1
|
Chen D, Wang J, Cao J, Zhu G. cAMP-PKA signaling pathway and anxiety: Where do we go next? Cell Signal 2024; 122:111311. [PMID: 39059755 DOI: 10.1016/j.cellsig.2024.111311] [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: 06/24/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Cyclic adenosine monophosphate (cAMP) is an intracellular second messenger that is derived from the conversion of adenosine triphosphate catalysed by adenylyl cyclase (AC). Protein kinase A (PKA), the main effector of cAMP, is a dimeric protein kinase consisting of two catalytic subunits and two regulatory subunits. When cAMP binds to the regulatory subunits of PKA, it leads to the dissociation and activation of PKA, which allows the catalytic subunit of PKA to phosphorylate target proteins, thereby regulating various physiological functions and metabolic processes in cellular function. Recent researches also implicate the involvement of cAMP-PKA signaling in the pathologenesis of anxiety disorder. However, there are still debates on the prevention and treatment of anxiety disorders from this signaling pathway. To review the function of cAMP-PKA signaling in anxiety disorder, we searched the publications with the keywords including "cAMP", "PKA" and "Anxiety" from Pubmed, Embase, Web of Science and CNKI databases. The results showed that the number of publications on cAMP-PKA pathway in anxiety disorder tended to increase. Bioinformatics results displayed a close association between the cAMP-PKA pathway and the occurrence of anxiety. Mechanistically, cAMP-PKA signaling could influence brain-derived neurotrophic factor and neuropeptide Y and participate in the regulation of anxiety. cAMP-PKA signaling could also oppose the dysfunctions of gamma-aminobutyric acid (GABA), intestinal flora, hypothalamic-pituitary-adrenal axis, neuroinflammation, and signaling proteins (MAPK and AMPK) in anxiety. In addition, chemical agents with the ability to activate cAMP-PKA signaling demonstrated therapy potential against anxiety disorders. This review emphasizes the central roles of cAMP-PKA signaling in anxiety and the targets of the cAMP-PKA pathway would be potential candidates for treatment of anxiety. Nevertheless, more laboratory investigations to improve the therapeutic effect and reduce the adverse effect, and continuous clinical research will warrant the drug development.
Collapse
Affiliation(s)
- Daokang Chen
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jingji Wang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei 230061, China.
| | - Jian Cao
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
2
|
Kokane SS, Butler BD, Antonio JH, Armant RJ, Hoch AC, Coelho CS, Brady BN, Chamseddine HH, Perrotti LI. Interactions between estradiol and ERK, but not mTOR, signaling is necessary for enhanced cocaine-induced conditioned place preference in female rats. Pharmacol Biochem Behav 2023; 232:173653. [PMID: 37804867 DOI: 10.1016/j.pbb.2023.173653] [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: 07/17/2023] [Revised: 09/12/2023] [Accepted: 10/03/2023] [Indexed: 10/09/2023]
Abstract
Women rapidly progress from recreational cocaine use to dependence, consume greater quantities of cocaine, experience more positive subjective effects of cocaine and have higher incidences of relapse during abstinence. These effects have been replicated in animal models of cocaine addiction and indicate an enhanced sensitivity and therefore, vulnerability of females to cocaine addiction. Furthermore, it has been demonstrated that estradiol (E2) is a key mediator of the aforementioned effects of cocaine in women and female animals. However, studies identifying the influence of E2 on cocaine-associated reward and its underlying neurobiological mechanisms are lacking. Here, we further explored the influence of E2 on cocaine conditioned place preference in female rats. We show that E2 mediates cocaine-conditioned reward by potentiating cocaine-context associations. In addition, the E2-mediated increases in cocaine-induced CPP are associated with increased activation of ERK1/2 and mTOR proteins in the nucleus accumbens, dorsal striatum, and ventral tegmental area. To assess the involvement of ERK1/2 and mTOR in E2-mediated enhanced cocaine-CPP, we inhibited ERK1/2 and/or mTOR activity during cocaine-conditioning and before CPP-test. Inhibition of ERK1/2 during conditioning blocked cocaine-CPP in females, inhibition mTOR was without effect, and inhibiting ERK1/2 and mTOR before CPP-test blocked cocaine-CPP. In conclusion, we have established that E2 enhances cocaine-conditioned reward by potentiating cocaine-context associations formed during conditioning. Additionally, activation of ERK1/2 during cocaine-conditioning is necessary for the potentiation of cocaine-conditioned reward by E2. SIGNIFICANCE STATEMENT: Studies characterizing the molecular substrates underlying the effects of E2 during the formation of cocaine-context associations are virtually unknown. In this study, we established the influence of E2 during the formation of cocaine-CPP and characterized the role of ERK1/2 and mTOR activity on this effect within significant nodes of the reward pathway. The elucidation of the role of E2 in cocaine-induced intracellular signaling fills a significant gap in our knowledge regarding the mechanisms by which E2 affects intracellular signaling pathways to indicate the motivational salience of a stimulus. These data are crucial to our understanding of how fluctuating hormone levels can render females increasing sensitive to the rewarding effects of cocaine.
Collapse
Affiliation(s)
- Saurabh S Kokane
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Brandon D Butler
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Josimar Hernandez Antonio
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Ross J Armant
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Adam C Hoch
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Clinton S Coelho
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Blake N Brady
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Houda H Chamseddine
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA
| | - Linda I Perrotti
- Department of Psychology, College of Science, The University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
3
|
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
|
4
|
Laviolette SR. Molecular and neuronal mechanisms underlying the effects of adolescent nicotine exposure on anxiety and mood disorders. Neuropharmacology 2020; 184:108411. [PMID: 33245960 DOI: 10.1016/j.neuropharm.2020.108411] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/16/2020] [Accepted: 11/21/2020] [Indexed: 12/28/2022]
Abstract
Tobacco addiction is highly co-morbid with a variety of mental health conditions, including schizophrenia, mood and anxiety disorders. Nicotine, the primary psychoactive compound in tobacco-related products is known to functionally modulate brain circuits that are disturbed in these disorders. Nicotine can potently regulate the transmission of various neurochemicals, including dopamine (DA), γ-amino-butyric acid (GABA) and glutamate, within various mesocorticolimbic structures, such as the ventral tegmental area (VTA), nucleus accumbens (NAc) and prefrontal cortex (PFC), all of which show pathologies in these disorders. Many neuropsychiatric diseases have etiological origins during neurodevelopment, typically occurring during vulnerable periods of adolescent or pre-natal brain development. During these neurodevelopmental periods, exposure to extrinsic drug insults can induce enduring and long-term pathophysiological sequelae that ultimately increase the risk of developing chronic mental health disorders in later life. These vulnerability factors are of growing concern given rising rates of adolescent nicotine exposure via traditional tobacco use and the increasing use of alternative nicotine delivery formats such as vaping and e-cigarettes. A large body of clinical and pre-clinical evidence points to an important role for adolescent exposure to nicotine and increased vulnerability to developing mood and anxiety disorders in later life. This review will examine current clinical and pre-clinical evidence that pinpoints specific mechanisms within the mesocorticolimbic circuitry and molecular biomarkers linked to the association between adolescent nicotine exposure and increased risk of developing mood and anxiety-related disorders. This article is part of the special issue on 'Vulnerabilities to Substance Abuse'.
Collapse
Affiliation(s)
- Steven R Laviolette
- Addiction Research Group, Dept. of Anatomy & Cell Biology, Dept. of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, N6A 3K7, ON, Canada.
| |
Collapse
|
5
|
Diazepam and SL-327 synergistically attenuate anxiety-like behaviours in mice - Possible hippocampal MAPKs specificity. Neuropharmacology 2020; 180:108302. [PMID: 32931814 DOI: 10.1016/j.neuropharm.2020.108302] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/20/2020] [Accepted: 09/08/2020] [Indexed: 01/10/2023]
Abstract
Intracellular signalling pathways have been extensively studied as therapeutic targets for the treatment of mental diseases. Our attention has been caught by two kinases potentially involved in anxiety, ERK1/2 and CaMKII. The study aimed to examine changes in the activation of ERK1/2 and CaMKII concerning anxiolytic-like behaviours in mice. To evaluate anxiety-related response in mice, we used the open field test and the elevated plus maze test. Behavioural studies were complemented with the immunoblotting analysis to identify proteins of interest in the cortex, hippocampus, and striatum. We analysed the phosphorylation status of ERK1/2 and CaMKII in mice treated with a well-known anxiolytic drug - diazepam. Next, the blockade of ERK1/2 pathway by SL-327, a selective MEK1/2 inhibitor, was checked for anxiolytic action. Finally, the co-administration of subeffective doses of diazepam and SL-327 was investigated for a potential synergistic anxiolytic effect. Anxiolytic effects of acute diazepam are accompanied by decreased p-ERK1/2 and upregulation of p-CaMKII. Subchronic treatment with SL-327 leads to the manifestation of anxiolytic-like behaviours and changes in the phosphorylation status of both kinases in a diazepam-like manner. Co-administration of subeffective doses of SL-327 and diazepam induces anxiolysis, which is CaMKII-independent and correlates to selectively decreased phosphoactive ERK1/2 in the hippocampus. The MEK-ERK pathway is significantly involved in anxiolytic action of diazepam and its prolonged inhibition produces anxiolytic-like phenotype in mice. ERK inhibition could be used to manage anxiety symptoms in a benzodiazepine-sparing regimen for treatment of anxiety.
Collapse
|
6
|
Della Valle R, Mohammadmirzaei N, Knox D. Single prolonged stress alters neural activation in the periacqueductal gray and midline thalamic nuclei during emotional learning and memory. Learn Mem 2019; 26:1-9. [PMID: 31527186 PMCID: PMC6749928 DOI: 10.1101/lm.050310.119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 07/20/2019] [Indexed: 12/25/2022]
Abstract
Clinical and preclinical studies that have examined the neurobiology of persistent fear memory in posttraumatic stress disorder (PTSD) have focused on the medial prefrontal cortex, hippocampus, and amygdala. Sensory systems, the periaqueductal gray (PAG), and midline thalamic nuclei have been implicated in fear and extinction memory, but whether neural activity in these substrates is sensitive to traumatic stress (at baseline or during emotional learning and memory) remains unexplored. To address this, we used the single prolonged stress (SPS) model of traumatic stress. SPS and control rats were either subjected to fear conditioning (CS-fear) or presented with CSs alone (CS-only) during fear conditioning. All rats were then subjected to extinction training and testing. A subset of rats were euthanized after each behavioral stage and c-Fos and c-Jun used to measure neural activation in all substrates. SPS lowered c-Jun levels in the dorsomedial and lateral PAG at baseline, but the elevated c-Jun expression in the PAG during emotional learning and memory. SPS also altered c-Fos expression during fear and extinction learning/memory in midline thalamic nuclei. These findings suggest changes in neural function in the PAG and midline thalamic nuclei could contribute to persistent fear memory induced by traumatic stress. Interestingly, SPS effects were also observed in animals that never learned fear or extinction (i.e., CS-only). This raises the possibility that traumatic stress could have broader effects on the psychological function that are dependent on the PAG and midline thalamic nuclei.
Collapse
Affiliation(s)
- Rebecca Della Valle
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware 19716, USA
| | - Negin Mohammadmirzaei
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware 19716, USA
| | - Dayan Knox
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware 19716, USA
| |
Collapse
|
7
|
Jia Y, Han Y, Wang X, Han F. Role of apoptosis in the Post-traumatic stress disorder model-single prolonged stressed rats. Psychoneuroendocrinology 2018; 95:97-105. [PMID: 29843020 DOI: 10.1016/j.psyneuen.2018.05.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 12/14/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a stress-related mental disorder which occurs following exposure to traumatic events. A number of brain neuroimaging studies have revealed that PTSD patients have reduced volume and abnormal functions in the hippocampus and the amygdala. However, the pathogenesis of abnormalities in certain brain regions, as induced by PTSD, remains unclear. Recent studies, using the single prolonged stress (SPS) model, an animal model of PTSD, have found that abnormal apoptosis in certain brain regions, including the hippocampus, the amygdala, and the medial prefrontal cortex (mPFC); these areas are closely associated with emotion and cognition. In this review, we summarize the mechanism of apoptosis in SPS rats, including the endoplasmic reticulum (ER) and the mitochondria pathways. For the ER pathway, three individual pathways: PERK, IRE1, and ATF6 showed different roles on apoptosis and neuroprotection. Three key factors are thought to be involved in the mitochondrial pathway and PTSD-induced apoptosis: corticosteroid receptors, apoptosis-related factors, and anti-apoptosis factors. We have investigated the role of these factors and have attempted to identify which factors of the pathways are more focused towards neuronal protection, and which are more direct towards apoptosis. We also discussed the role of autophagy and the specific differences between autophagy and apoptosis in SPS rats. Finally, we discussed emerging researches related to anti-apoptosis treatment, including PERK inhibitors, IRE1 inhibitors, and metformin; collectively, these were exciting, but limited, This review provides a summary of the current understanding of apoptosis in SPS rats and the potential anti-apoptosis treatment strategies for PTSD.
Collapse
Affiliation(s)
- Yunbo Jia
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Yunhe Han
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Xinyue Wang
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China
| | - Fang Han
- PTSD laboratory, Department of Histology and Embryology, Basic Medical University, China Medical University, Shenyang, 110122, China.
| |
Collapse
|
8
|
Jobson CLM, Renard J, Szkudlarek H, Rosen LG, Pereira B, Wright DJ, Rushlow W, Laviolette SR. Adolescent Nicotine Exposure Induces Dysregulation of Mesocorticolimbic Activity States and Depressive and Anxiety-like Prefrontal Cortical Molecular Phenotypes Persisting into Adulthood. Cereb Cortex 2018; 29:3140-3153. [DOI: 10.1093/cercor/bhy179] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/06/2018] [Indexed: 12/17/2022] Open
Abstract
Abstract
Considerable evidence demonstrates strong comorbidity between nicotine dependence and mood and anxiety disorders. Nevertheless, the neurobiological mechanisms linking adolescent nicotine exposure to mood and anxiety disorders are not known. Disturbances in the mesocorticolimbic dopamine (DA) system, comprising the prefrontal cortex (PFC), ventral tegmental area (VTA), and nucleus accumbens (NAc), are correlates of mood and anxiety-related symptoms and this circuitry is strongly influenced by acute or chronic nicotine exposure. Using a combination of behavioral pharmacology, in vivo neuronal electrophysiology and molecular analyses, we examined and compared the effects of chronic nicotine exposure in rats during adolescence versus adulthood to characterize the mechanisms by which adolescent nicotine may selectively confer increased risk of developing mood and anxiety-related symptoms in later life. We report that exposure to nicotine, selectively during adolescence, induces profound and long-lasting neuronal, molecular and behavioral disturbances involving PFC DA D1R and downstream extracellular-signal-related kinase 1-2 (ERK 1-2) signaling. Remarkably, adolescent nicotine induced a persistent state of hyperactive DA activity in the ventral tegmental area (VTA) concomitant with hyperactive neuronal activity states in the PFC. Our findings identify several unique neuronal and molecular biomarkers that may serve as functional risk mechanisms for the long-lasting neuropsychiatric effects of adolescent smoking behaviors.
Collapse
Affiliation(s)
- Christina L M Jobson
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Justine Renard
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Hanna Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Laura G Rosen
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Brian Pereira
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Daniel J Wright
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Walter Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
9
|
Zhang N, Zhang L, Feng L, Yao L. Cananga odorata essential oil reverses the anxiety induced by 1-(3-chlorophenyl) piperazine through regulating the MAPK pathway and serotonin system in mice. JOURNAL OF ETHNOPHARMACOLOGY 2018; 219:23-30. [PMID: 29545208 DOI: 10.1016/j.jep.2018.03.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 11/16/2017] [Accepted: 03/09/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cananga odorata essential oil, known as ylang-ylang essential oil (YYO), was commonly used in the aromatherapy for relaxation and mood adjusting use. In our previous study, YYO played anxiolytic effects on the mice in several behavioral tests that based on the instinctive responses to novel environments. AIM OF THE STUDY To investigate the effects and mechanisms of YYO reversing the anxiety induced by 5-HT2C receptor agonist 1-(3-chlorophenyl) piperazine (m-CPP). MATERIALS AND METHODS m-CPP was administrated to the male ICR mice to develop an anxiety model. The anxiolytic effect of YYO (0.1%, 1% and 10%, v/v) was evaluated in the elevated plus maze (EPM) test after odor exposure. Western blot was used to detect the phosphorylation levels of extracellular signal-regulated kinase 1/2 (ERK1/2) and cAMP response element-binding protein (CREB) and the expression of c-Fos in the prefrontal cortex (PFC) and hippocampus after the EPM test. Serotonin and its metabolite change in the brain were detected by liquid chromatogram with an electrochemical detector. The effect of YYO on the plasma corticosterone level was evaluated using enzyme-linked immunosorbent assay (ELISA) after the odor exposure. RESULTS The behavior analysis showed that m-CPP (2 mg/kg and 4 mg/kg) could induce anxiety behaviors in the mice while diazepam (2 mg/kg) reversed the anxiety behavior induced by m-CPP. YYO dose-dependently increased the time and number of entries in the open arms (p < 0.05) compared to the Tween 80 group. YYO reduced the phosphorylation levels of ERK1/2 (p < 0.05) in both PFC and hippocampus. Down-regulations of phosphor-CREB (p < 0.05) and c-Fos (p < 0.05) were only observed in the hippocampus. YYO also affected the brain serotonin metabolism and reduced the blood plasma corticosterone level of the m-CPP treated mice. CONCLUSION YYO odor exposure could reverse the anxiety behaviors generated by m-CPP. The anxiolytic effect of YYO was associated with the ERK1/2/CREB pathway in the hippocampus and relevant to the serotonin system.
Collapse
Affiliation(s)
- Nan Zhang
- School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, China.
| | - Lei Zhang
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, China.
| | - Linyin Feng
- CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai, China.
| | - Lei Yao
- School of Agriculture and Biology, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai, China.
| |
Collapse
|
10
|
Jing Li J, Szkudlarek H, Renard J, Hudson R, Rushlow W, Laviolette SR. Fear Memory Recall Potentiates Opiate Reward Sensitivity through Dissociable Dopamine D1 versus D4 Receptor-Dependent Memory Mechanisms in the Prefrontal Cortex. J Neurosci 2018; 38:4543-4555. [PMID: 29686048 PMCID: PMC6705931 DOI: 10.1523/jneurosci.3113-17.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 03/09/2018] [Accepted: 04/04/2018] [Indexed: 12/18/2022] Open
Abstract
Disturbances in prefrontal cortical (PFC) dopamine (DA) transmission are well established features of psychiatric disorders involving pathological memory processing, such as post-traumatic stress disorder and opioid addiction. Transmission through PFC DA D4 receptors (D4Rs) has been shown to potentiate the emotional salience of normally nonsalient emotional memories, whereas transmission through PFC DA D1 receptors (D1Rs) has been demonstrated to selectively block recall of reward- or aversion-related associative memories. In the present study, using a combination of fear conditioning and opiate reward conditioning in male rats, we examined the role of PFC D4/D1R signaling during the processing of fear-related memory acquisition and recall and subsequent sensitivity to opiate reward memory formation. We report that PFC D4R activation potentiates the salience of normally subthreshold fear conditioning memory cues and simultaneously potentiates the rewarding effects of systemic or intra-ventral tegmental area (VTA) morphine conditioning cues. In contrast, blocking the recall of salient fear memories with intra-PFC D1R activation, blocks the ability of fear memory recall to potentiate systemic or intra-VTA morphine place preference. These effects were dependent upon dissociable PFC phosphorylation states involving calcium-calmodulin-kinase II or extracellular signal-related kinase 1-2, following intra-PFC D4 or D1R activation, respectively. Together, these findings reveal new insights into how aberrant PFC DAergic transmission and associated downstream molecular signaling pathways may modulate fear-related emotional memory processing and concomitantly increase opioid addiction vulnerability.SIGNIFICANCE STATEMENT Post-traumatic stress disorder is highly comorbid with addiction. In this study, we use a translational model of fear memory conditioning to examine how transmission through dopamine D1 or D4 receptors, in the prefrontal cortex (PFC), may differentially control acquisition or recall of fear memories and how these mechanisms might regulate sensitivity to the rewarding effects of opioids. We demonstrate that PFC D4 activation not only controls the salience of fear memory acquisition, but potentiates the rewarding effects of opioids. In contrast, PFC D1 receptor activation blocks recall of fear memories and prevents potentiation of opioid reward effects. Together, these findings demonstrate novel PFC mechanisms that may account for how emotional memory disturbances might increase the addictive liability of opioid-class drugs.
Collapse
Affiliation(s)
| | | | | | - Roger Hudson
- Departments of Anatomy and Cell Biology, and
- Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Walter Rushlow
- Departments of Anatomy and Cell Biology, and
- Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Steven R Laviolette
- Departments of Anatomy and Cell Biology, and
- Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario N6A 5C1, Canada
| |
Collapse
|
11
|
Lisieski MJ, Eagle AL, Conti AC, Liberzon I, Perrine SA. Single-Prolonged Stress: A Review of Two Decades of Progress in a Rodent Model of Post-traumatic Stress Disorder. Front Psychiatry 2018; 9:196. [PMID: 29867615 PMCID: PMC5962709 DOI: 10.3389/fpsyt.2018.00196] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 04/25/2018] [Indexed: 12/21/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a common, costly, and often debilitating psychiatric condition. However, the biological mechanisms underlying this disease are still largely unknown or poorly understood. Considerable evidence indicates that PTSD results from dysfunction in highly-conserved brain systems involved in stress, anxiety, fear, and reward. Pre-clinical models of traumatic stress exposure are critical in defining the neurobiological mechanisms of PTSD, which will ultimately aid in the development of new treatments for PTSD. Single prolonged stress (SPS) is a pre-clinical model that displays behavioral, molecular, and physiological alterations that recapitulate many of the same alterations observed in PTSD, illustrating its validity and giving it utility as a model for investigating post-traumatic adaptations and pre-trauma risk and protective factors. In this manuscript, we review the present state of research using the SPS model, with the goals of (1) describing the utility of the SPS model as a tool for investigating post-trauma adaptations, (2) relating findings using the SPS model to findings in patients with PTSD, and (3) indicating research gaps and strategies to address them in order to improve our understanding of the pathophysiology of PTSD.
Collapse
Affiliation(s)
- Michael J Lisieski
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Andrew L Eagle
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Alana C Conti
- Research and Development Service, John D. Dingell Veterans Affairs Medical Center, Detroit, MI, United States.,Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, United States
| | - Israel Liberzon
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, United States.,Mental Health Service, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, United States
| | - Shane A Perrine
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
12
|
Knox D, Stanfield BR, Staib JM, David NP, DePietro T, Chamness M, Schneider EK, Keller SM, Lawless C. Using c-Jun to identify fear extinction learning-specific patterns of neural activity that are affected by single prolonged stress. Behav Brain Res 2017; 341:189-197. [PMID: 29292158 DOI: 10.1016/j.bbr.2017.12.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/08/2017] [Accepted: 12/28/2017] [Indexed: 01/15/2023]
Abstract
Neural circuits via which stress leads to disruptions in fear extinction is often explored in animal stress models. Using the single prolonged stress (SPS) model of post traumatic stress disorder and the immediate early gene (IEG) c-Fos as a measure of neural activity, we previously identified patterns of neural activity through which SPS disrupts extinction retention. However, none of these stress effects were specific to fear or extinction learning and memory. C-Jun is another IEG that is sometimes regulated in a different manner to c-Fos and could be used to identify emotional learning/memory specific patterns of neural activity that are sensitive to SPS. Animals were either fear conditioned (CS-fear) or presented with CSs only (CS-only) then subjected to extinction training and testing. C-Jun was then assayed within neural substrates critical for extinction memory. Inhibited c-Jun levels in the hippocampus (Hipp) and enhanced functional connectivity between the ventromedial prefrontal cortex (vmPFC) and basolateral amygdala (BLA) during extinction training was disrupted by SPS in the CS-fear group only. As a result, these effects were specific to emotional learning/memory. SPS also disrupted inhibited Hipp c-Jun levels, enhanced BLA c-Jun levels, and altered functional connectivity among the vmPFC, BLA, and Hipp during extinction testing in SPS rats in the CS-fear and CS-only groups. As a result, these effects were not specific to emotional learning/memory. Our findings suggest that SPS disrupts neural activity specific to extinction memory, but may also disrupt the retention of fear extinction by mechanisms that do not involve emotional learning/memory.
Collapse
Affiliation(s)
- Dayan Knox
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States.
| | - Briana R Stanfield
- Center for Molecular and Behavioral Neuroscience, Rutgers University, Newark, NJ 07102, United States
| | - Jennifer M Staib
- Department of Neuroscience, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Nina P David
- School of Public Policy and Administration, University of Delaware, Newark, DE 19716, United States
| | - Thomas DePietro
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - Marisa Chamness
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - Elizabeth K Schneider
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - Samantha M Keller
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| | - Caroline Lawless
- Department of Psychological and Brain Sciences, University of Delaware, Newark, DE 19716, United States
| |
Collapse
|
13
|
Goodman J, McIntyre CK. Impaired Spatial Memory and Enhanced Habit Memory in a Rat Model of Post-traumatic Stress Disorder. Front Pharmacol 2017; 8:663. [PMID: 29018340 PMCID: PMC5614977 DOI: 10.3389/fphar.2017.00663] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 09/06/2017] [Indexed: 01/25/2023] Open
Abstract
High levels of emotional arousal can impair spatial memory mediated by the hippocampus, and enhance stimulus-response (S-R) habit memory mediated by the dorsolateral striatum (DLS). The present study was conducted to determine whether these memory systems may be similarly affected in an animal model of post-traumatic stress disorder (PTSD). Sprague-Dawley rats were subjected to a “single-prolonged stress” (SPS) procedure and 1 week later received training in one of two distinct versions of the plus-maze: a hippocampus-dependent place learning task or a DLS-dependent response learning task. Results indicated that, relative to non-stressed control rats, SPS rats displayed slower acquisition in the place learning task and faster acquisition in the response learning task. In addition, extinction of place learning and response learning was impaired in rats exposed to SPS, relative to non-stressed controls. The influence of SPS on hippocampal spatial memory and DLS habit memory observed in the present study may be relevant to understanding some common features of PTSD, including hippocampal memory deficits, habit-like avoidance responses to trauma-related stimuli, and greater likelihood of developing drug addiction and alcoholism.
Collapse
Affiliation(s)
- Jarid Goodman
- School of Behavioral and Brain Sciences, University of Texas at Dallas, RichardsonTX, United States
| | - Christa K McIntyre
- School of Behavioral and Brain Sciences, University of Texas at Dallas, RichardsonTX, United States
| |
Collapse
|
14
|
He YQ, Lang XQ, Lin L, Ji L, Yuan XY, Chen Q, Ran YM, Chen HS, Li L, Wang JM, Wang ZG, Gregersen H, Zou DW, Liang HP, Yang M. P2X3 receptor-mediated visceral hyperalgesia and neuronal sensitization following exposure to PTSD-like stress in the dorsal root ganglia of rats. Neurogastroenterol Motil 2017; 29. [PMID: 27781340 DOI: 10.1111/nmo.12976] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 09/22/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Patients with posttraumatic stress disorder (PTSD) often share co-morbidity with chronic pain conditions. Recent studies suggest a role of P2X3 receptors and ATP signaling in pain conditions. However, the underlying mechanisms of visceral hyperalgesia following exposure to PTSD-like stress conditions remain unclarified. METHODS The behavior and hormones relevant for PTSD were studied. Visceromotor responses (VMR) and the abdominal withdrawal reflexes (AWR) to colorectal distention (CRD) were recorded to determine P2X3-receptor-mediated alteration of hyperalgesia following single-prolonged stress (SPS) exposure. Immunofluorescence, Western blotting, and patch-clamp were used. KEY RESULTS The escape latency, adrenocorticotropic hormone and cortisol were increased on days 7-14. Visceromotor responses and AWR was reduced at day 1 in SPS rats but increased to higher levels than in controls after exposure to day 7. Intrathecal administration of the P2X3-receptor antagonist TNP-ATP abolished the CRD response. Based on immunofluorescence and Western blotting analysis, SPS-treated rats exhibited reduced P2X3 expression in dorsal root ganglia (DRG) after day 1 compared with controls. P2X3 expression in DRG was enhanced on day 7 after SPS and the increase of the P2X3 expression was maintained on day 14 and 21 compared with controls. The P2X3-receptor agonist α,β-me ATP (10 μM) induced a fast desensitizing inward current in DRG neurons of both control and SPS-treated rats. The average peak current densities in SPS-treated group were increased 3.6-fold. TNP-ATP (100 nM) markedly blocked all fast α,β-me ATP-induced inward currents in the DRG neurons both in control and SPS-treated rats. CONCLUSIONS & INFERENCES The data indicate an important role of P2X3 signaling in visceral hyperalgesia following PTSD-like stress.
Collapse
Affiliation(s)
- Y-Q He
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - X-Q Lang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - L Lin
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - L Ji
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - X-Y Yuan
- Department of Gastroenterology, The Ninth People's Hospital of Chongqing, Chongqing, China
| | - Q Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Y-M Ran
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China
| | - H-S Chen
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Chongqing, China
| | - L Li
- Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - J-M Wang
- Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - Z-G Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China.,Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - H Gregersen
- GIOME and the Key Laboratory for Biorheological Science and Technology of Ministry of Education, Bioengineering College of Chongqing University, Chongqing, China
| | - D-W Zou
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - H-P Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China.,Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - M Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Department of Gastroenterology, Daping Hospital, Third Military Medical University, Chongqing, China.,Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
15
|
Neuro-Epigenetic Indications of Acute Stress Response in Humans: The Case of MicroRNA-29c. PLoS One 2016; 11:e0146236. [PMID: 26730965 PMCID: PMC4711717 DOI: 10.1371/journal.pone.0146236] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/15/2015] [Indexed: 12/17/2022] Open
Abstract
Stress research has progressively become more integrative in nature, seeking to unfold crucial relations between the different phenotypic levels of stress manifestations. This study sought to unravel stress-induced variations in expression of human microRNAs sampled in peripheral blood mononuclear cells and further assess their relationship with neuronal and psychological indices. We obtained blood samples from 49 healthy male participants before and three hours after performing a social stress task, while undergoing functional magnetic resonance imaging (fMRI). A seed-based functional connectivity (FC) analysis was conducted for the ventro-medial prefrontal cortex (vmPFC), a key area of stress regulation. Out of hundreds of microRNAs, a specific increase was identified in microRNA-29c (miR-29c) expression, corresponding with both the experience of sustained stress via self-reports, and alterations in vmPFC functional connectivity. Explicitly, miR-29c expression levels corresponded with both increased connectivity of the vmPFC with the anterior insula (aIns), and decreased connectivity of the vmPFC with the left dorso-lateral prefrontal cortex (dlPFC). Our findings further revealed that miR-29c mediates an indirect path linking enhanced vmPFC-aIns connectivity during stress with subsequent experiences of sustained stress. The correlative patterns of miR-29c expression and vmPFC FC, along with the mediating effects on subjective stress sustainment and the presumed localization of miR-29c in astrocytes, together point to an intriguing assumption; miR-29c may serve as a biomarker in the blood for stress-induced functional neural alterations reflecting regulatory processes. Such a multi-level model may hold the key for future personalized intervention in stress psychopathology.
Collapse
|
16
|
Wu ZM, Zheng CH, Zhu ZH, Wu FT, Ni GL, Liang Y. SiRNA-mediated serotonin transporter knockdown in the dorsal raphe nucleus rescues single prolonged stress-induced hippocampal autophagy in rats. J Neurol Sci 2015; 360:133-40. [PMID: 26723990 DOI: 10.1016/j.jns.2015.11.056] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 11/26/2015] [Accepted: 11/30/2015] [Indexed: 02/02/2023]
Abstract
The neurobiological mechanisms underlying the development of post-traumatic stress disorder (PTSD) remain elusive. One of the hypotheses is the dysfunction of serotonin (5-HT) neurotransmission, which is critically regulated by serotonin transporter (SERT). Therefore, we hypothesized that attenuation of SERT gene expression in the hippocampus could prevent hippocampal autophagy and the development of PTSD-like behavior. To this end, we infused SLC6A4 siRNAs into the dorsal raphe nucleus (DRN) to knockdown SERT gene expression after a single prolonged stress (SPS) treatment in rats. Then, we evaluated the effects of SERT gene knockdown on anxiety-related behaviors and extinction of contextual fear memory. We also examined the histological changes and the expression of Beclin-1, LC3-I, and LC3-II in the hippocampus. We found that SPS treatment did not alter anxiety-related behaviors but prolonged the extinction of contextual fear memory, and such a behavioral phenomenon was correlated with increased hippocampal autophagy, decreased 5-HT level, and increased expression of Beclin-1 and LC3-II/LC3-I ratio in the hippocampus. Furthermore, intra-DRN infusion of SLC6A4 siRNAs promoted the extinction of contextual fear memory, prevented hippocampal autophagy, increased 5-HT level, and decreased expression of Beclin-1 and LC3-II/LC3-I ratio. These results indicated that SERT may play a critical role in the pathogenesis of hippocampal autophagy, and is likely involved in the development of PTSD.
Collapse
Affiliation(s)
- Zhong-Min Wu
- Department of anatomy, Medical College of Taizhou University, Taizhou 318000, China; Department of Neurology, First People's Hospital of Linhai City, Linhai 317000, China
| | - Chun-Hua Zheng
- Outpatient Office, Taizhou Hospital, Taizhou 317000, China
| | - Zhen-Hua Zhu
- Department of Pediatrics, Taizhou Central Hospital, Taizhou 318000, China
| | - Feng-Tian Wu
- City Colloege of Zhejiang University, Hanzhou 310031, China
| | - Gui-Lian Ni
- Department of Neurology, First People's Hospital of Linhai City, Linhai 317000, China
| | - Yong Liang
- Department of anatomy, Medical College of Taizhou University, Taizhou 318000, China.
| |
Collapse
|
17
|
Borges G, Berrocoso E, Mico JA, Neto F. ERK1/2: Function, signaling and implication in pain and pain-related anxio-depressive disorders. Prog Neuropsychopharmacol Biol Psychiatry 2015; 60:77-92. [PMID: 25708652 DOI: 10.1016/j.pnpbp.2015.02.010] [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: 10/24/2014] [Revised: 01/31/2015] [Accepted: 02/15/2015] [Indexed: 12/15/2022]
Abstract
Despite the increasing knowledge regarding pain modulation, the understanding of the mechanisms behind a complex and pathologic chronic pain condition is still insufficient. These knowledge gaps might result in ineffective therapeutic approaches to relieve painful sensations. As a result, severe untreated chronic pain frequently triggers the onset of new disorders such as depression and/or anxiety, and therefore, both the diagnosis and treatment of patients suffering from chronic pain become seriously compromised, prompting a self-perpetuating cycle of symptomatology. The extracellular signal-regulated kinases 1 and 2 (ERK1/2) are molecules strongly implicated in the somatic component of pain at the spinal cord level and have been emerging as mediators of the emotional-affective component as well. Although these molecules might represent good biomarkers, their use as pharmacological targets is still open to discussion as paradoxical information has been obtained. Here we review the current scientific literature regarding ERK1/2 signaling in the modulation of pain, depression and anxiety, including the emotional-affective spheres of the pain experience.
Collapse
Affiliation(s)
- Gisela Borges
- Neuropsycopharmacology and Psychobiology Research Group, Department of Neuroscience (Pharmacology and Psychiatry), University of Cádiz, 11003 Cádiz, Spain; Departamento de Biologia Experimental, Centro de Investigação Médica da Faculdade de Medicina da Universidade do Porto (CIM-FMUP), 4200-319 Porto, Portugal; Grupo de Morfofisiologia do Sistema Nervoso, Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal
| | - Esther Berrocoso
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain; Neuropsychopharmacology and Psychobiology Research Group, Psychobiology Area, Department of Psychology, University of Cádiz, 11510 Cádiz, Spain
| | - Juan Antonio Mico
- Neuropsycopharmacology and Psychobiology Research Group, Department of Neuroscience (Pharmacology and Psychiatry), University of Cádiz, 11003 Cádiz, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Instituto de Salud Carlos III, Madrid, Spain
| | - Fani Neto
- Departamento de Biologia Experimental, Centro de Investigação Médica da Faculdade de Medicina da Universidade do Porto (CIM-FMUP), 4200-319 Porto, Portugal; Grupo de Morfofisiologia do Sistema Nervoso, Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal.
| |
Collapse
|
18
|
Changes in the glucocorticoid receptor and Ca²⁺/calreticulin-dependent signalling pathway in the medial prefrontal cortex of rats with post-traumatic stress disorder. J Mol Neurosci 2014; 56:24-34. [PMID: 25407822 DOI: 10.1007/s12031-014-0464-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 11/07/2014] [Indexed: 12/23/2022]
Abstract
The glucocorticoid receptor (GR), calreticulin (CRT) and protein kinase C (PKC) have all been implicated in the Ca(2+)-dependent signalling pathway, which plays an important role in the plasticity of the central nervous system, learning and memory. The medial prefrontal cortex (mPFC) is known to be involved in mechanisms of learning and memory. In the present study, single prolonged stress (SPS) was used as an animal model of post-traumatic stress disorder (PTSD). The Morris water maze test was used to detect rats' ability for spatial memory and learning. A fluorescence spectrophotometer was used to measure the concentration of intracellular Ca(2+) in mPFC. Immunohistochemistry, immunofluorescence, western blot and reverse transcription polymerase chain reaction were used to explore changes in GR, CRT and PKC in mPFC of SPS rats. The concentration of Ca(2+) in mPFC was increased in the SPS rats. We found increased intensity of GR and CRT immunoreactivity and increased messenger RNA (mRNA) levels of GR, CRT and PKC in mPFC of the SPS groups, although the degree and time of increase was different among them. The protein levels of cytoplasmic GR, cytoplasmic CRT and cytoplasmic pPKC increased in mPFC of the SPS groups, whereas the protein level of nuclear GR decreased in comparison with the control group. As a conclusion, changed CRT and GR/PKC were involved in the mechanism of SPS-induced dysfunctional mPFC.
Collapse
|
19
|
Ji LL, Tong L, Xu BK, Fu CH, Shu W, Peng JB, Wang ZY. Intra-hippocampal administration of ZIP alleviates depressive and anxiety-like responses in an animal model of posttraumatic stress disorder. Behav Brain Funct 2014; 10:28. [PMID: 25178800 PMCID: PMC4158004 DOI: 10.1186/1744-9081-10-28] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 08/13/2014] [Indexed: 01/20/2023] Open
Abstract
Background Given that impairment of fear extinction has been implicated in the pathogenesis of posttraumatic stress disorder (PTSD), effective pharmacological interventions that facilitate fear extinction may provide alternative strategies to conventional treatment. It is generally accepted that the zeta inhibitory peptide (ZIP), a controversial inhibitor of protein kinase M zeta (PKMζ), could erase certain types of previously established long-term memories. However, it is unclear whether ZIP administration may alleviate PTSD-associated depressive and anxiety-like abnormalities. Methods Here we developed a re-stressed single-prolonged stress (SPS) paradigm, a modified prevalent animal model of PTSD, and assayed the expressions of PKMζ in the hippocampus after SPS procedure. Next, Seven days prior to re-stress, ZIP was injected into the hippocampus, and the depressive and anxiety-like behavior was examined by the subsequent forced swim (FS), open-field and elevated plus maze (EPM) test. Results Rats given ZIP prior to FS exhibited a reduction of immobility time in FS test, and more open arms (OA) entries and longer OA duration in EPM. They also spent longer time in the center of the open field. Conclusions Our results suggested that re-stressed SPS could reproduce behavioral alteration similar to that observed in patients with PTSD, and these behavioral symptoms co-morbid with PTSD could be effectively alleviated by the intro-hippocampal administration of ZIP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhen-Yu Wang
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang 110001, People's Republic of China.
| |
Collapse
|
20
|
Activation of extracellular signal-regulated kinase1/2 in the medial prefrontal cortex contributes to stress-induced hyperalgesia. Mol Neurobiol 2014; 50:1013-23. [PMID: 24799176 DOI: 10.1007/s12035-014-8707-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 04/01/2014] [Indexed: 01/07/2023]
Abstract
Stressful stimuli can exacerbate persistent pain disorder. However, the underlying mechanism is still unknown. Here, to reveal the underlying mechanism for stressful stimuli-induced hyperalgesia in chronic pain, we investigated the effect of extracellular signal-regulated kinase1/2 (ERK1/2) activation on pain hypersensitivity using single-prolonged stress (SPS) model, complete Freund's adjuvant (CFA) model and SPS + CFA model. The experimental results revealed significantly reduced paw withdrawal threshold in the SPS, CFA, and SPS + CFA group compared with the control group. However, the increased phosphorylation of ERK1/2 in the medial prefrontal cortex (mPFC) was observed in the SPS- or SPS + CFA-exposed group but not the CFA group compared with control group. There was also a significant increase in mPFC ERK1/2 phosphorylation and mechanical allodynia after SPS + CFA treatment compared to SPS or CFA treatment alone. Furthermore, inhibiting ERK1/2 phosphorylation by microinjection of U0126, a MAPK kinase (MEK) inhibitor, into the mPFC attenuated SPS + CFA- and SPS- but not CFA-induced mechanical allodynia, anxiety-like behavior, and cognitive impairments. These results suggest that the activation of ERK1/2 in the mPFC may contribute to the process of stress-induced cognitive and emotional disorders, leading to an increase in pain sensitivity.
Collapse
|
21
|
Effect of glucose-regulated protein 94 and endoplasmic reticulum modulator caspase-12 in medial prefrontal cortex in a rat model of posttraumatic stress disorder. J Mol Neurosci 2014; 54:147-55. [PMID: 24610447 DOI: 10.1007/s12031-014-0263-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 02/11/2014] [Indexed: 10/25/2022]
Abstract
Posttraumatic stress disorder (PTSD) is an anxiety disorder caused by a life-threatening traumatic experience, which affects a patient's quality of life and social stability. The objective of this study was to investigate the change of the glucose-regulated protein (GRP) 94 and apoptosis-related caspase-12 expression in medial prefrontal cortex (mPFC) in rats to determine whether endoplasmic reticulum apoptosis pathway plays an important role in single-prolonged stress (SPS), a well-established PTSD animal model, and therefore to provide experimental evidence to reveal PTSD pathogenesis. A total of 120 healthy male Wistar rats were selected for this study, randomly divided into a normal control group and SPS groups of 1, 4, 7, 14, and 28 days. Behavioral studies of the learning and memory capabilities of rats were observed by using Morris water maze. Morphological changes were detected using transmission electron microscopy (TEM). Immunohistochemistry, Western blot, and reverse transcription polymerase chain reaction (RT-PCR) were used to detect the expressions of caspase-12 and GRP94 expressions in mPFC. Our results showed that compared with control groups, after the SPS exposure, the average escape latency was prolonged in place navigation test (P < 0.05), and swimming time in the third quadrant in spatial probe test shortened. The morphological change of mPFC in each SPS group bears typical apoptotic characteristics. The expressions of GRP94 and caspase-12 gradually increased on 1 and 4 days, peaked on 7 days after the SPS exposure, and then decreased. These results suggest that SPS exposure can induce apoptotic neurons and a change of caspase-12 and GRP94 expression in the mPFC, which may be one of the pathogenesis of mPFC abnormal function in PTSD.
Collapse
|
22
|
Xiao H, Run X, Cao X, Su Y, Sun Z, Tian C, Sun S, Liang Z. Temperature control can abolish anesthesia-induced tau hyperphosphorylation and partly reverse anesthesia-induced cognitive impairment in old mice. Psychiatry Clin Neurosci 2013; 67:493-500. [PMID: 23992430 DOI: 10.1111/pcn.12091] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Revised: 10/15/2012] [Accepted: 12/12/2012] [Indexed: 12/28/2022]
Abstract
AIMS Anesthesia is related to cognitive impairment and the risk for Alzheimer's disease. Hypothermia during anesthesia can lead to abnormal hyperphosphorylation of tau, which has been speculated to be involved in anesthesia-induced cognitive impairment. The aim of this study was to investigate whether maintenance of the tau phosphorylation level by body temperature control during anesthesia could reverse the cognitive dysfunction in C57BL/6 mice. METHODS Eighteen-month-old mice were repeatedly anesthetized during a 2-week period with or without maintenance of body temperature, control mice were treated with normal saline instead of anesthetics. Tau phosphorylation level in mice brain was detected on western blot, and cognitive performance was measured using the Morris water maze (MWM). RESULTS After anesthesia-induced hypothermia in old mice, tau was hyperphosphorylated and the cognitive performance, measured on MWM, was impaired. When body temperature was controlled during anesthesia, however, the tau hyperphosphorylation was completely avoided, and there was partial recovery in cognitive impairment measured on the MWM. CONCLUSION Hyperphosphorylation of tau in the brain after anesthesia is an important event, and it might be, although not solely, responsible for postoperative cognitive decline.
Collapse
Affiliation(s)
- Haibing Xiao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Neurology, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | | | | | | | | | | | | | | |
Collapse
|
23
|
FGF2 blocks PTSD symptoms via an astrocyte-based mechanism. Behav Brain Res 2013; 256:472-80. [PMID: 24013012 DOI: 10.1016/j.bbr.2013.08.048] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 08/23/2013] [Accepted: 08/28/2013] [Indexed: 12/20/2022]
Abstract
Although posttraumatic stress disorder (PTSD) is characterized by traumatic memories or experiences and increased arousal, which can be partly alleviated by antidepressants, the underlying cellular mechanisms are not fully understood. As emerging studies have focused on the critical role of astrocytes in pathological mood disorders, we hypothesized that several 'astrocyte-related' mechanisms underlying PTSD exist. In the present study, using the single prolonged stress (SPS) model, we investigated the effects of intraperitoneal FGF2 on SPS-induced PTSD behavior response as well as the astrocytic activation after FGF2 administration in SPS rats. Behavioral data showed that intraperitoneal FGF2 inhibited SPS-induced hyperarousal and anxiety behavior; however, immunohistochemistry showed that SPS-induced astrocytic inhibition was activated by intraperitoneal FGF2. Quantitative western blotting showed that intraperitoneal FGF2 up-regulated glial fibrillary acidic protein (GFAP), but not NeuN, expression in the hippocampus. We suggest that intraperitoneal FGF2 could block the SPS-induced fear response and anxiety behavior in PTSD via astrocyte-based but not neuron-based mechanisms.
Collapse
|
24
|
Perrotti LI, Dennis TS, Jiao X, Servatius RJ, Pang KCH, Beck KD. Activation of extracellular signal-regulated kinase (ERK) and ΔFosB in emotion-associated neural circuitry after asymptotic levels of active avoidance behavior are attained. Brain Res Bull 2013; 98:102-10. [PMID: 23932962 DOI: 10.1016/j.brainresbull.2013.07.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 06/20/2013] [Accepted: 07/09/2013] [Indexed: 12/30/2022]
Abstract
Avoidance susceptibility may constitute a vulnerability to develop anxiety disorders, and Wistar-Kyoto (WKY) rats exhibit unique features in their acquisition of avoidance behavior that appear to promote susceptibility to this form of learning, namely the absence of the commonly observed "warm-up" effect. The present study sought to determine if strain differences in acquired avoidance behavior, between WKY and Sprague Dawley rats, could be attributed to differences in dopamine-related plasticity, represented by extracellular signal-regulated kinase (ERK) activity, and prolonged neuronal activation, represented by ΔFosB accumulation, in three key areas of the brain: the medial prefrontal cortex (mPFC), dorsal striatum (DS), and basolateral amygdala (BLA). Consistent with earlier work, WKY rats exhibited a higher level of asymptotic performance of avoidance behavior, which included an absence of warm-up in the first few trials of later training sessions, and they exhibited more non-reinforced anticipatory responses in the single minute prior to the initiation of the first warning signal presentation of each training session. In the brain, phosyphorylated ERK2 (pERK2) activation was higher in avoidance trained rats in both the mPFC and DS, although the difference in DS was mostly observed in WKY rats. Avoidance-training was associated with higher levels of ΔFosB expression in the mPFC of SD rats, but not WKY rats. The strain differences in pERK2 activation in the DS and ΔFosB levels in the mPFC may underlie the strain-specific differences observed in warm-up, the emission of non-reinforced anticipatory responses, and general differences in asymptotic performance of active avoidance behavior. The mPFC and DS require further study as potential neural targets for understanding avoidance susceptibility and, as a result, anxiety vulnerability.
Collapse
Affiliation(s)
- Linda I Perrotti
- Department of Psychology, The University of Texas at Arlington, Arlington, TX 76019, USA.
| | | | | | | | | | | |
Collapse
|
25
|
He YQ, Chen Q, Ji L, Wang ZG, Bai ZH, Stephens RL, Yang M. PKCγ receptor mediates visceral nociception and hyperalgesia following exposure to PTSD-like stress in the spinal cord of rats. Mol Pain 2013; 9:35. [PMID: 23837410 PMCID: PMC3751645 DOI: 10.1186/1744-8069-9-35] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 07/04/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Clinical studies indicate that patients with post-traumatic stress disorder (PTSD) frequently share comorbidity with numerous chronic pain conditions. However, the sustained effects of PTSD-like stress over time on visceral nociception and hyperalgesia have been rarely studied, and the underlying mechanisms of stress-induced modulation of visceral hyperalgesia remain elusive. The purpose of this study was to investigate the characterization of visceral nociception and hyperalgesia over time in rats exposed to PTSD-like stress, and to explore the potential role of protein kinase C gamma (PKCγ) in mediating visceral hyperalgesia following exposure to PTSD-like stress. RESULTS On day 1, the rats exposed to single-prolonged stress (SPS, an established animal model for PTSD) exhibited an analgesic response and its visceromotor response (VMR) to graded colorectal distention (CRD) at 40 and 60 mmHg was reduced compared with the control group (all P < 0.05). On day 6, the VMR returned to the baseline value. However, as early as 7 days after SPS, VMR dramatically increased compared with its baseline value and that in the controls (all P < 0.001) and this increase persisted for 28 days, with the peak on day 9. Abdominal withdrawal reflex (AWR) scores were higher in SPS rats than in controls on days 7, 9, 14, 21 and 28 (all P < 0.001). Intrathecal administration of GF109203X (an inhibitor of PKC gamma), attenuated the SPS-induced increase in both VMR and AWR scores on days 7, 14, 21 and 28 (all P < 0.05). PKCγ protein expression determined by immunofluorescence was reduced in the spinal cord within 3 days after the exposure to SPS (P < 0.01), which returned to normal levels between days 4 and 6, and significantly increased from day 7, and this increase was maintained on days 14, 21, and 28 (all P < 0.001), with the peak on day 9. In addition, Western blotting showed a consistent trend in the changes of PKCγ protein expression. CONCLUSIONS The modified SPS alters visceral sensitivity to CRD, and contributes to the maintenance of visceral hyperalgesia, which is associated with enhanced PKCγ expression in the spinal cord. Functional blockade of the PKCγ receptors attenuates SPS-induced visceral hyperalgesia. Thus, the present study identifies a specific molecular mechanism for visceral hyperalgesia which may pave the way for novel therapeutic strategies for PTSD-like conditions.
Collapse
|
26
|
Treadway MT, Buckholtz JW, Zald DH. Perceived stress predicts altered reward and loss feedback processing in medial prefrontal cortex. Front Hum Neurosci 2013; 7:180. [PMID: 23730277 PMCID: PMC3657626 DOI: 10.3389/fnhum.2013.00180] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 04/22/2013] [Indexed: 12/20/2022] Open
Abstract
Stress is a significant risk factor for the development of psychopathology, particularly symptoms related to reward processing. Importantly, individuals display marked variation in how they perceive and cope with stressful events, and such differences are strongly linked to risk for developing psychiatric symptoms following stress exposure. However, many questions remain regarding the neural architecture that underlies inter-subject variability in perceptions of stressors. Using functional magnetic resonance imaging (fMRI) during a Monetary Incentive Delay (MID) paradigm, we examined the effects of self-reported perceived stress levels on neural activity during reward anticipation and feedback in a sample of healthy individuals. We found that subjects reporting more uncontrollable and overwhelming stressors displayed blunted neural responses in medial prefrontal cortex (mPFC) following feedback related to monetary gains as well monetary losses. This is consistent with preclinical models that implicate the mPFC as a key site of vulnerability to the noxious effects of uncontrollable stressors. Our data help translate these findings to humans, and elucidate some of the neural mechanisms that may underlie stress-linked risk for developing reward-related psychiatric symptoms.
Collapse
Affiliation(s)
- Michael T Treadway
- Department of Psychiatry, Center for Depression, Anxiety and Stress Research, McLean Hospital/Harvard Medical School Belmont, MA, USA
| | | | | |
Collapse
|
27
|
Serova LI, Laukova M, Alaluf LG, Sabban EL. Intranasal infusion of melanocortin receptor four (MC4R) antagonist to rats ameliorates development of depression and anxiety related symptoms induced by single prolonged stress. Behav Brain Res 2013; 250:139-47. [PMID: 23680165 DOI: 10.1016/j.bbr.2013.05.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 04/29/2013] [Accepted: 05/06/2013] [Indexed: 01/13/2023]
Abstract
Brain melanocortinergic systems and specifically melanocortin receptor four (MC4R) are implicated in modulation of anxiety- and depressive-like behavior induced by mild or moderate stress. Here we examine whether blockage of central MC4Rs with HS014 before severe traumatic stress may protect against development of anxiety and depression co-morbid with post-traumatic stress disorder (PTSD). Male rats were treated intranasally (IN) with vehicle or varied doses of HS014, 30min prior to single prolonged stress (SPS) animal model of PTSD. IN administration of 100μg HS014 pre-SPS improved despair behavior in forced swim (FS) immediately after immobilization stress part of SPS protocol. During all 4 intervals of 20min FS these rats spent less time immobile than rats given vehicle or 3.5ng HS014. This dose of HS014 also had a long-term beneficial effect manifested as reduction of immobility time in forced swim test performed after SPS. However, both HS014 doses were effective in ameliorating development of anxiety-like behavior after traumatic stress. Thus, rats given IN HS014 prior to SPS exhibited less open arms (OA) visits in elevated plus maze (EPM), spent longer time in OA and less in closed arms, had lower anxiety index, higher risk assessment and more head dips over borders in OA. They also spent longer time in the center of the open field and defecated less. Reduced grooming behavior in EPM was observed with 100μg HS014. This is the first study revealing pronounced resilience effects of HS014 on development of behavioral symptoms co-morbid with PTSD.
Collapse
Affiliation(s)
- Lidia I Serova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | |
Collapse
|
28
|
Liu H, Han F, Shi Y. Effect of calreticulin on Ca2+/CaM kinaseIIα and endoplasmic reticulum stress in hippocampal in a rat model of post-traumatic stress disorder. Neurochem Res 2013; 38:1407-14. [PMID: 23575574 DOI: 10.1007/s11064-013-1038-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/07/2013] [Accepted: 04/02/2013] [Indexed: 11/24/2022]
Abstract
The purpose of the present study was to examine the changes in the molecular chaperone calreticulin (CRT), calcium signaling pathway Ca(2+)-calmodulin (CaM)-CaM kinaseIIα (CaMKIIα), and the endoplasmic reticulum (ER) apoptotic modulator caspase-12 in hippocampal neurons of rats exposed to single-prolonged stress (SPS), a model of post-traumatic stress disorder (PTSD). Molecular markers and proteins were assessed using immunohistochemistry, western blot and reverse transcript-polymerase chain reaction in rats exposed to SPS at 1 day (1d), 4 and 7 days post-stress and time matched controls. We found that at 7 days, SPS rats had the highest CRT expression. The intracellular free Ca(2+) and the CaM expression reached peak at 1 day post-SPS whereas the CaMKIIα had the opposite trend. Caspase-12 was most active at 4 days and was found to decrease thereafter. Signs of apoptosis were identified using transmission electron microscopy in the rats exposed to SPS. The results indicate that signs of ER stress in the hippocampus of rats exposed to SPS trigger the molecular changes in the intracellular cytoplasm which in turn activate the apoptotic pathway through caspase-12. Therefore, we propose that the hippocampal apoptosis could be one of the pathological mechanisms related to the memory disorders in PTSD.
Collapse
Affiliation(s)
- Hong Liu
- Department of Histology and Embryology, Institute of Pathology and Pathophysiology, Basic Medical Sciences College, China Medical University, 92 North 2nd Road, Shenyang, 110001, Liaoning Province, People's Republic of China
| | | | | |
Collapse
|
29
|
Eagle AL, Knox D, Roberts MM, Mulo K, Liberzon I, Galloway MP, Perrine SA. Single prolonged stress enhances hippocampal glucocorticoid receptor and phosphorylated protein kinase B levels. Neurosci Res 2012. [PMID: 23201176 DOI: 10.1016/j.neures.2012.11.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Animal models of posttraumatic stress disorder (PTSD) can explore neurobiological mechanisms by which trauma enhances fear and anxiety reactivity. Single prolonged stress (SPS) shows good validity in producing PTSD-like behavior. While SPS-induced behaviors have been linked to enhanced glucocorticoid receptor (GR) expression, the molecular ramifications of enhanced GR expression have yet to be identified. Phosphorylated protein kinase B (pAkt) is critical for stress-mediated enhancement in general anxiety and memory, and may be regulated by GRs. However, it is currently unknown if pAkt levels are modulated by SPS, as well as if the specificity of GR and pAkt related changes contribute to anxiety-like behavior after SPS. The current study set out to examine the effects of SPS on GR and pAkt protein levels in the amygdala and hippocampus and to examine the specificity of these changes to unconditioned anxiety-like behavior. Levels of GR and pAkt were increased in the hippocampus, but not amygdala. Furthermore, SPS had no effect on unconditioned anxiety-like behavior suggesting that generalized anxiety is not consistently observed following SPS. The results suggest that SPS-enhanced GR expression is associated with phosphorylation of Akt, and also suggest that these changes are not related to an anxiogenic phenotype.
Collapse
Affiliation(s)
- Andrew L Eagle
- Department of Psychiatry and Behavioral Neuroscience, Wayne State University School of Medicine, Detroit, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
Xiao B, Yu B, Wang HT, Han F, Shi YX. Single-prolonged stress induces apoptosis by activating cytochrome C/caspase-9 pathway in a rat model of post-traumatic stress disorder. Cell Mol Neurobiol 2011; 31:37-43. [PMID: 20803313 PMCID: PMC11498605 DOI: 10.1007/s10571-010-9550-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Accepted: 07/23/2010] [Indexed: 10/19/2022]
Abstract
The purpose of this study was to provide a novel insight into the mechanism of how amygdala might participate in PTSD by investigating the changes of cytochrome c oxidase (COX), caspase-9, and caspase-3 in the amygdala of single-prolonged stress (SPS) rats. A total of 80 healthy, male Wistar rats were selected for this study. The models of post-traumatic stress disorder (PTSD) were created by SPS, which is an established animal model for PTSD. The change of COX was detected by light microscope and transmission electron microscopy (TEM). The expression of caspase-9 and caspase-3 in the basolateral amygdala was examined by immunofluorescence and reverse transcription-polymerase chain reaction (RT-PCR). SPS exposure resulted in a significant change of COX in the SPS model groups compared with the normal control group. Evaluation by enzymohistochemistry indicated translocation of COX from mitochondria to cytoplasm. The expression of both caspase-9 and caspase-3 significantly increased 1 day after SPS stimulation, then gradually increased and peaked at SPS 7d. This findings suggest changes of COX, caspase-9, and caspase-3 in the amygdala of SPS rats, which may play important roles in the pathogenesis of PTSD.
Collapse
Affiliation(s)
- Bing Xiao
- Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 92 North 2nd Road, Shenyang, 110001 Liaoning Province China
| | - Bo Yu
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, 110004 Liaoning Province China
| | - Hai-tao Wang
- Department of Histology and Embryology, North China Coal Medical University, Tangshan, 063000 China
| | - Fang Han
- Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 92 North 2nd Road, Shenyang, 110001 Liaoning Province China
| | - Yu-xiu Shi
- Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, 92 North 2nd Road, Shenyang, 110001 Liaoning Province China
| |
Collapse
|
31
|
Activation of mitogen-activated protein kinase in descending pain modulatory system. JOURNAL OF SIGNAL TRANSDUCTION 2010; 2011:468061. [PMID: 21637376 PMCID: PMC3101953 DOI: 10.1155/2011/468061] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2010] [Accepted: 10/14/2010] [Indexed: 01/10/2023]
Abstract
The descending pain modulatory system is thought to undergo plastic changes following peripheral tissue injury and exerts bidirectional (facilitatory and inhibitory) influence on spinal nociceptive transmission. The mitogen-activated protein kinases (MAPKs) superfamily consists of four main members: the extracellular signal-regulated protein kinase1/2 (ERK1/2), the c-Jun N-terminal kinases (JNKs), the p38 MAPKs, and the ERK5. MAPKs not only regulate cell proliferation and survival but also play important roles in synaptic plasticity and memory formation. Recently, many studies have demonstrated that noxious stimuli activate MAPKs in several brain regions that are components of descending pain modulatory system. They are involved in pain perception and pain-related emotional responses. In addition, psychophysical stress also activates MAPKs in these brain structures. Greater appreciation of the convergence of mechanisms between noxious stimuli- and psychological stress-induced neuroplasticity is likely to lead to the identification of novel targets for a variety of pain syndromes.
Collapse
|