PMID: 26210872 DOI: 10.1016/j.expneurol.2015.07.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Approximately 1 in 6 infants are born prematurely each year. Typically, these infants spend 25 days in the Neonatal Intensive Care Unit (NICU) where they experience 10-18 painful and inflammatory procedures each day. Remarkably, pre-emptive analgesics and/or anesthesia are administered less than 25% of the time. Unalleviated pain during the perinatal period is associated with permanent decreases in pain sensitivity, blunted cortisol responses and high rates of neuropsychiatric disorders. To date, the mechanism(s) by which these long-term changes in stress and pain behavior occur, and whether such alterations can be prevented by appropriate analgesia at the time of insult, remains unclear. Work in our lab using a rodent model of early life pain suggests that inflammatory pain experienced on the day of birth blunts adult responses to stress- and pain-provoking stimuli, and dysregulates the hypothalamic pituitary adrenal (HPA) axis in part through a permanent upregulation in central endogenous opioid tone. This review focuses on the long-term impact of neonatal inflammatory pain on adult anxiety- and stress-related responses, and underlying neuroanatomical changes in the context of endogenous pain control and the HPA axis. These two systems are in a state of exaggerated developmental plasticity early in postnatal life, and work in concert to respond to noxious or aversive stimuli. We present empirical evidence from animal and clinical studies, and discuss historical perspectives underlying the lack of analgesia/anesthetic use for early life pain in the modern NICU.
Collapse
Affiliation(s)
- Nicole C Victoria
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303, USA.
| | - Anne Z Murphy
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave, Atlanta, GA 30303, USA.
| |
Collapse
|
31
|
Zai CC, Gonçalves VF, Tiwari AK, Gagliano SA, Hosang G, de Luca V, Shaikh SA, King N, Chen Q, Xu W, Strauss J, Breen G, Lewis CM, Farmer AE, McGuffin P, Knight J, Vincent JB, Kennedy JL. A genome-wide association study of suicide severity scores in bipolar disorder. J Psychiatr Res 2015; 65:23-9. [PMID: 25917933 DOI: 10.1016/j.jpsychires.2014.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/16/2014] [Accepted: 11/13/2014] [Indexed: 10/24/2022]
Abstract
BACKGROUND Suicide claims one million lives worldwide annually, making it a serious public health concern. The risk for suicidal behaviour can be partly explained by genetic factors, as suggested by twin and family studies (reviewed in (Zai et al. 2012)). Recently, genome-wide association studies (GWASs) of suicide attempt on large samples of bipolar disorder (BD) patients from multiple sites have identified a number of novel candidate genes. GWASs of suicide behaviour severity, from suicidal ideation to serious suicide attempt, have not been reported for BD. METHODS We conducted a GWAS of suicide behaviour severity in three independent BD samples:212 small nuclear families with BD probands from Toronto, Canada, 428 BD cases from Toronto, and 483 BD cases from the UK. We carried out imputation with 1000 Genome Project data as reference using IMPUTE2. Quality control and data analysis was conducted using PLINK and R. We conducted the quantitative analyses of suicide behaviour severity in the three samples separately, and derived an overall significance by a meta-analysis using the METAL software. RESULTS We did not find genome-wide significant association of any tested markers in any of the BD samples, but we found a number of suggestive associations, including regions on chromosomes 8 and 10 (p < 1e-5). CONCLUSIONS Our GWAS findings suggest that likely many gene variants of small effects contribute collectively to the risk for suicidal behaviour severity in BD. Larger independent replications are required to strengthen the findings from the GWAS presented here.
Collapse
Affiliation(s)
- Clement C Zai
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Vanessa F Gonçalves
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Arun K Tiwari
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Sarah A Gagliano
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Georgina Hosang
- Department of Psychology, Goldsmiths, University of London, New Cross, London, United Kingdom; MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Vincenzo de Luca
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada
| | - Sajid A Shaikh
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Nicole King
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Qian Chen
- Cancer Care Ontario, Toronto, Ontario, Canada
| | - Wei Xu
- Dalla Lana School of Public Health, University of Toronto, Ontario, Canada; Ontario Cancer Institute, Prince Margaret Hospital, Toronto, Ontario, Canada
| | - John Strauss
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Gerome Breen
- MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Cathryn M Lewis
- MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Anne E Farmer
- MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Peter McGuffin
- MRC Social, Genetic & Developmental Psychiatry Centre, King's College London, London, United Kingdom
| | - Jo Knight
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada
| | - John B Vincent
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada; Molecular Neuropsychiatry and Development Laboratory (MiND), Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - James L Kennedy
- Neurogenetics Section, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada; Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Ontario, Canada.
| |
Collapse
|
32
|
Chauhan E, Bali A, Singh N, Jaggi AS. Pharmacological investigations on cross adaptation in mice subjected to stress immobilization. Life Sci 2015; 127:98-105. [DOI: 10.1016/j.lfs.2015.02.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 01/21/2015] [Accepted: 02/17/2015] [Indexed: 11/15/2022]
|
33
|
Stress and opioids: role of opioids in modulating stress-related behavior and effect of stress on morphine conditioned place preference. Neurosci Biobehav Rev 2015; 51:138-50. [PMID: 25636946 DOI: 10.1016/j.neubiorev.2014.12.018] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 12/24/2014] [Accepted: 12/31/2014] [Indexed: 11/21/2022]
Abstract
Research studies have defined the important role of endogenous opioids in modulating stress-associated behavior. The release of β-endorphins in the amygdala in response to stress helps to cope with a stressor by inhibiting the over-activation of HPA axis. Administration of mu opioid agonists reduces the risk of developing post-traumatic stress disorder (PTSD) following a traumatic event by inhibiting fear-related memory consolidation. Similarly, the release of endogenous enkephalin and nociceptin in the basolateral amygdala and the nucleus accumbens tends to produce the anti-stress effects. An increase in dynorphin levels during prolonged exposure to stress may produce learned helplessness, dysphoria and depression. Stress also influences morphine-induced conditioned place preference (CPP) depending upon the intensity and duration of the stressor. Acute stress inhibits morphine CPP, while chronic stress potentiates CPP. The development of dysphoria due to increased dynorphin levels may contribute to chronic stress-induced potentiation of morphine CPP. The activation of ERK/cyclic AMP responsive element-binding (CREB) signaling in the mesocorticolimbic area, glucocorticoid receptors in the basolateral amygdala, and norepinephrine and galanin system in the nucleus accumbens may decrease the acute stress-induced inhibition of morphine CPP. The increase in dopamine levels in the nucleus accumbens and augmentation of GABAergic transmission in the median prefrontal cortex may contribute in potentiating morphine CPP. Stress exposure reinstates the extinct morphine CPP by activating the orexin receptors in the nucleus accumbens, decreasing the oxytocin levels in the lateral septum and amygdala, and altering the GABAergic transmission (activation of GABAA and inactivation of GABAB receptors). The present review describes these varied interactions between opioids and stress along with the possible mechanism.
Collapse
|
34
|
Melo I, Drews E, Zimmer A, Bilkei-Gorzo A. Enkephalin knockout male mice are resistant to chronic mild stress. GENES BRAIN AND BEHAVIOR 2014; 13:550-8. [PMID: 24804898 DOI: 10.1111/gbb.12139] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/08/2014] [Accepted: 04/24/2014] [Indexed: 12/12/2022]
Abstract
Enhanced stress reactivity or sensitivity to chronic stress increases the susceptibility to mood pathologies such as major depression. The opioid peptide enkephalin is an important modulator of the stress response. Previous studies using preproenkephalin knockout (PENK KO) mice showed that these animals exhibit abnormal stress reactivity and show increased anxiety behavior in acute stress situations. However, the consequence of enkephalin deficiency in the reactivity to chronic stress conditions is not known. In this study, we therefore submitted wild-type (WT) and PENK KO male mice to chronic stress conditions, using the chronic mild stress (CMS) protocol. Subsequently, we studied the CMS effects on the behavioral and hormonal level and also performed gene expression analyses. In WT animals, CMS increased the expression of the enkephalin gene in the paraventricular nucleus (PVN) of the hypothalamus and elevated the corticosterone levels. In addition, WT mice exhibited enhanced anxiety in the zero-maze test and depression-related behaviors in the sucrose preference and forced swim tests. Surprisingly, in PENK KO mice, we did not detect anxiety and depression-related behavioral changes after the CMS procedure, and even measured a decreased hormonal stress response. These results indicate that PENK KO mice are resistant to the CMS effects, suggesting that enkephalin enhances the reactivity to chronic stress.
Collapse
Affiliation(s)
- I Melo
- Institute of Molecular Psychiatry, University of Bonn, Bonn, Germany
| | | | | | | |
Collapse
|
35
|
Hurd YL, Michaelides M, Miller ML, Jutras-Aswad D. Trajectory of adolescent cannabis use on addiction vulnerability. Neuropharmacology 2013; 76 Pt B:416-24. [PMID: 23954491 DOI: 10.1016/j.neuropharm.2013.07.028] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 07/16/2013] [Accepted: 07/19/2013] [Indexed: 12/22/2022]
Abstract
The adolescent brain is a period of dynamic development making it vulnerable to environmental factors such as drug exposure. Of the illicit drugs, cannabis is most used by teenagers since it is perceived by many to be of little harm. This perception has led to a growing number of states approving its legalization and increased accessibility. Most of the debates and ensuing policies regarding cannabis were done without consideration of its impact on one of the most vulnerable population, namely teens, or without consideration of scientific data. We provide an overview of the endocannabinoid system in relation to adolescent cannabis exposure and provide insights regarding factors such as genetics and behavioral traits that confer risk for subsequent addiction. While it is clear that more systematic scientific studies are needed to understand the long-term impact of adolescent cannabis exposure on brain and behavior, the current evidence suggests that it has a far-reaching influence on adult addictive behaviors particularly for certain subsets of vulnerable individuals. This article is part of a Special Issue entitled 'NIDA 40th Anniversary Issue'.
Collapse
Affiliation(s)
- Yasmin L Hurd
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; James J. Peters Veterans Administration, Bronx, NY, USA.
| | | | | | | |
Collapse
|
36
|
Temporal association of elevated cholecystokininergic tone and adolescent trauma is critical for posttraumatic stress disorder-like behavior in adult mice. Proc Natl Acad Sci U S A 2013; 110:6589-94. [PMID: 23576730 DOI: 10.1073/pnas.1219601110] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Adolescent trauma (AT) is a common risk factor for adult-onset posttraumatic stress disorder (PTSD). However, the vulnerability to AT among different individuals varies dramatically, indicating that other cofactors are important. Despite extensive studies, the identification of those cofactors has had little success. Here, we found that after subjected to traumatic stress at postnatal day 25 (P25), a stage that is comparable to the human adolescent period, inducible/reversible forebrain-specific cholecystokinin receptor-2 transgenic (IF-CCKR-2 tg) mice exhibited a significantly higher level of PTSD-like behavior at a later life (adult) stage compared with their wild-type littermates. Moreover, in these traumatized IF-CCKR-2 tg mice, both the glucocorticoid negative feedback inhibition and spatial learning and memory were impaired. Interestingly, if the CCKR-2 transgene was specifically suppressed during the time of AT exposure, these observations were largely diminished, indicating that a temporal association of the elevated CCKergic tone and AT is pathogenically critical. Treatment of traumatized IF-CCKR-2 tg mice with fluoxetine, a selective serotonin reuptake inhibitor, for a period of 4 wk significantly attenuated the PTSD-like behavior and the impaired glucocorticoid negative feedback inhibition, but not the memory deficit, implying that the memory deficit is an independent post-AT clinical entity and not a consequence of PTSD. Taken together, these results reveal a dynamic role of the CCKergic system in the development of post-AT psychopathologies and suggest that a timely antagonism of CCKR-2 activity during AT exposure is a potential preventive strategy for post-AT psychopathologies including PTSD and cognitive dysfunction.
Collapse
|
37
|
Neuropsychological Functions of μ- and δ-Opioid Systems. ISRN ADDICTION 2013; 2013:674534. [PMID: 25938117 PMCID: PMC4392981 DOI: 10.1155/2013/674534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/08/2013] [Indexed: 12/19/2022]
Abstract
Brain opioid innervation is involved in many pathophysiological processes related to drug addiction. The main idea of the present review is that μ-/δ-opioid innervation is an intrinsic component of the motor/approach behavior network, which is activated synergetically with dopaminergic mesocorticolimbic network. Contribution of opioid innervation to the motor/approach behavior processing includes generation of positive emotions and inhibition of pain and stress reactions in order that the individual would be able to reach the vital goal. We cite the neuroanatomical data which showed that motor subcortical nuclei contain the most abundant opioid innervation and its activation is an obligatory component of positive emotions. In the majority of life situations, motor/approach behavior network concomitantly activates pain/stress control opioid network. Intensive cognitive activity induces activation of opioid innervation as well, and both enhancing and impairing effects of opioid agonists on cognitive functioning were demonstrated. Overall, the functioning of endogenous opioid networks may be summarized as following: NO physical/cognitive activity = NO positive emotions plus NO pain/stress control. We suppose that contemporary findings concerning neuropsychological functions of endogenous opioid system explain many controversial issues in neuropsychiatric conditions predisposing to drug addiction and neurological mechanisms of opioid addiction.
Collapse
|
38
|
Seo JH, Kim TW, Kim CJ, Sung YH, Lee SJ. Treadmill exercise during pregnancy ameliorates post‑traumatic stress disorder‑induced anxiety‑like responses in maternal rats. Mol Med Rep 2012; 7:389-95. [PMID: 23174863 DOI: 10.3892/mmr.2012.1197] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 11/20/2012] [Indexed: 11/05/2022] Open
Abstract
Post‑traumatic stress disorder (PTSD) is an anxiety disorder triggered by life‑threatening events that cause intense fear. Exercise is known to have protective effects on neuropsychiatric diseases. The present study investigated whether treadmill exercise during pregnancy reduced or alleviated symptoms of PTSD in maternal rats. To induce predator stress in pregnant rats, rats were exposed to a hunting dog in an enclosed room. Exposure time was three 10‑min daily sessions separated by 1 h, starting at week 1 of pregnancy until delivery. Pregnant rats in the exercise group were forced to run on a treadmill for 30 min once a day, starting one week following pregnancy until delivery. Rats receiving predator stress during pregnancy exhibited PTSD anxiety‑like behaviors following delivery. Expression of 5‑hydroxytryptamine (5‑HT) and its synthesizing enzyme tryptophan hydroxylase (TPH) in the dorsal raphe was increased compared with unstressed rats. Expression of c‑Fos and neuronal nitric oxide synthases (nNOS) in the hypothalamus and locus coeruleus were higher in the rats receiving stress during pregnancy compared with unstressed rats. By contrast, treadmill exercise during pregnancy ameliorated anxiety‑like behaviors and reduced the expression of 5‑HT, TPH, c‑Fos and nNOS in the PTSD maternal rats. The results of the present study indicate that exercise during pregnancy is suitable for use as a therapeutic strategy to reduce anxiety‑related disorders, including PTSD.
Collapse
Affiliation(s)
- Jin-Hee Seo
- Division of Sports Science, Baekseok University, Cheonan 330‑704, Republic of Korea
| | | | | | | | | |
Collapse
|
39
|
Yu H, Watt H, Kesavan C, Johnson PJ, Wergedal JE, Mohan S. Lasting consequences of traumatic events on behavioral and skeletal parameters in a mouse model for post-traumatic stress disorder (PTSD). PLoS One 2012; 7:e42684. [PMID: 22927935 PMCID: PMC3425500 DOI: 10.1371/journal.pone.0042684] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 07/11/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) is an anxiety disorder that not only affects mental health, but may also affect bone health. However, there have been no studies to examine the direct relationship between PTSD and bone. METHODOLOGY/PRINCIPAL FINDINGS We employed electric shocks in mice to simulate traumatic events that cause PTSD. We also injected the anxiogenic drug FG-7142 prior to electric shocks. Electric shocks created lasting conditioned fear memory in all mice. In young mice, electric shocks elicited not only behavioral response but also skeletal response, and injection of FG-7142 appeared to increase both types of response. For example in behavioral response within the first week, mice shocked alone froze an average of 6.2 sec in 10 sec tests, and mice injected with FG-7142 froze 7.6 sec, both significantly different (P<0.05) from control mice, which only froze 1.3 sec. In skeletal response at week 2, shocks alone reduced 6% bone mineral content (BMC) in total body (P = 0.06), while shocks with FG-7142 injection reduced not only 11% BMC (P<0.05) but also 6% bone mineral density (BMD) (P<0.05). In addition, FG-7142 injection also caused significant reductions of BMC in specific bones such as femur, lumbar vertebra, and tibia at week 3. Strong negative correlations (R(2) = -0.56, P<0.05) and regression (y = 0.2527-0.0037 * x, P<0.01) between freezing behavior and total body BMC in young mice indicated that increased contextual PTSD-like behavior was associated with reduced bone mass acquisition. CONCLUSIONS/SIGNIFICANCE This is the first study to document evidence that traumatic events induce lasting consequences on both behavior and skeletal growth, and electric shocks coupled with injection of anxiogenic FG-7142 in young mice can be used as a model to study the effect of PTSD-like symptoms on bone development.
Collapse
Affiliation(s)
- Hongrun Yu
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California, United States of America
- Department of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Heather Watt
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California, United States of America
| | - Chandrasekhar Kesavan
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California, United States of America
- Department of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Patrick J. Johnson
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California, United States of America
| | - Jon E. Wergedal
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California, United States of America
- Department of Medicine, Loma Linda University, Loma Linda, California, United States of America
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial VA Medical Center, Loma Linda, California, United States of America
- Department of Medicine, Loma Linda University, Loma Linda, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Bupesh M, Abellán A, Medina L. Genetic and experimental evidence supports the continuum of the central extended amygdala and a mutiple embryonic origin of its principal neurons. J Comp Neurol 2012; 519:3507-31. [PMID: 21800302 DOI: 10.1002/cne.22719] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The central extended amygdala is the major output center for telencephalic control of ingestion, fear responses, stress, and anxiety. In spite of the abundant data supporting the similarity in neurochemistry, connections, and function along the extended amygdala, embryological support for this continuum is lacking. By using a combination of in vitro migration assays, in situ hybridization, and immunostaining, here we show that its major components, including central amygdala and lateral bed nucleus of the stria terminalis (BST), are mosaics formed by different proportions of dorsal lateral ganglionic eminence (LGE)-, ventral LGE-, and medial ganglionic eminence (MGE)-derived principal neurons. The dorsal LGE produces Pax6-expressing neurons that primarily populate lateral parts of the central extended amygdala, including the capsular and part of lateral central amygdala, but also produces a few cells for the lateral BST. Based on correlation with preproenkephalin, many of these cells are likely enkephalinergic. The ventral LGE produces Islet1-expressing neurons that populate primarily the central and medial parts of the central amygdala but also produces numerous neurons for the lateral BST. Correlation with corticotropin-releasing factor suggests that these neurons express this neuropeptide. The MGE produces the majority of neurons of the lateral BST, but its ventrocaudal subdivision also produces an important subpopulation of projection neurons containing somatostatin for medial aspects of the central amygdala. Thus, distinct principal neurons originate in different embryonic domains, but the same domains contribute neurons to most subdivisions of the central extended amygdala, which may explain the similarity in neurochemistry and connections along the corridor.
Collapse
Affiliation(s)
- Munisamy Bupesh
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institut of Biomedical Research of Lleida (IRBLLEIDA), 25008 Lleida Spain
| | | | | |
Collapse
|
41
|
Rodgers KM, Bercum FM, McCallum DL, Rudy JW, Frey LC, Johnson KW, Watkins LR, Barth DS. Acute neuroimmune modulation attenuates the development of anxiety-like freezing behavior in an animal model of traumatic brain injury. J Neurotrauma 2012; 29:1886-97. [PMID: 22435644 PMCID: PMC3390983 DOI: 10.1089/neu.2011.2273] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Chronic anxiety is a common and debilitating result of traumatic brain injury (TBI) in humans. While little is known about the neural mechanisms of this disorder, inflammation resulting from activation of the brain's immune response to insult has been implicated in both human post-traumatic anxiety and in recently developed animal models. In this study, we used a lateral fluid percussion injury (LFPI) model of TBI in the rat and examined freezing behavior as a measure of post-traumatic anxiety. We found that LFPI produced anxiety-like freezing behavior accompanied by increased reactive gliosis (reflecting neuroimmune inflammatory responses) in key brain structures associated with anxiety: the amygdala, insula, and hippocampus. Acute peri-injury administration of ibudilast (MN166), a glial cell activation inhibitor, suppressed both reactive gliosis and freezing behavior, and continued neuroprotective effects were apparent several months post-injury. These results support the conclusion that inflammation produced by neuroimmune responses to TBI play a role in post-traumatic anxiety, and that acute suppression of injury-induced glial cell activation may have promise for the prevention of post-traumatic anxiety in humans.
Collapse
Affiliation(s)
- Krista M. Rodgers
- Department of Psychology and Neuroscience, University of Colorado–Boulder, Boulder, Colorado
| | - Florencia M. Bercum
- Department of Psychology and Neuroscience, University of Colorado–Boulder, Boulder, Colorado
| | - Danielle L. McCallum
- Department of Psychology and Neuroscience, University of Colorado–Boulder, Boulder, Colorado
| | - Jerry W. Rudy
- Department of Psychology and Neuroscience, University of Colorado–Boulder, Boulder, Colorado
| | - Lauren C. Frey
- Department of Neurology, University of Colorado–Denver, and Colorado Injury Control Research Center, Denver, Colorado
| | | | - Linda R. Watkins
- Department of Psychology and Neuroscience, University of Colorado–Boulder, Boulder, Colorado
| | - Daniel S. Barth
- Department of Psychology and Neuroscience, University of Colorado–Boulder, Boulder, Colorado
| |
Collapse
|
42
|
Jutras-Aswad D, Jacobs MM, Yiannoulos G, Roussos P, Bitsios P, Nomura Y, Liu X, Hurd YL. Cannabis-dependence risk relates to synergism between neuroticism and proenkephalin SNPs associated with amygdala gene expression: case-control study. PLoS One 2012; 7:e39243. [PMID: 22745721 PMCID: PMC3382183 DOI: 10.1371/journal.pone.0039243] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Accepted: 05/17/2012] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Many young people experiment with cannabis, yet only a subgroup progress to dependence suggesting individual differences that could relate to factors such as genetics and behavioral traits. Dopamine receptor D2 (DRD2) and proenkephalin (PENK) genes have been implicated in animal studies with cannabis exposure. Whether polymorphisms of these genes are associated with cannabis dependence and related behavioral traits is unknown. METHODOLOGY/PRINCIPAL FINDINGS Healthy young adults (18-27 years) with cannabis dependence and without a dependence diagnosis were studied (N = 50/group) in relation to a priori-determined single nucleotide polymorphisms (SNPs) of the DRD2 and PENK genes. Negative affect, Impulsive Risk Taking and Neuroticism-Anxiety temperamental traits, positive and negative reward-learning performance and stop-signal reaction times were examined. The findings replicated the known association between the rs6277 DRD2 SNP and decisions associated with negative reinforcement outcomes. Moreover, PENK variants (rs2576573 and rs2609997) significantly related to Neuroticism and cannabis dependence. Cigarette smoking is common in cannabis users, but it was not associated to PENK SNPs as also validated in another cohort (N = 247 smokers, N = 312 non-smokers). Neuroticism mediated (15.3%-19.5%) the genetic risk to cannabis dependence and interacted with risk SNPs, resulting in a 9-fold increase risk for cannabis dependence. Molecular characterization of the postmortem human brain in a different population revealed an association between PENK SNPs and PENK mRNA expression in the central amygdala nucleus emphasizing the functional relevance of the SNPs in a brain region strongly linked to negative affect. CONCLUSIONS/SIGNIFICANCE Overall, the findings suggest an important role for Neuroticism as an endophenotype linking PENK polymorphisms to cannabis-dependence vulnerability synergistically amplifying the apparent genetic risk.
Collapse
Affiliation(s)
- Didier Jutras-Aswad
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Michelle M. Jacobs
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Georgia Yiannoulos
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Panos Roussos
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Panos Bitsios
- Department of Psychiatry and Behavioral Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Yoko Nomura
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Psychology, Queens College, Queens, New York, United States of America
| | - Xun Liu
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
- Institute of Psychology, Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Yasmin L. Hurd
- Department of Psychiatry, Mount Sinai School of Medicine, New York, New York, United States of America
- Department of Psychiatry and Behavioral Sciences, Faculty of Medicine, University of Crete, Heraklion, Crete, Greece
- Department of Neuroscience, Mount Sinai School of Medicine, New York, New York, United States of America
- James J Peters VA Medical Center, New York, New York, United States of America
| |
Collapse
|
43
|
Inhibiting the breakdown of endogenous opioids and cannabinoids to alleviate pain. Nat Rev Drug Discov 2012; 11:292-310. [DOI: 10.1038/nrd3673] [Citation(s) in RCA: 139] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Taylor JJ, Borckardt JJ, George MS. Endogenous opioids mediate left dorsolateral prefrontal cortex rTMS-induced analgesia. Pain 2012; 153:1219-1225. [PMID: 22444187 DOI: 10.1016/j.pain.2012.02.030] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/11/2011] [Accepted: 02/24/2012] [Indexed: 01/20/2023]
Abstract
The concurrent rise of undertreated pain and opiate abuse poses a unique challenge to physicians and researchers alike. A focal, noninvasive form of brain stimulation called repetitive transcranial magnetic stimulation (rTMS) has been shown to produce acute and chronic analgesic effects when applied to dorsolateral prefrontal cortex (DLPFC), but the anatomical and pharmacological mechanisms by which prefrontal rTMS induces analgesia remain unclear. Data suggest that DLPFC mediates top-down analgesia via gain modulation of the supraspinal opioidergic circuit. This potential pathway might explain how prefrontal rTMS reduces pain. The purpose of this sham-controlled, double-blind, crossover study was to determine whether left DLPFC rTMS-induced analgesia was sensitive to μ-opioid blockade. Twenty-four healthy volunteers were randomized to receive real or sham TMS after either intravenous saline or naloxone pretreatment. Acute hot and cold pain via quantitative sensory testing and hot allodynia via block testing on capsaicin-treated skin were assessed at baseline and at 0, 20, and 40 minutes after TMS treatment. When compared to sham, real rTMS reduced hot pain and hot allodynia. Naloxone pretreatment significantly reduced the analgesic effects of real rTMS. These results demonstrate that left DLPFC rTMS-induced analgesia requires opioid activity and suggest that rTMS drives endogenous opioidergic pain relief in the human brain. Further studies with chronic dosing regimens of drugs that block or augment the actions of opiates are needed to determine whether TMS can augment opiates in chronic or postoperative pain management.
Collapse
Affiliation(s)
- Joseph J Taylor
- Brain Stimulation Laboratory, Department of Psychiatry, Medical University of South Carolina, Charleston, SC, USA Ralph H. Johnson VA Medical Center, Charleston, SC, USA
| | | | | |
Collapse
|
45
|
Wang H, Zuo D, He B, Qiao F, Zhao M, Wu Y. Conditioned fear stress combined with single-prolonged stress: a new PTSD mouse model. Neurosci Res 2012; 73:142-52. [PMID: 22426496 DOI: 10.1016/j.neures.2012.03.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 03/06/2012] [Accepted: 03/07/2012] [Indexed: 01/15/2023]
Abstract
There are still some defects in current single-prolonged stress (SPS) model and conditioned fear (CF) stress model of post-traumatic stress disorder (PTSD). The purpose of this study is to evaluate a novel mouse model of PTSD. Male KM mice suffered the double stresses-SPS and CF. After incubation time, the novel model exhibited the PTSD-like behaviors: sensitive fear and conditioned fear, low activities and defects in novel object recognition abilities. The apoptosis in the hippocampus was significantly increased, which was induced by the double stresses and further caused the synaptic structure damages in the hippocampus. The electron microscopy analysis further proved the synaptic losses and neuronal impairments in the hippocampus. Our results indicated this combined stresses mouse model was better than the SPS model and CF model. In addition, in order to further verify this model, paroxetine was administered after the double stresses. The results showed that paroxetine administration reduced PTSD-like behaviors, hippocampal apoptosis and structure damages. We conclude that this mouse model is novel and more predictably mimicked the clinical characteristics of PTSD, and this model can be further used for investigating the mechanisms of PTSD and screening effective therapeutics agents.
Collapse
Affiliation(s)
- Haifeng Wang
- Department of Pharmacology, Shenyang Pharmaceutical University, Mailbox 41, Wenhua Road 103, Shenyang 110016, China
| | | | | | | | | | | |
Collapse
|
46
|
Stewart A, Gaikwad S, Kyzar E, Green J, Roth A, Kalueff AV. Modeling anxiety using adult zebrafish: a conceptual review. Neuropharmacology 2012; 62:135-43. [PMID: 21843537 PMCID: PMC3195883 DOI: 10.1016/j.neuropharm.2011.07.037] [Citation(s) in RCA: 263] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Revised: 07/22/2011] [Accepted: 07/23/2011] [Indexed: 11/21/2022]
Abstract
Zebrafish (Danio rerio) are rapidly emerging as a useful animal model in neurobehavioral research. Mounting evidence shows the suitability of zebrafish to model various aspects of anxiety-related states. Here, we evaluate established and novel approaches to uncover the molecular substrates, genetic pathways and neural circuits of anxiety using adult zebrafish. Experimental approaches to modeling anxiety in zebrafish include novelty-based paradigms, pharmacological and genetic manipulations, as well as innovative video-tracking, 3D-reconstructions, bioinformatics-based searchable databases and omics-based tools. Complementing traditional rodent models of anxiety, we provide a conceptual framework for the wider application of zebrafish and other aquatic models in anxiety research. This article is part of a Special Issue entitled 'Anxiety and Depression'.
Collapse
Affiliation(s)
- Adam Stewart
- Department of Pharmacology and Neuroscience Program, Tulane Neurophenotyping Platform, Zebrafish Neuroscience Research Consortium, Tulane University Medical School, 1430 Tulane Ave., New Orleans, LA 70112, USA
| | - Siddharth Gaikwad
- Department of Pharmacology and Neuroscience Program, Tulane Neurophenotyping Platform, Zebrafish Neuroscience Research Consortium, Tulane University Medical School, 1430 Tulane Ave., New Orleans, LA 70112, USA
| | - Evan Kyzar
- Department of Pharmacology and Neuroscience Program, Tulane Neurophenotyping Platform, Zebrafish Neuroscience Research Consortium, Tulane University Medical School, 1430 Tulane Ave., New Orleans, LA 70112, USA
| | - Jeremy Green
- Department of Pharmacology and Neuroscience Program, Tulane Neurophenotyping Platform, Zebrafish Neuroscience Research Consortium, Tulane University Medical School, 1430 Tulane Ave., New Orleans, LA 70112, USA
| | - Andrew Roth
- Department of Pharmacology and Neuroscience Program, Tulane Neurophenotyping Platform, Zebrafish Neuroscience Research Consortium, Tulane University Medical School, 1430 Tulane Ave., New Orleans, LA 70112, USA
| | - Allan V. Kalueff
- Department of Pharmacology and Neuroscience Program, Tulane Neurophenotyping Platform, Zebrafish Neuroscience Research Consortium, Tulane University Medical School, 1430 Tulane Ave., New Orleans, LA 70112, USA
| |
Collapse
|
47
|
Baranova KA, Rybnikova EA, Samoilov MO. Involvement of the transcription factor c-Fos in the protective effect of hypoxic preconditioning in a model of post-traumatic stress disorder in rats. NEUROCHEM J+ 2011. [DOI: 10.1134/s1819712411040039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
48
|
Abstract
This paper is the thirty-third consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2010 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior (Section 2), and the roles of these opioid peptides and receptors in pain and analgesia (Section 3); stress and social status (Section 4); tolerance and dependence (Section 5); learning and memory (Section 6); eating and drinking (Section 7); alcohol and drugs of abuse (Section 8); sexual activity and hormones, pregnancy, development and endocrinology (Section 9); mental illness and mood (Section 10); seizures and neurologic disorders (Section 11); electrical-related activity and neurophysiology (Section 12); general activity and locomotion (Section 13); gastrointestinal, renal and hepatic functions (Section 14); cardiovascular responses (Section 15); respiration (Section 16); and immunological responses (Section 17).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, USA.
| |
Collapse
|
49
|
Sudakov SK, Nazarova GA, Kolyasnikova KN, Kolpakov AA, Bashkatova VG. Effect of Peripheral Administration of Peptide Ligands of δ-Opioid Receptors on Anxiety Level and Locomotor Activity of Rats. Bull Exp Biol Med 2011; 151:661-3. [DOI: 10.1007/s10517-011-1409-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
50
|
Costanzi M, Cannas S, Saraulli D, Rossi-Arnaud C, Cestari V. Extinction after retrieval: Effects on the associative and nonassociative components of remote contextual fear memory. Learn Mem 2011; 18:508-18. [DOI: 10.1101/lm.2175811] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|