1
|
Shen B, Wang Z, Yu H, Shen X, Li L, Ru Y, Yang C, Du G, Lai C, Gao Y. Medicinal cannabis oil improves anxiety-like and depressive-like behaviors in CCS mice via the BDNF/TRPC6 signaling pathway. Behav Brain Res 2024; 467:115005. [PMID: 38641178 DOI: 10.1016/j.bbr.2024.115005] [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: 02/14/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
BACKGROUND Post-traumatic stress disorder (PTSD) refers to a chronic impairing psychiatric disorder occurring after exposure to the severe traumatic event. Studies have demonstrated that medicinal cannabis oil plays an important role in neuroprotection, but the mechanism by which it exerts anti-PTSD effects remains unclear. METHODS The chronic complex stress (CCS) simulating the conditions of long voyage stress for 4 weeks was used to establish the PTSD mice model. After that, behavioral tests were used to evaluate PTSD-like behaviors in mice. Mouse brain tissue index was detected and hematoxylin-eosin staining was used to assess pathological changes in the hippocampus. The indicators of cell apoptosis and the BDNF/TRPC6 signaling activation in the mice hippocampus were detected by western blotting or real-time quantitative reverse transcription PCR experiments. RESULTS We established the PTSD mice model induced by CCS, which exhibited significant PTSD-like phenotypes, including increased anxiety-like and depression-like behaviors. Medicinal cannabis oil treatment significantly ameliorated PTSD-like behaviors and improved brain histomorphological abnormalities in CCS mice. Mechanistically, medicinal cannabis oil reduced CCS-induced cell apoptosis and enhanced the activation of BDNF/TRPC6 signaling pathway. CONCLUSIONS We constructed a PTSD model with CCS and medicinal cannabis oil that significantly improved anxiety-like and depressive-like behaviors in CCS mice, which may play an anti-PTSD role by stimulating the BDNF/TRPC6 signaling pathway.
Collapse
Affiliation(s)
- Baoying Shen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Zhixing Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; Qinghai University, Xining, Qinghai 810016, China
| | - Huijing Yu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Xin Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Lin Li
- Jilin Sihuan Aokang Pharmaceutical Co., Ltd., Jilin 133400, China
| | - Yi Ru
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Chunqi Yang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; Faculty of Environment and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Guangxu Du
- Jilin Sihuan Aokang Pharmaceutical Co., Ltd., Jilin 133400, China
| | - Chengcai Lai
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| | - Yue Gao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
2
|
Activated cell-cycle CDK4/CyclinD1-pRB-E2F1 signaling pathway is involved in the apoptosis of dorsal raphe nucleus in the rat model of PTSD. Biochem Biophys Res Commun 2022; 602:142-148. [PMID: 35272144 DOI: 10.1016/j.bbrc.2022.02.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/14/2022] [Accepted: 02/26/2022] [Indexed: 11/23/2022]
Abstract
Dysregulation of the dorsal raphe nucleus (DRN) has been revealed to contribute to cognitive and arousal impairments associated with post-traumatic stress disorder (PTSD) in an animal model. In our research an acute exposure to single prolonged stress (SPS) was used to establish PTSD rat model and the effects related to cell-cycle signaling pathway in DRN were examined. Apoptosis in DRN was detected by TUNEL staining, showing that DRN apoptosis number was sharply increased after SPS. SPS triggered cell-cycle CDK4/CyclinD1-pRB-E2F1 signal pathway. Treatment with CDK4 inhibitor Abemaciclib successfully attenuated the DRN apoptosis and rescued decreased spatial learning and memory abilities in SPS rats, indicating that activation of CDK4/CyclinD1-pRB-E2F1 pathway was involved in DRN apoptosis, which may be one of the pathogenesis for PTSD.
Collapse
|
3
|
Yang Z, Huang J, Liao Y, Gan S, Zhu S, Xu S, Shu Y, Lu W. ER Stress is Involved in Mast Cells Degranulation via IRE1α/miR-125/Lyn Pathway in an Experimental Intracerebral Hemorrhage Mouse Model. Neurochem Res 2022; 47:1598-1609. [PMID: 35171433 DOI: 10.1007/s11064-022-03555-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/16/2022] [Accepted: 02/08/2022] [Indexed: 10/25/2022]
Abstract
The degranulation of mast cells accounts for the development of neuroinflammation following intracerebral hemorrhage (ICH). Inhibition of IRE1α, a sensor signaling protein related to endoplasmic reticulum stress, has been shown to exert anti-inflammatory effects in several neurological diseases. The objective of this study was to investigate the effects of IRE1α inhibition on mast cells degranulation in an ICH mouse model and to explore the contribution of miR-125/Lyn pathway in IRE1α-mediated mast cells degranulation. Male mice were subjected to ICH by intraparenchymal injection of autologous blood. STF083010, an inhibitor of IRE1α, was administered intranasally at 1 h after ICH induction. AntimiR-125 was delivered by intracerebroventricular (i.c.v.) injection prior to ICH induction to elucidate the possible mechanisms. Western blot analysis, immunofluorescence staining, neurological test, hematoma volume, brain water content, toluidine blue staining and reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR) were performed. Endogenous phosphorylated IRE1α (p-IRE1α), tryptase, interleukin-17A (IL-17A), tumor necrosis factor α (TNF-α) and tryptase mRNA were increased in time dependent manner while miR-125b-2-3p was decreased after ICH. Inhibition of IRE1α, with STF083010, remarkably reduced brain water content, improved neurological function, decreased hematoma volume, upregulated the expression of miR-125b-2-3p, decreased the number of mast cells, and downregulated the protein expression of Lyn kinase, XBP1s (spliced X-box binding protein-1), tryptase, IL-17A and TNF-α. The downregulation of Lyn kinase, tryptase, IL-17A, TNF-α, and decreased mast cells number were reversed by antimiR-125. The present findings demonstrate that IRE1α inhibition attenuates mast cells degranulation and neuroinflammation, at least partially, through IRE1α/miR-125/Lyn signaling pathway after ICH.
Collapse
Affiliation(s)
- Zhengyu Yang
- Department of Anatomy & Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.,Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Juan Huang
- Department of Anatomy & Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.,Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Yuhui Liao
- Department of Anatomy & Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.,Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Shengwei Gan
- Department of Anatomy & Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.,Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Shujuan Zhu
- Department of Anatomy & Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.,Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Shiye Xu
- Department of Anatomy & Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.,Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Yue Shu
- Department of Anatomy & Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China.,Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Weitian Lu
- Department of Anatomy & Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China. .,Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Feemster JC, Steele TA, Palermo KP, Ralston CL, Tao Y, Bauer DA, Edgar L, Rivera S, Walters-Smith M, Gossard TR, Teigen LN, Timm PC, Richardson JW, Robert Auger R, Kolla B, McCarter SJ, Boeve BF, Silber MH, St. Louis EK. Abnormal rapid eye movement sleep atonia control in chronic post-traumatic stress disorder. Sleep 2021; 45:6484914. [PMID: 34958372 PMCID: PMC8919203 DOI: 10.1093/sleep/zsab259] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 10/12/2021] [Indexed: 12/30/2022] Open
Abstract
STUDY OBJECTIVES Post-traumatic stress disorder (PTSD) and rapid eye movement (REM) sleep behavior disorder (RBD) share some common features including prominent nightmares and sleep disturbances. We aimed to comparatively analyze REM sleep without atonia (RSWA) between patients with chronic PTSD with and without dream enactment behavior (DEB), isolated RBD (iRBD), and controls. METHODS In this retrospective study, we comparatively analyzed 18 PTSD with DEB (PTSD+DEB), 18 PTSD without DEB, 15 iRBD, and 51 controls matched for age and sex. We reviewed medical records to determine PTSD clinical features and quantitatively analyzed RSWA. We used nonparametric analyses to compare clinical and polysomnographic features. RESULTS PTSD patients, both with and without DEB, had significantly higher RSWA than controls (all p < .025, excepting submentalis phasic duration in PTSD+DEB). Most RSWA measures were also higher in PTSD+DEB than in PTSD without DEB patients (all p < .025). CONCLUSIONS PTSD patients have higher RSWA than controls, whether DEB is present or not, indicating that REM sleep atonia control is abnormal in chronic PTSD. Further prospective studies are needed to determine whether neurodegenerative risk and disease markers similar to RBD might occur in PTSD patients.
Collapse
Affiliation(s)
- John C Feemster
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Tyler A Steele
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Kyle P Palermo
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,St. Olaf College, Northfield, MN, USA
| | - Christy L Ralston
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Cornell College, Mount Vernon, IA, USA
| | - Yumeng Tao
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Cornell College, Mount Vernon, IA, USA
| | - David A Bauer
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,St. Olaf College, Northfield, MN, USA
| | - Liam Edgar
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,St. Olaf College, Northfield, MN, USA
| | - Sonia Rivera
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Maxwell Walters-Smith
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Thomas R Gossard
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Medicine, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Luke N Teigen
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Medicine, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Paul C Timm
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Medicine, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Jarrett W Richardson
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Psychiatry, Mayo Clinic and Foundation, Rochester, MN, USA
| | - R Robert Auger
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Psychiatry, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Bhanuprakash Kolla
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Psychiatry, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Stuart J McCarter
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Bradley F Boeve
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Michael H Silber
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Erik K St. Louis
- Mayo Sleep Behavior and Neurophysiology Research Laboratory, Mayo Center for Sleep Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Division of Pulmonary and Critical Care Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Medicine, Mayo Clinic and Foundation, Rochester, MN, USA,Department of Neurology, Mayo Clinic and Foundation, Rochester, MN, USA,Mayo Clinic Health System Southwest Wisconsin, La Crosse, WI, USA,Corresponding author. Erik K. St. Louis, Mayo Center for Sleep Medicine, Departments of Medicine and Neurology, Mayo Clinic College of Medicine, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
5
|
Yang L, Chen Z, Li J, Ding P, Wang Y. Effects of Escitalopram on Endoplasmic Reticulum Stress and Oxidative Stress Induced by Tunicamycin. Front Neurosci 2021; 15:737509. [PMID: 34759791 PMCID: PMC8573126 DOI: 10.3389/fnins.2021.737509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/06/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Major depressive disorder (MDD) was reported to be associated with endoplasmic reticulum stress (ERS) combined with oxidative stress (OS) (ERS/OS). Here, we aimed to investigate the effects of escitalopram (ESC) on blood-brain barrier (BBB) permeability and ERS/OS-related pathways in brain microvascular endothelial cells (bEnd.3 cells) induced by tunicamycin (TM). Methods: bEnd.3 cells were divided into four groups: control, TM, ESC, and ESC + TM groups. CCK-8 and flow cytometry were used to detect cell survival and apoptosis, respectively. The expression levels of proteins involved in cell permeability and ERS/OS-related pathways were assessed by western blot and immunofluorescence. Malondialdehyde (MDA) concentration and superoxide dismutase (SOD) activity were determined by commercial kits. Results: We revealed that TM-induced bEnd.3 cells exhibited remarkably decreased viability and increased apoptosis rate, while ESC treatment reversed these changes. Additionally, TM treatment resulted in markedly increased PERK, GRP78, ATF6, XBP1, and CHOP protein expression levels. On the contrary, the expression of PERK, GRP78, XBP1, and CHOP was obviously reduced in TM-induced bEnd.3 cells after ESC treatment. Moreover, TM significantly reduced the expression of p-eNOS and P-gp and increased the expression of CaMKII and MMP9 compared with the control group. However, ESC reversed these changes in TM-induced bEnd.3 cells. Furthermore, the expression of SOD was significantly decreased, while MDA was significantly increased by TM treatment. In contrast, the expression of SOD was dramatically increased, while MDA was remarkably decreased by ESC treatment. Conclusion: Our results demonstrated that ESC can inhibit ERS/OS and BBB permeability of TM-induced bEnd.3 cells. ESC may alleviate cognitive impairment and prevent comorbidities in MDD patients through ERS/OS.
Collapse
Affiliation(s)
- Lixia Yang
- Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - ZhengHong Chen
- The First Affiliated Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Jie Li
- Department of Psychiatry and Mental Health, Guizhou Medical University, Guiyang, China
| | - PengJin Ding
- Department of Psychiatry and Mental Health, Guizhou Medical University, Guiyang, China
| | - Yiming Wang
- Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
6
|
Hou Y, Li M, Jin Y, Xu F, Liang S, Xue C, Wang K, Zhao W. Protective effects of tetramethylpyrazine on dysfunction of the locus coeruleus in rats exposed to single prolonged stress by anti-ER stress mechanism. Psychopharmacology (Berl) 2021; 238:2923-2936. [PMID: 34231002 DOI: 10.1007/s00213-021-05908-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 06/21/2021] [Indexed: 10/20/2022]
Abstract
Post-traumatic stress disorder (PTSD) is a serious stress-related neuropsychiatric disorder caused by major traumatic events. Abnormal activity of the locus coeruleus (LC)-noradrenergic system is related to the development of PTSD-like symptoms. Our previous studies have indicated that endoplasmic reticulum (ER) stress induced neuronal apoptosis of LC in rats with PTSD. The purpose of this study was to further investigate the role of ER stress pathways in LC neuronal dysfunction and elucidate the effect of the bioactive component tetramethylpyrazine (TMP) against ER stress response. We used an acute exposure to single prolonged stress (SPS) to model PTSD in rats. There were higher norepinephrine (NE) levels in the brain, increased tyrosine hydroxylase expression in LC, and enhanced anxiety-like behaviors in rats exposed to SPS, which were observed by enzyme-linked immunosorbent assay, western blot analysis and elevated plus maze test, respectively. In addition, the three major pathways of ER stress were activated by SPS exposure, which may be involved in the dysregulation of the LC-noradrenergic system of rats with PTSD. Furthermore, we found that TMP administration significantly suppressed the increased responsiveness of LC-noradrenergic system, effectively reduced the anxiety response of SPS rats, and selectively attenuated the activation of pro-apoptotic ER stress pathways. The results suggest that TMP was efficient in improving the LC-NE dysfunction induced by excessive ER stress. TMP exhibited a significant neuroprotective effect and potential therapeutics on PTSD-like symptoms.
Collapse
Affiliation(s)
- Yun Hou
- Department of Histology and Embryology, Binzhou Medical University, No. 346 Guanhai Road, Yantai, 264003, Shandong, China
| | - Meifeng Li
- Department of Intensive Care Unit, Yantai Yuhuangding Hospital Qingdao University, Yantai, 264000, Shandong, China
| | - Yinchuan Jin
- Department of Medical Psychology, Fourth Military Medical University, No. 169 West Changle Road, Xi'an, 710032, China
| | - Feibo Xu
- Department of Histology and Embryology, Binzhou Medical University, No. 346 Guanhai Road, Yantai, 264003, Shandong, China
| | - Shaohua Liang
- Department of Anatomy, Binzhou Medical University, No. 346 Guanhai Road, Yantai, 264003, Shandong, China
| | - Chen Xue
- Clinical Medicine College, Binzhou Medical University, No. 346 Guanhai Road, Yantai, 264003, Shandong, China
| | - Kaili Wang
- Clinical Medicine College, Binzhou Medical University, No. 346 Guanhai Road, Yantai, 264003, Shandong, China
| | - Wei Zhao
- Department of Histology and Embryology, Binzhou Medical University, No. 346 Guanhai Road, Yantai, 264003, Shandong, China.
| |
Collapse
|
7
|
Pillon MC, Gordon J, Frazier MN, Stanley RE. HEPN RNases - an emerging class of functionally distinct RNA processing and degradation enzymes. Crit Rev Biochem Mol Biol 2021; 56:88-108. [PMID: 33349060 PMCID: PMC7856873 DOI: 10.1080/10409238.2020.1856769] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/06/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
HEPN (Higher Eukaryotes and Prokaryotes Nucleotide-binding) RNases are an emerging class of functionally diverse RNA processing and degradation enzymes. Members are defined by a small α-helical bundle encompassing a short consensus RNase motif. HEPN dimerization is a universal requirement for RNase activation as the conserved RNase motifs are precisely positioned at the dimer interface to form a composite catalytic center. While the core HEPN fold is conserved, the organization surrounding the HEPN dimer can support large structural deviations that contribute to their specialized functions. HEPN RNases are conserved throughout evolution and include bacterial HEPN RNases such as CRISPR-Cas and toxin-antitoxin associated nucleases, as well as eukaryotic HEPN RNases that adopt large multi-component machines. Here we summarize the canonical elements of the growing HEPN RNase family and identify molecular features that influence RNase function and regulation. We explore similarities and differences between members of the HEPN RNase family and describe the current mechanisms for HEPN RNase activation and inhibition.
Collapse
Affiliation(s)
- Monica C. Pillon
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Jacob Gordon
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Meredith N. Frazier
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Robin E. Stanley
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, NC 27709, USA
| |
Collapse
|
8
|
Chen L, Xia YF, Shen SF, Tang J, Chen JL, Qian K, Chen Z, Qin ZH, Sheng R. Syntaxin 17 inhibits ischemic neuronal injury by resuming autophagy flux and ameliorating endoplasmic reticulum stress. Free Radic Biol Med 2020; 160:319-333. [PMID: 32828953 DOI: 10.1016/j.freeradbiomed.2020.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Previous studies have shown that syntaxin 17 (STX17) is involved in mediating the fusion of autophagosomes and lysosomes. This study aimed to investigate the role and mechanism of STX17 in neuronal injury following cerebral ischemia/reperfusion. The ischemia/reperfusion (I/R) models were established by transient middle cerebral artery occlusion (tMCAO) in mice and oxygen glucose deprivation/reperfusion (O/R) in primary cultured cortical neurons and HT22 cells. Cerebral ischemia/reperfusion significantly up-regulated the expression of STX17 in neurons. Lentivirus mediated knockdown of STX17 in neurons reduced neuronal viability and increased LDH leakage. Injection of AAV9-shSTX17 into the brain of mice then subjected to tMCAO also significantly augmented the infarct area and exacerbated neurobehavioral deficits and mortality. Depletion of STX17 caused accumulation of autophagic marker/substrate LC3 II and p62, blockade of the autophagic flux, and the accumulation of dysfunctional lysosomes. Knockdown of STX17 also aggravated endoplasmic reticulum (ER) stress-dependent neuronal apoptosis induced by ischemia/reperfusion. Importantly, induction of autophagy-lysosomal pathway and alleviation of ER stress partially rescued STX17 knockdown-induced neuronal damage. These results suggest that STX17 may ameliorate ischemia/reperfusion-induced neuronal damage by enhancing autophagy flux and reducing ER stress-dependent neuronal apoptosis.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Yun-Fei Xia
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Shu-Fang Shen
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Jie Tang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Jia-Li Chen
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Ke Qian
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Zhong Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China.
| |
Collapse
|
9
|
Huang J, Lu W, Doycheva DM, Gamdzyk M, Hu X, Liu R, Zhang JH, Tang J. IRE1α inhibition attenuates neuronal pyroptosis via miR-125/NLRP1 pathway in a neonatal hypoxic-ischemic encephalopathy rat model. J Neuroinflammation 2020; 17:152. [PMID: 32375838 PMCID: PMC7203836 DOI: 10.1186/s12974-020-01796-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/31/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Inhibition of inositol-requiring enzyme-1 alpha (IRE1α), one of the sensor signaling proteins associated with endoplasmic reticulum (ER) stress, has been shown to alleviate brain injury and improve neurological behavior in a neonatal hypoxic-ischemic encephalopathy (HIE) rat model. However, there is no information about the role of IRE1α inhibitor as well as its molecular mechanisms in preventing neuronal pyroptosis induced by NLRP1 (NOD-, LRR- and pyrin domain-containing 1) inflammasome. In the present study, we hypothesized that IRE1α can degrade microRNA-125-b-2-3p (miR-125-b-2-3p) and activate NLRP1/caspased-1 pathway, and subsequently promote neuronal pyroptosis in HIE rat model. METHODS Ten-day old unsexed rat pups were subjected to hypoxia-ischemia (HI) injury, and the inhibitor of IRE1α, STF083010, was administered intranasally at 1 h after HI induction. AntimiR-125 or NLRP1 activation CRISPR was administered by intracerebroventricular (i.c.v) injection at 24 h before HI induction. Immunofluorescence staining, western blot analysis, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), brain infarct volume measurement, neurological function tests, and Fluoro-Jade C staining were performed. RESULTS Endogenous phosphorylated IRE1α (p-IRE1α), NLRP1, cleaved caspase-1, interleukin-1β (IL-1β), and interleukin-18 (IL-18) were increased and miR-125-b-2-3p was decreased in HIE rat model. STF083010 administration significantly upregulated the expression of miR-125-b-2-3p, reduced the infarct volume, improved neurobehavioral outcomes and downregulated the protein expression of NLRP1, cleaved caspase-1, IL-1β and IL-18. The protective effects of STF083010 were reversed by antimiR-125 or NLRP1 activation CRISPR. CONCLUSIONS IRE1α inhibitor, STF083010, reduced neuronal pyroptosis at least in part via miR-125/NLRP1/caspase-1 signaling pathway after HI.
Collapse
Affiliation(s)
- Juan Huang
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
| | - Weitian Lu
- Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
| | - Desislava Met Doycheva
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
| | - Xiao Hu
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Rui Liu
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA
- Department of Anesthesiology, Loma Linda University, Loma Linda, CA, 92350, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, CA, 92350, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Risley Hall, 11041 Campus St, Loma Linda, CA, 92350, USA.
| |
Collapse
|
10
|
Díaz-Hung ML, Martínez G, Hetz C. Emerging roles of the unfolded protein response (UPR) in the nervous system: A link with adaptive behavior to environmental stress? INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 350:29-61. [PMID: 32138903 DOI: 10.1016/bs.ircmb.2020.01.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Stressors elicit a neuroendocrine response leading to increased levels of glucocorticoids, allowing the organism to adapt to environmental changes and maintain homeostasis. Glucocorticoids have a broad effect in the body, modifying the activity of the immune system, metabolism, and behavior through the activation of receptors in the limbic system. Chronic exposition to stressors operates as a risk factor for psychiatric diseases such as depression and posttraumatic stress disorder. Among the cellular alterations observed as a consequence of environmental stress, alterations to organelle function at the level of mitochondria and endoplasmic reticulum (ER) are emerging as possible factors contributing to neuronal dysfunction. ER proteostasis alterations elicit the unfolded protein response (UPR), a conserved signaling network that re-establish protein homeostasis. In addition, in the context of brain function, the UPR has been associated to neurodevelopment, synaptic plasticity and neuronal connectivity. Recent studies suggest a role of the UPR in the adaptive behavior to stress, suggesting a mechanistic link between environmental and cellular stress. Here, we revise recent evidence supporting an evolutionary connection between the neuroendocrine system and the UPR to modulate behavioral adaptive responses.
Collapse
Affiliation(s)
- Mei-Li Díaz-Hung
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Gabriela Martínez
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, United States.
| |
Collapse
|
11
|
Li MX, Li Q, Sun XJ, Luo C, Li Y, Wang YN, Chen J, Gong CZ, Li YJ, Shi LP, Zheng YF, Li RC, Huang XL, Xiong QJ, Chen H. Increased Homer1-mGluR5 mediates chronic stress-induced depressive-like behaviors and glutamatergic dysregulation via activation of PERK-eIF2α. Prog Neuropsychopharmacol Biol Psychiatry 2019; 95:109682. [PMID: 31265863 DOI: 10.1016/j.pnpbp.2019.109682] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/26/2019] [Accepted: 06/27/2019] [Indexed: 10/26/2022]
Abstract
Glutamatergic dysregulation has served as one common pathophysiology of major depressive disorder (MDD) and a promising target for treatment intervention. Previous studies implicate neurotransmission via metabotropic glutamate receptors (mGluRs) and Homer1 in stress-induced anhedonia, but the mechanisms have not been well elucidated. In the present study, we used two different animal models of depression, chronic social defeat stress (CSDS) and chronic restraint stress (CRS), to investigate the expression of Homer1 isoforms and functional interaction with mGluRs. We found that chronic stress selectively upregulated the expression of Homer1b/c in the hippocampus, whereas the level of Homer1a was unchanged. Additionally, there was a significant negative correlation between the levels of Homer1-mGluR5 signaling and depressive-like behaviors. Both application of paired-pulse low-frequency stimulation (PP-LFS) and the selective group 1 mGluRs agonist (RS)-3,5-dihydroxyphenylglycine (DHPG) significantly enhanced mGluR-dependent long-term depression (LTD) at CA3-CA1 pyramidal cell synapses in slices from susceptible mice, whereas there was no change in NMDAR-dependent LTD induced by LFS. Furthermore, these effects were associated with the internalization of surface AMPARs in hippocampal pyramidal neurons, including reduced the expression of AMPARs and amplitude of AMPARs-mediated mEPSC. Finally, we found that chronic stress activated the KR-like ER kinase-eukaryotic initiation factor 2α (PERK-eIF2α) signaling pathway, subsequently phosphorylated cAMP response element binding protein (CREB) at the S129 and reduced the BDNF level, eventually leading to the impairment of synaptic transmission and depressive-like behaviors. Therefore, our study suggests that PERK-eIF2α acts as a critical target downstream of Homer1-mGluR5 complex to mediate chronic stress-induced depressive-like behaviors, and highlights them as a potential target for the treatment of mood disorder.
Collapse
Affiliation(s)
- Ming-Xing Li
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Tongji-Wisconsin Stem Cell Application Technology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Li
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xue-Jiao Sun
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Can Luo
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yong Li
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Ya-Nan Wang
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chen-Zi Gong
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ya-Jie Li
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li-Ping Shi
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yi-Feng Zheng
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Rong-Chun Li
- Department of Pain Management, Wuhan Pu-Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao-Lin Huang
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qiu-Ju Xiong
- Department of Pain Management, Wuhan Pu-Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; Tongji-Wisconsin Stem Cell Application Technology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
12
|
He Y, Ruganzu JB, Lin C, Ding B, Zheng Q, Wu X, Ma R, Liu Q, Wang Y, Jin H, Qian Y, Peng X, Ji S, Zhang L, Yang W, Lei X. Tanshinone IIA ameliorates cognitive deficits by inhibiting endoplasmic reticulum stress-induced apoptosis in APP/PS1 transgenic mice. Neurochem Int 2019; 133:104610. [PMID: 31778727 DOI: 10.1016/j.neuint.2019.104610] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/15/2019] [Accepted: 11/23/2019] [Indexed: 02/06/2023]
Abstract
Our previous data indicated that tanshinone IIA (tan IIA) improves learning and memory in a mouse model of Alzheimer's disease (AD) induced by streptozotocin via restoring cholinergic function, attenuating oxidative stress and blocking p38 MAPK signal pathway activation. This study aims to estimate whether tan IIA inhibits endoplasmic reticulum (ER) stress-induced apoptosis to prevent cognitive decline in APP/PS1 transgenic mice. Tan IIA (10 mg/kg and 30 mg/kg) was intraperitoneally administered to the six-month-old APP/PS1 mice for 30 consecutive days. β-amyloid (Aβ) plaques were measured by immunohistochemisty and Thioflavin S staining, apoptotic cells were observed by TUNEL, ER stress markers and apoptosis signaling proteins were investigated by western blotting and RT-PCR. Our results showed that tan IIA significantly ameliorates cognitive deficits and improves spatial learning ability of APP/PS1 mice in the nest-building test, novel object recognition test and Morris water maze test. Furthermore, tan IIA significantly reduced the deposition of Aβ plaques and neuronal apoptosis, and markedly prevented abnormal expression of glucose regulated protein 78 (GRP78), initiation factor 2α (eIF2α), inositol-requiring enzyme 1α (IRE1α), activating transcription factor 6 (ATF6), as well as suppressed the activation of C/EBP homologous protein (CHOP) and c-Jun N-terminal kinase (JNK) pathways in the parietal cortex and hippocampus. Moreover, tan IIA induced an up-regulation of the Bcl-2/Bax ratio and down-regulation of caspase-3 protein activity. Taken together, the above findings indicated that tan IIA improves learning and memory through attenuating Aβ plaques deposition and inhibiting ER stress-induced apoptosis. These results suggested that tan IIA might become a promising therapeutic candidate drug against AD.
Collapse
Affiliation(s)
- Yingying He
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - John Bosco Ruganzu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Chengheng Lin
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Bo Ding
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Quzhao Zheng
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Xiangyuan Wu
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Ruiyang Ma
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Qian Liu
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Yang Wang
- Medical Undergraduates of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Hui Jin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Yihua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Xiaoqian Peng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Shengfeng Ji
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China
| | - Liangliang Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, 710061, China
| | - Weina Yang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi province, 710061, China.
| | - Xiaomei Lei
- Department of Child Health Care, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi province, 710004, China.
| |
Collapse
|
13
|
Chen X, Jiang Y, Wang J, Liu Y, Xiao M, Song C, Bai Y, Yinuo Han N, Han F. Synapse impairment associated with enhanced apoptosis in post-traumatic stress disorder. Synapse 2019; 74:e22134. [PMID: 31562782 DOI: 10.1002/syn.22134] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/17/2019] [Accepted: 09/20/2019] [Indexed: 12/31/2022]
Abstract
Synapse impairment is associated with post-traumatic stress disorder (PTSD), which is characterized by enhanced apoptosis in the hippocampus, amygdala, and other brain regions. However, there are no detailed studies on the relationship between apoptosis and synaptic connectivity in PTSD. In this review, we discuss results from various studies describing the synaptic changes observed in the PTSD brain. A decreased number of dendrites/spines or increased number of immature spines in the hippocampus, medial prefrontal cortex, and other brain regions has been reported. Studies on axon guidance, myelination, and the cytoskeleton suggest that PTSD may involve axon overgrowth and overbranching. Apoptosis affects synapse formation; low levels of caspase maintain the balance between growth cone attraction and repulsion and inhibit axon elongation. PTSD enhances neuronal apoptosis through caspase activation, which disrupts the balance between growth cone attraction and repulsion and alters growth cone trajectory, leading to axon mistargeting. Meanwhile, caspase activation induces dendritic pruning and dendrite degeneration. These events contribute to the formation of fewer and aberrant synapses, which is associated with enhanced apoptosis in PTSD.
Collapse
Affiliation(s)
- Xinzhao Chen
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yifan Jiang
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Jiayu Wang
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yishu Liu
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Menglei Xiao
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Congshan Song
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Yu Bai
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| | - Nancy Yinuo Han
- Faculty of Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fang Han
- PTSD Laboratory, Department of Histology and Embryology, Basic Medical College, China Medical University, Shenyang, China
| |
Collapse
|
14
|
Luo G, Jian Z, Zhu Y, Zhu Y, Chen B, Ma R, Tang F, Xiao Y. Sirt1 promotes autophagy and inhibits apoptosis to protect cardiomyocytes from hypoxic stress. Int J Mol Med 2019; 43:2033-2043. [PMID: 30864731 PMCID: PMC6443335 DOI: 10.3892/ijmm.2019.4125] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/13/2019] [Indexed: 01/11/2023] Open
Abstract
Sirtuin 1 (Sirt1) exerts its cardioprotective effects in various cardiovascular diseases via multiple cellular activities. However, the therapeutic implications of Sirt1 in hypoxic cardiomyocytes and the underlying mechanisms remain elusive. The present study investigated whether Sirt1 regulates autophagy and apoptosis in hypoxic H9C2 cardiomyocytes and in an experimental hypoxic mouse model. Right ventricular outflow tract biopsies were obtained from patients with cyanotic or acyanotic congenital heart diseases. Adenovirus Ad‑Sirt1 was used to activate Sirt1 and Ad‑Sh‑Sirt1 was used to inhibit Sirt1 expression in H9C2 cells, in order to investigate the effect of Sirt1 on cellular autophagy and apoptosis. SRT1720, a pharmacological activator of Sirt1 and EX‑527, a Sirt1 antagonist, were administered to mice to explore the role of Sirt1 in hypoxic cardiomyocytes in vivo. The levels of autophagy and apoptosis‑related proteins were evaluated using western blotting. Apoptosis was investigated by TUNEL staining and Annexin V/7‑aminoactinomycin D flow cytometry analysis. Heart tissue samples from cyanotic patients exhibited increased autophagy and apoptosis, as well as elevated Sirt1 levels, compared with the noncyanotic control samples. The data from the western blot analysis revealed that Sirt1 promoted autophagic flux and reduced apoptosis in hypoxic H9C2 cells. In addition, Sirt1 activated AMP‑activated protein kinase (AMPK), and the AMPK inhibitor Compound C abolished the effect of Sirt1 on autophagy activation. Further exploration of the mechanism revealed that Sirt1 protects hypoxic cardiomyocytes from apoptosis, at least in part, through inositol requiring kinase enzyme 1α (IRE1α). Consistent with the in vitro results, treatment with the Sirt1 activator SRT1720 activated AMPK, inhibited IRE1α, enhanced autophagy, and decreased apoptosis in the heart tissues of normoxic mice compared with the hypoxia control group. Opposite changes were observed in hypoxic mice treated with the Sirt1 inhibitor EX‑527. These results suggested that Sirt1 promoted autophagy via AMPK activation and reduced hypoxia‑induced apoptosis via the IRE1α pathway, to protect cardiomyocytes from hypoxic stress.
Collapse
Affiliation(s)
- Guiping Luo
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Zhao Jian
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yun Zhu
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yu Zhu
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Baicheng Chen
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Ruiyan Ma
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Fuqin Tang
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Yingbin Xiao
- Department of Cardiovascular Surgery, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
15
|
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
|
16
|
Liu L, Xu L, Zhang S, Wang D, Dong G, Chen H, Li X, Shu C, Wang R. STF-083010, an inhibitor of XBP1 splicing, attenuates acute renal failure in rats by suppressing endoplasmic reticulum stress-induced apoptosis and inflammation. Exp Anim 2018; 67:373-382. [PMID: 29695649 PMCID: PMC6083033 DOI: 10.1538/expanim.17-0131] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is one of the driving forces of ischemia/reperfusion
(IR)-induced acute renal failure (ARF). STF-083010, an inhibitor of the endonuclease
activity of inositol-requiring enzyme-1 (IRE1), has the potential to block the initiation
of a prolonged unfolded protein response (UPR) that is stimulated by ER stress and
alleviates the impairments due to ER stress. In the current study, it was hypothesized
that STF-083010 was capable of ameliorating ER stress-related damages in IR-induced ARF.
Rats were administrated with STF-083010 and were subjected to induction of ARF using a
ligation method. Then the effect of STF-083010 administration on the renal structure and
function, oxidative stress, and inflammation in model rats was assessed. Furthermore, the
levels of expression of UPR members and downstream effectors regulating apoptosis were
detected as well. The results showed that establishment of the ARF model induced ER stress
and impaired the renal structure and function. Administration of STF-083010 ameliorated
impairments in the structure and function of the kidneys and the effect was associated
with the suppressed oxidative stress and inflammation. At the molecular level, STF-083010
inhibited the prolonged UPR by downregulating the expressions of GRP78, p-IRE1, XBP1s,
CHOP, and caspase 3, partially explaining the decreased apoptotic rate. The current study
evaluated the potential of STF-083010 in treating ER stress-induced symptoms in ARF for
the first time, and the findings demonstrated that STF-083010 resulted in effective
treatment outcomes of ARF.
Collapse
Affiliation(s)
- Lei Liu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, Shandong 250021, P.R. China.,Department of General Practice, The Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272000, P.R. China
| | - Lu Xu
- Department of Blood Purification, The Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272000, P.R. China
| | - Shaoqing Zhang
- Department of General Practice, The Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272000, P.R. China
| | - Dong Wang
- Department of Nephrology, The Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272000, P.R. China
| | - Guoxia Dong
- Department of General Practice, The Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272000, P.R. China
| | - Hanwen Chen
- Department of General Practice, The Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272000, P.R. China
| | - Xinjian Li
- Department of Nephrology, The Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining, Shandong 272000, P.R. China
| | - Chi Shu
- High-tech Zone Laboratory of Public Test and Analysis Service, 18-32 Puhe Road, Shenyang 110179, P.R. China
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, 324 Jingwuweiqi Road, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
17
|
Chen D, Dixon BJ, Doycheva DM, Li B, Zhang Y, Hu Q, He Y, Guo Z, Nowrangi D, Flores J, Filippov V, Zhang JH, Tang J. IRE1α inhibition decreased TXNIP/NLRP3 inflammasome activation through miR-17-5p after neonatal hypoxic-ischemic brain injury in rats. J Neuroinflammation 2018; 15:32. [PMID: 29394934 PMCID: PMC5797348 DOI: 10.1186/s12974-018-1077-9] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 01/22/2018] [Indexed: 12/18/2022] Open
Abstract
Background The endoplasmic reticulum (ER) is responsible for the control of correct protein folding and protein function which is crucial for cell survival. However, under pathological conditions, such as hypoxia–ischemia (HI), there is an accumulation of unfolded proteins thereby triggering the unfolded protein response (UPR) and causing ER stress which is associated with activation of several stress sensor signaling pathways, one of them being the inositol requiring enzyme-1 alpha (IRE1α) signaling pathway. The UPR is regarded as a potential contributor to neuronal cell death and inflammation after HI. In the present study, we sought to investigate whether microRNA-17 (miR-17), a potential IRE1α ribonuclease (RNase) substrate, arbitrates downregulation of thioredoxin-interacting protein (TXNIP) and consequent NLRP3 inflammasome activation in the immature brain after HI injury and whether inhibition of IRE1α may attenuate inflammation via miR-17/TXNIP regulation. Methods Postnatal day 10 rat pups (n = 287) were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2). STF-083010, an IRE1α RNase inhibitor, was intranasally delivered at 1 h post-HI or followed by an additional one administration per day for 2 days. MiR-17-5p mimic or anti-miR-17-5p inhibitor was injected intracerebroventricularly at 48 h before HI. Infarct volume and body weight were used to evaluate the short-term effects while brain weight, gross and microscopic brain tissue morphologies, and neurobehavioral tests were conducted for the long-term evaluation. Western blots, immunofluorescence staining, reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR), and co-immunoprecipitation (Co-IP) were used for mechanism studies. Results Endogenous phosphorylated IRE1α expression was significantly increased after HI. Intranasal administration of STF-083010 alleviated brain injury and improved neurological behavior. MiR-17-5p expression was reduced after HI, and this decrease was attenuated by STF-083010 treatment. MiR-17-5p mimic administration ameliorated TXNIP expression, NLRP3 inflammasome activation, caspase-1 cleavage, and IL-1β production, as well as brain infarct volume. Conversely, anti-miR-17-5p inhibitor reversed IRE1α inhibition-induced decrease in TXNIP expression and inflammasome activation, as well as exacerbated brain injury after HI. Conclusions IRE1a-induced UPR pathway may contribute to inflammatory activation and brain injury following neonatal HI. IRE1a activation, through decay of miR-17-5p, elevated TXNIP expression to activate NLRP3 inflammasome and aggravated brain damage. Electronic supplementary material The online version of this article (10.1186/s12974-018-1077-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Di Chen
- Cerebrovascular Diseases Laboratory, Institute of Neuroscience, Chongqing Medical University, Chongqing, 400016, China
| | - Brandon J Dixon
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Desislava M Doycheva
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Bo Li
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Yang Zhang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Qin Hu
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Yue He
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Zongduo Guo
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Derek Nowrangi
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Jerry Flores
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Valery Filippov
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.,Department of Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Jiping Tang
- Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
18
|
Ni H, Rui Q, Li D, Gao R, Chen G. The Role of IRE1 Signaling in the Central Nervous System Diseases. Curr Neuropharmacol 2018; 16:1340-1347. [PMID: 29663887 PMCID: PMC6251047 DOI: 10.2174/1570159x16666180416094646] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 08/07/2017] [Accepted: 02/28/2018] [Indexed: 01/14/2023] Open
Abstract
The accumulation of misfolded or unfolded proteins in endoplasmic reticulum (ER) lumen results in the activation of an adaptive stress process called the unfolded protein response (UPR). As the most conserved signaling branch of the UPR, Inositol-requiring enzyme 1 (IRE1) possesses both Ser/Thr kinase and RNase activities operating as major stress sensors, mediating both adaptive and pro-apoptotic pathways under ER stress. Over the last three decades, a mounting body of evidence has shown that IRE1 signaling dysfunction is involved in the pathology of various neurological disorders. Targeting this pathway has emerged as a promising therapeutic strategy against these diseases. In this review, we provide a general overview about the expression and physiological function of IRE1 signaling and its pathophysiological roles in the central nervous system diseases.
Collapse
Affiliation(s)
| | | | - Di Li
- Address correspondence to this author at the Department of Translational Medicine Center, The First People’s Hospital of Zhangjiagang City, Suzhou, Jiangsu, P.R. China; Tel: +86-18921962599; E-mail:
| | | | | |
Collapse
|
19
|
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
|
20
|
Xiao B, Wang JG, Han F, Shi YX. Effects of calcium-dependent molecular chaperones and endoplasmic reticulum in the amygdala in rats under single‑prolonged stress. Mol Med Rep 2017; 17:1099-1104. [PMID: 29115545 DOI: 10.3892/mmr.2017.7976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 10/17/2017] [Indexed: 11/05/2022] Open
Abstract
The purpose of the present study was to investigate the role of endoplasmic reticulum (ER)‑resident molecular chaperone proteins to identify whether these proteins were involved in post‑traumatic stress disorder (PTSD). The present study detected changes of calreticulin (CRT), calnexin (CNX) and ERp57 in the amygdala of rats, which may with aim of providing a novel insight into the modulation effect of amygdala in PTSD. Single‑prolonged stress (SPS) was applied to create the models of PTSD in rats. The expression levels of CRT, CNX and ERp57 were examined using immunohistochemistry or immunofluorescence, western blot analysis and reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR). The results showed that SPS induced significant changes in CRT, CNX and ERp57 expression levels. Furthermore, the expression levels of CRT, CNX and ERp57 were significantly upregulated when compared to that in the control group after SPS exposure by western blot analysis (P<0.05). RT‑qPCR analysis supported these results, indicating an upregulation of mRNA expression level. Taken together, the present findings suggest that SPS may induce changes to the expression of CRT, CNX and ERp57 in the amygdala of rats. The present study provides an insight into the effects of ER‑resident molecular chaperones in the amygdala participating in PTSD, and provides the experimental basis and a mechanism for the pathophysiology of PTSD.
Collapse
Affiliation(s)
- Bing Xiao
- Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Jian-Gang Wang
- Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Fang Han
- Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Yu-Xiu Shi
- Department of Histology and Embryology, Basic Medical Sciences College, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|