1
|
Kartalci Ş, Özgen N, Kartal F, Arslan AK, Koç A, Düzova H. The Effect of Electroconvulsive Therapy on Hippocampal Endoplasmic Reticulum Stress in a Rat Model of Depression. J ECT 2025:00124509-990000000-00288. [PMID: 40245333 DOI: 10.1097/yct.0000000000001144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
OBJECTIVE The mechanisms underlying the effectiveness of electroconvulsive therapy (ECT), one of the most effective treatments for depression, are still unknown. While the increasing endoplasmic reticulum (ER) stress in depression can retrieve with pharmacological agents, ECT's effectiveness has not been examined yet. In this study, we tested how hippocampal ER stress parameters change after repeated ECT in rats in the chronic unpredictable mild stress (CUMS) model. METHODS Rats were divided into 4 groups. Two groups were included in the experimental group, where the CUMS model was applied for 21 days. Then, the experimental ECT model was applied to 2 groups, one experimental and 1 control group, for 21 days. Weight changes results were evaluated at the end of the 21st and 42nd day. In the final step, 3 prominent ER stress-related proteins (Grp-78, Xbp1, and Atf-4) were measured by western blot in the removed hippocampus. RESULTS We found that rats administered CUMS exhibited depression-like behavioral responses such as weight loss. In CUMS-treated rats (group 3), Grp-78 levels increased, ATF4 levels did not change, and Xbp1 levels decreased. While ECT increased Grp-78 levels in normal rats, it did not change Atf-4 levels and reduced Xbp1 levels (group 2). When ECT was applied to rats undergoing the CUMS model, Grp-78 and Xbp1 levels decreased, while Atf-4 levels did not change (group 4). CONCLUSIONS These findings show that increased ER stress may play a role in the pathophysiology of depression and that this increase can be reversed with ECT treatment. These findings need to be confirmed by clinical studies in humans.
Collapse
Affiliation(s)
- Şükrü Kartalci
- From the Department of Pschiatry, The Private Medicabil Hospital, Bursa, Turkey
| | - Nazmi Özgen
- Department of Biostatististics and Medical İnformatics, İnönü University, Faculty of Medicine, Malatya, Turkey
| | - Fatma Kartal
- Kırıkkale University, Faculty of Medicine, Department of psychiatry, Kırıkkale, Turkey
| | - Ahmet Kadir Arslan
- Department of Biostatististics and Medical İnformatics, İnönü University, Faculty of Medicine, Malatya, Turkey
| | - Ahmet Koç
- Department of Medical Genetics, İnönü University, Faculty of Medicine, Malatya, Turkey
| | - Halil Düzova
- Department Of Physiology, Inonu University, Faculty Of Medicine, Malatya, Turkey
| |
Collapse
|
2
|
Al Bazzal A, Mtairek MA, Awde MH, Kanso H, Hajj F, Al Amin F, Kazan Z, Mohammed NA, Hamdar H. Stress-related psychiatric disorders. PROGRESS IN BRAIN RESEARCH 2025; 291:161-173. [PMID: 40222778 DOI: 10.1016/bs.pbr.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Stress is a natural human emotion that motivates us to face difficulties and risks. Everyone experiences stress to some extent, but when it becomes chronic or reaches a level that cannot be managed, its effects begin to manifest. It is a common condition that most individuals confront, and its effects on the body and brain have become more obvious in recent years. Social and environmental interactions activate systemic reactions primarily controlled by the brain via immunological, neuroendocrine, and metabolic pathways. Long-term stress disrupts homeostasis, activating stress mediators that attempt to restore balance but frequently cause cumulative damage, particularly to the hippocampus, amygdala, and hypothalamus. Furthermore, persistent stress can have a direct and indirect effect on initiating psychiatric illnesses such as depression, anxiety, ADHD, and schizophrenia. Studies on neuroimaging show anatomical and functional alterations in stress-affected regions such as the prefrontal cortex and the hippocampus, which are linked to emotional dysregulation and cognitive decline. To better understand how stress affects psychiatric disorders and exacerbates their symptoms, this chapter will first discuss the molecular mechanism and neurobiological changes it can cause. It will then demonstrate various neuroimaging techniques for studying the effects of stress and offer potential treatments to mitigate these negative effects.
Collapse
Affiliation(s)
- Abbas Al Bazzal
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon; Medical Learning Skills Academy, Beirut, Lebanon
| | - Mohammad Ali Mtairek
- Medical Learning Skills Academy, Beirut, Lebanon; Faculty of Medicine, Damascus University, Damascus, Syria
| | - Mohammad Hadi Awde
- Medical Learning Skills Academy, Beirut, Lebanon; Faculty of Medicine, Damascus University, Damascus, Syria
| | - Haidar Kanso
- Medical Learning Skills Academy, Beirut, Lebanon; Faculty of Medicine, Damascus University, Damascus, Syria
| | - Fatima Hajj
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon; Medical Learning Skills Academy, Beirut, Lebanon
| | - Fatima Al Amin
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon; Medical Learning Skills Academy, Beirut, Lebanon
| | - Zeinab Kazan
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon; Medical Learning Skills Academy, Beirut, Lebanon
| | | | - Hiba Hamdar
- Medical Learning Skills Academy, Beirut, Lebanon.
| |
Collapse
|
3
|
Munshi S, Alarbi AM, Zheng H, Kuplicki R, Burrows K, Figueroa-Hall LK, Victor TA, Aupperle RL, Khalsa SS, Paulus MP, Teague TK, Savitz J. Increased expression of ER stress, inflammasome activation, and mitochondrial biogenesis-related genes in peripheral blood mononuclear cells in major depressive disorder. Mol Psychiatry 2025; 30:574-586. [PMID: 39174649 PMCID: PMC12054637 DOI: 10.1038/s41380-024-02695-2] [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: 11/05/2023] [Accepted: 08/09/2024] [Indexed: 08/24/2024]
Abstract
A subset of major depressive disorder (MDD) is characterized by immune system dysfunction, but the intracellular origin of these immune changes remains unclear. Here we tested the hypothesis that abnormalities in endoplasmic reticulum (ER) stress, inflammasome activity and mitochondrial biogenesis contribute to the development of systemic inflammation in MDD. RT-qPCR was used to measure mRNA expression of key organellar genes from peripheral blood mononuclear cells (PBMCs) isolated from 186 MDD and 67 healthy control (HC) subjects. The comparative CT (2-ΔΔCT) method was applied to quantify mRNA expression using GAPDH as the reference gene. After controlling for age, sex, BMI, and medication status using linear regression models, expression of the inflammasome (NLRC4 and NLRP3) and the ER stress (XBP1u, XBP1s, and ATF4) genes was found to be significantly increased in the MDD versus the HC group. Sensitivity analyses excluding covariates yielded similar results. After excluding outliers, expression of the inflammasome genes was no longer statistically significant but expression of the ER stress genes (XBP1u, XBP1s, and ATF4) remained significant and the mitochondrial biogenesis gene, MFN2, was significantly increased in the MDD group. NLRC4 and MFN2 were positively correlated with serum C-reactive protein concentrations, while ASC trended significant. The altered expression of inflammasome activation, ER stress, and mitochondrial biogenesis pathway components suggest that dysfunction of these organelles may play a role in the pathogenesis of MDD.
Collapse
Affiliation(s)
- Soumyabrata Munshi
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, 1110 N. Stonewall Avenue, Oklahoma City, OK, 73117, USA.
| | - Ahlam M Alarbi
- Integrative Immunology Center, Department of Surgery and Department of Psychiatry, University of Oklahoma - School of Community Medicine, 4502 E. 41st St., Tulsa, OK, 74135, USA
| | - Haixia Zheng
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Rayus Kuplicki
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Kaiping Burrows
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Leandra K Figueroa-Hall
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Teresa A Victor
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
| | - Robin L Aupperle
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California at Los Angeles, 300 UCLA Medical Plaza, Los Angeles, CA, 90095, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| | - T Kent Teague
- Integrative Immunology Center, Department of Surgery and Department of Psychiatry, University of Oklahoma - School of Community Medicine, 4502 E. 41st St., Tulsa, OK, 74135, USA
- Department of Biochemistry and Microbiology, Center for Health Sciences, Oklahoma State University, 1111 W. 17th St., Tulsa, OK, 74107, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, 6655 S. Yale Ave., Tulsa, OK, 74136, USA
- Oxley College of Health and Natural Sciences, The University of Tulsa, Tulsa, OK, 74199, USA
| |
Collapse
|
4
|
Moradi Khankani A, Hossein Meftahi G. Pretreatment with 4-methylumbilliferon improves anxiety-like behaviors and memory impairment in stressed rats via modulation of neuronal cell death and oxidative stress. Brain Res 2024; 1844:149196. [PMID: 39181223 DOI: 10.1016/j.brainres.2024.149196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
This work was done to investigate the ameliorating impact of 4-methylumbilliferon (4-MU) on spatial learning and memory dysfunction and restraint stress (STR)-induced anxiety-like behaviors in male Wistar rats and the underlying mechanisms. Thirty-two animals were assigned into 4 cohorts: control, 4-MU, STR, and STR+4-MU. Animals were exposed to STR for 4 h per day for 14 consecutive days or kept in normal conditions (healthy animals without exposure to stress). 4-MU (25 mg/kg) was intraperitoneally administered once daily to STR rats before restraint stress for 14 consecutive days. The behavioral tests were performed through Morris water maze tests and elevated-plus maze to examine learning/memory function, and anxiety levels, respectively. The levels of the antioxidant defense biomarkers (GPX, SOD) and MDA as an oxidant molecule in the brain tissues were measured using commercial ELISA kits. Neuronal loss or density of neurons was evaluated using Nissl staining. STR exposure could cause significant alterations in the levels of the antioxidant defense biomarkers (MDA, GPX, and SOD) in the prefrontal cortex and hippocampus, induce anxiety, and impair spatial learning and memory function. Treatment with 4-MU markedly reduced anxiety levels and improved spatial learning and memory dysfunction via restoring the antioxidant defense biomarkers to normal values and reducing MDA levels. Moreover, more intact cells with normal morphologies were detected in STR-induced animals treated with 4-MU. 4-MU could attenuate the STR-induced anxiety-like behaviors and spatial learning and memory dysfunction by reducing oxidative damage and neuronal loss in the prefrontal cortex and hippocampus region. Taken together, our findings provide new insights regarding the potential therapeutic effects of 4-MU against neurobehavioral disorders induced by STR.
Collapse
Affiliation(s)
| | - Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Fujii C, Zorumski CF, Izumi Y. Endoplasmic reticulum stress, autophagy, neuroinflammation, and sigma 1 receptors as contributors to depression and its treatment. Neural Regen Res 2024; 19:2202-2211. [PMID: 38488553 PMCID: PMC11034583 DOI: 10.4103/1673-5374.391334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/02/2023] [Accepted: 11/24/2023] [Indexed: 04/24/2024] Open
Abstract
The etiological factors contributing to depression and other neuropsychiatric disorders are largely undefined. Endoplasmic reticulum stress pathways and autophagy are well-defined mechanisms that play critical functions in recognizing and resolving cellular stress and are possible targets for the pathophysiology and treatment of psychiatric and neurologic illnesses. An increasing number of studies indicate the involvement of endoplasmic reticulum stress and autophagy in the control of neuroinflammation, a contributing factor to multiple neuropsychiatric illnesses. Initial inflammatory triggers induce endoplasmic reticulum stress, leading to neuroinflammatory responses. Subsequently, induction of autophagy by neurosteroids and other signaling pathways that converge on autophagy induction are thought to participate in resolving neuroinflammation. The aim of this review is to summarize our current understanding of the molecular mechanisms governing the induction of endoplasmic reticulum stress, autophagy, and neuroinflammation in the central nervous system. Studies focused on innate immune factors, including neurosteroids with anti-inflammatory roles will be reviewed. In the context of depression, animal models that led to our current understanding of molecular mechanisms underlying depression will be highlighted, including the roles of sigma 1 receptors and pharmacological agents that dampen endoplasmic reticulum stress and associated neuroinflammation.
Collapse
Affiliation(s)
- Chika Fujii
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Charles F. Zorumski
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Yukitoshi Izumi
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
6
|
Gong X, Fan Z, Xu H, Qu Y, Li B, Li L, Yan Y, Wu L, Yan C. GABAergic interneurons in the hippocampal CA1 mediate contextual fear generalization in PTSD rats. J Neurochem 2024; 168:2587-2600. [PMID: 38705582 DOI: 10.1111/jnc.16123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/08/2024] [Accepted: 04/22/2024] [Indexed: 05/07/2024]
Abstract
Fear overgeneralization is widely accepted as a pathogenic marker of post-traumatic stress disorder (PTSD). Recently, GABAergic interneurons have been regarded as key players in the regulation of fear memory. The role of hippocampal GABAergic interneurons in contextual fear generalization of PTSD remains incompletely understood. In the present study, we established a rat model of PTSD with inescapable foot shocks (IFS) and observed the loss of GABAergic interneuron phenotype in the hippocampal cornu ammonis-1 (CA1) subfield. To determine whether the loss of GABAergic interneuron phenotype was associated with fear generalization in PTSD rats, we used adeno-associated virus (AAV) to reduce the expression of GAD67 in CA1 and observed its effect on fear generalization. The results showed that the reduction of GAD67 in CA1 enhanced contextual fear generalization in rats. We investigated whether the PERK pathway was involved in the GABAergic interneuron injury. Increased expression of p-PERK, CHOP, and Caspase12 in GABAergic interneurons of PTSD rats was observed. Then, we used salubrinal, an endoplasmic reticulum stress inhibitor, to modulate the PERK pathway. The salubrinal treatment significantly protected the GABAergic interneurons and relieved fear generalization in PTSD rats. In addition, the results showed that salubrinal down-regulated the expression of CHOP and Caspase12 in GABAergic interneurons of PTSD rats. In conclusion, this study provided evidence that the loss of GABAergic interneuron phenotype in CA1 may contribute to contextual fear generalization in PTSD. The PERK pathway is involved in the GABAergic interneuron injury of PTSD rats and modulating it can protect GABAergic interneurons and constrain contextual fear generalization.
Collapse
Affiliation(s)
- Xiayu Gong
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhixin Fan
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hanfang Xu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Qu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bozhi Li
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lanxin Li
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuqi Yan
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lili Wu
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Can Yan
- Research Center for Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
7
|
Hofstra BM, Hoeksema EE, Kas MJH, Verbeek DS. Cross-species analysis uncovers the mitochondrial stress response in the hippocampus as a shared mechanism in mouse early life stress and human depression. Neurobiol Stress 2024; 31:100643. [PMID: 38800537 PMCID: PMC11127276 DOI: 10.1016/j.ynstr.2024.100643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/03/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Depression, or major depressive disorder, poses a significant burden for both individuals and society, affecting approximately 10.8% of the general population. This psychiatric disorder leads to approximately 800,000 deaths per year. A combination of genetic and environmental factors such as early life stress (ELS) increase the risk for development of depression in humans, and a clear role for the hippocampus in the pathophysiology of depression has been shown. Nevertheless, the underlying mechanisms of depression remain poorly understood, resulting in a lack of effective treatments. To better understand the core mechanisms underlying the development of depression, we used a cross-species design to investigate shared hippocampal pathophysiological mechanisms in mouse ELS and human depression. Mice were subjected to ELS by a maternal separation paradigm, followed by RNA sequencing analysis of the adult hippocampal tissue. This identified persistent transcriptional changes linked to mitochondrial stress response pathways, with oxidative phosphorylation and protein folding emerging as the main mechanisms affected by maternal separation. Remarkably, there was a significant overlap between the pathways involved in mitochondrial stress response we observed and publicly available RNAseq data from hippocampal tissue of depressive patients. This cross-species conservation of changes in gene expression of mitochondria-related genes suggests that mitochondrial stress may play a pivotal role in the development of depression. Our findings highlight the potential significance of the hippocampal mitochondrial stress response as a core mechanism underlying the development of depression. Further experimental investigations are required to expand our understanding of these mechanisms.
Collapse
Affiliation(s)
- Bente M. Hofstra
- Department of Genetics, University of Groningen, University Medical Center Groningen, the Netherlands
- Department of Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | - Emmy E. Hoeksema
- Department of Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | - Martien JH. Kas
- Department of Behavioural Neuroscience, Groningen Institute for Evolutionary Life Sciences, University of Groningen, the Netherlands
| | - Dineke S. Verbeek
- Department of Genetics, University of Groningen, University Medical Center Groningen, the Netherlands
| |
Collapse
|
8
|
Shayan E, Maheri F, Aflaki F, Mousavi SE, Zarrindast MR, Fakhraei N, Rezayat Sorkhabadi SM, Shushtarian SMM. Synergistic effects of citicoline and silymarin nanomicelles in restraint stress-exposed mice. Behav Brain Res 2024; 464:114929. [PMID: 38428646 DOI: 10.1016/j.bbr.2024.114929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 02/12/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
This study evaluated the effects of citicoline and silymarin nanomicelles (SMnm) in repeated restraint stress (RRS). METHOD Mice were exposed to RRS for four consecutive days, 2 hrs. daily. On day 5 of the study, SMnm (25 and 50 mg/kg, i.p.) and citicoline (25 and 75 mg/kg), and a combination of them (25 mg/kg, i.p.) were initiated. On day 18, anxiety-like behavior, behavioral despair, and exploratory behavior were evaluated. The prefrontal cortex (PFC) and the hippocampus were dissected measuring brain-derived neurotrophic factor (BDNF), cAMP response element-binding protein (CREB), and tumor necrosis factor-alpha (TNF-α) through Western Blot and ELISA, respectively. RESULTS In RR-exposed mice, anxiety-like behavior in the elevated plus maze (EPM) was enhanced by reductions in open arm time (OAT%) P < 0.001, and open arm entry (OAE%) P < 0.001. In the forced swimming test (FST), the immobility increased P < 0.001 while the swimming and climbing reduced P < 0.001. In the open field test (OFT), general motor activity was raised P < 0.05. Further, body weights reduced P < 0.001, and tissue BDNF and pCREB expressions decreased P < 0.001 while TNF-α increased P < 0.001. Conversely, SMnm, citicoline and their combination could reduce anxiety-like behavior P < 0.001. The combination group reduced the depressive-like behaviors P < 0.001. Moreover, body weights were restored P < 0.001. Besides, BDNF and pCREB expressions increased while TNF-α reduced, P < 0.001. CONCLUSION The combination synergistically improved emotion-like behaviors, alleviating the inflammation and upregulating the hippocampal BDNF-mediated CREB signaling pathway.
Collapse
Affiliation(s)
- Elham Shayan
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Fatemeh Maheri
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Fatemeh Aflaki
- Department of Biophysics and Biochemistry, Faculty of Advance Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Seyyedeh-Elaheh Mousavi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Mohammad-Reza Zarrindast
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Nahid Fakhraei
- Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed-Mohammad-Masoud Shushtarian
- Department of Biophysics and Biochemistry, Faculty of Advance Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
9
|
Liu Y, Sun Z, Sun Q, Wang L, Wang C, Li Y, Ma C, Shi W, Zhang G, Dong Y, Zhang X, Cong B. The effects of restraint stress on ceramide metabolism disorders in the rat liver: the role of CerS6 in hepatocyte injury. Lipids Health Dis 2024; 23:68. [PMID: 38431645 PMCID: PMC10908211 DOI: 10.1186/s12944-024-02019-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/15/2024] [Indexed: 03/05/2024] Open
Abstract
BACKGROUND Stress is implicated in various pathological conditions leading to liver injury. Existing evidence suggests that excessive stress can induce mitochondrial damage in hepatocytes, yet the underlying mechanism remains unclear. Ceramide synthase 6 (CerS6)-derived C16:0 ceramide is recognised as a lipotoxic substance capable of causing mitochondrial damage. However, the role of CerS6 in stress has received insufficient attention. This study aimed to explore the involvement of CerS6 in stress-induced hepatic damage and its associated mechanisms. METHODS The rat restraint stress model and a corticosterone (CORT)-induced hepatocyte stress model were employed for in vivo and in vitro experimental analyses, respectively. Changes in mitochondrial damage and ceramide metabolism in hepatocytes induced by stress were evaluated. The impact of CORT on mitochondrial damage and ceramide metabolism in hepatocytes was assessed following CerS6 knockdown. Mitochondria were isolated using a commercial kit, and ceramides in liver tissue and hepatocytes were detected by LC-MS/MS. RESULTS In comparison to the control group, rats subjected to one week of restraint exhibited elevated serum CORT levels. The liver displayed significant signs of mitochondrial damage, accompanied by increased CerS6 and mitochondrial C16:0 ceramide, along with activation of the AMPK/p38 MAPK pathway. In vitro studies demonstrated that CORT treatment of hepatocytes resulted in mitochondrial damage, concomitant with elevated CerS6 and mitochondrial C16:0 ceramide. Furthermore, CORT induced sequential phosphorylation of AMPK and p38 MAPK proteins, and inhibition of the p38 MAPK pathway using SB203580 mitigated the CORT-induced elevation in CerS6 protein. Knocking down CerS6 in hepatocytes inhibited both the increase in C16:0 ceramide and the release of mitochondrial cytochrome c induced by CORT. CONCLUSIONS CerS6-associated C16:0 ceramide plays a mediating role in stress-induced mitochondrial damage in hepatocytes. The molecular mechanism is linked to CORT-induced activation of the AMPK/p38 MAPK pathway, leading to upregulated CerS6.
Collapse
Affiliation(s)
- Yichang Liu
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
- Department of Forensic Medicine, College of Medicine, Nantong University, Nantong, 226000, China
| | - Zhaoling Sun
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Qiuli Sun
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Li Wang
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Chuan Wang
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Yingmin Li
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Chunling Ma
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Weibo Shi
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Guozhong Zhang
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
- Hebei Province Laboratory of Experimental Animal, Shijiazhuang, 050017, China
| | - Yiming Dong
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China
| | - Xiaojing Zhang
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China.
| | - Bin Cong
- Department of Forensic Medicine, Hebei Medical University, No. 361 Zhong Shan Rd, Shijiazhuang, 050017, Hebei, China.
- Hainan Tropical Forensic Medicine Academician Workstation, Haikou, 571199, China.
| |
Collapse
|
10
|
Munshi S, Alarbi A, Zheng H, Kuplicki R, Burrows K, Figueroa-Hall L, Victor T, Aupperle R, Khalsa S, Paulus M, Teague TK, Savitz J. Increased expression of ER stress, inflammasome activation, and mitochondrial biogenesis-related genes in peripheral blood mononuclear cells in major depressive disorder. RESEARCH SQUARE 2024:rs.3.rs-3564760. [PMID: 38260352 PMCID: PMC10802690 DOI: 10.21203/rs.3.rs-3564760/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
A subset of major depressive disorder (MDD) is characterized by immune system dysfunction, but the intracellular origin of these immune changes remains unclear. Here we tested the hypothesis that abnormalities in the endoplasmic reticulum (ER) stress, inflammasome activity and mitochondrial biogenesis contribute to the development of systemic inflammation in MDD. RT-qPCR was used to measure mRNA expression of key organellar genes from peripheral blood mononuclear cells (PBMCs) isolated from 186 MDD and 67 healthy control (HC) subjects. The comparative CT (2-ΔΔCT) method was applied to quantify mRNA expression using GAPDH as the reference gene. After controlling for age, sex, BMI, and medication status using linear regression models, expression of the inflammasome (NLRC4 and NLRP3) and the ER stress (XBP1u, XBP1s, and ATF4) genes was found to be significantly increased in the MDD versus the HC group. After excluding outliers, expression of the inflammasome genes was no longer statistically significant but expression of the ER stress genes (XBP1u, XBP1s, and ATF4) and the mitochondrial biogenesis gene, MFN2, was significantly increased in the MDD group. ASC and MFN2 were positively correlated with serum C-reactive protein concentrations. The altered expression of inflammasome activation, ER stress, and mitochondrial biogenesis pathway components suggest that dysfunction of these organelles may play a role in the pathogenesis of MDD.
Collapse
|
11
|
Ghaffari-Nasab A, Javani G, Yousefi H, Sharafkhani R, Taghizadeh S. Prolonged stress-induced depression-like behaviors in aged rats are mediated by endoplasmic reticulum stress and apoptosis in the hippocampus. Neurosci Res 2024; 198:39-46. [PMID: 37392834 DOI: 10.1016/j.neures.2023.06.011] [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: 03/28/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/03/2023]
Abstract
Structural and functional recovery from stress-induced depression is impaired in the context of aging brain. Since investigating the molecular substrates that facilitate behavioral recovery may have important implications for understanding brain plasticity and resilience of individuals, we studied depressive-like behaviors in young and aged rats 6 weeks after chronic stress exposure as a recovery period and examined the levels of TNF-α and IL-6 inflammatory cytokines, NADH oxidase activity, NADPH oxidase, endoplasmic reticulum (ER) stress markers, and apoptosis in the hippocampus. Young (3 months old) and aged (22 months old) male Wistar rats were divided into four groups; young control (Young), depression model of young rats that received chronic stress procedure followed by a 6-week recovery period (Young+S), aged control (Aged), and depression model of aged rats that received chronic stress procedure followed by a 6-week recovery period (Aged+S). After the recovery period, aged but not young rats showed depression-like behaviors, evaluated by the sucrose preference test (SPT) and forced swimming test (FST), coincided with the altered levels of TNF-α, IL-6, NADH oxidase activity, NADPH oxidase, GRP78, CHOP, and cleaved caspase-12 in the hippocampus of these animals. These data suggested that oxidative and ER stress-induced apoptosis in the aging hippocampus may affect the recovery-related outcomes after the stress paradigm.
Collapse
Affiliation(s)
- Arshad Ghaffari-Nasab
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, the Islamic Republic of Iran
| | - Gonja Javani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, the Islamic Republic of Iran
| | - Hadi Yousefi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, the Islamic Republic of Iran.
| | - Rahim Sharafkhani
- School of Health, Khoy University of Medical Sciences, Khoy, the Islamic Republic of Iran
| | - Sajjad Taghizadeh
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, the Islamic Republic of Iran
| |
Collapse
|
12
|
Yang N, Wang Y, Luo X, Zhan G. Chronic restraint stress induces abnormal behaviors in pain sensitivity and cognitive function in mice: the role of Keap1/Nrf2 pathway. Stress 2024; 27:2316050. [PMID: 38377152 DOI: 10.1080/10253890.2024.2316050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/03/2024] [Indexed: 02/22/2024] Open
Abstract
Stress is a series of physical and psychological responses to external and internal environmental stimuli. Growing studies have demonstrated the detrimental impacts of acute restraint stress (ARS) and chronic restraint stress (CRS) on animal behavior. However, the related pathogenesis and therapeutic mechanisms remain unclear. Hence, the present study aimed to examine whether unfolded protein response (UPR) and Kelch-like ECH-associated protein 1 (Keap1)-nuclear factor erythroid 2 related factor 2 (Nrf2) pathway are associated with ARS- and CRS- induced abnormal behaviors of pain sensitivity and cognitive function. We here used four behavioral tests to evaluate pain sensitivity and cognitive function in ARS and CRS mice. CRS markedly decreased Paw Withdrawal Mechanical Threshold (PWMT) and Tail-flick Latency (TFL) scores, whereas ARS altered TFL but had no effect on PWMT scores. Additionally, CRS, but not ARS, significantly changed behaviors in nest building behavior and MWMT. Intriguingly, the expression of Keap1 and Nrf2 protein were decreased in the spinal cord and hippocampus in CRS mice, but not in ARS mice. Moreover, neither the ARS nor the CRS groups significantly differed from the control group in terms of endoplasmic reticulum stress (ERS). Taken together, this study demonstrated that CRS could induce abnormal pain sensitivity and cognitive function probably via Keap1/Nrf2 pathway in spinal cord and hippocampus. It is therefore likely that effective intervention of Keap1/Nrf2 pathway may contribute to preventing and treating hyperalgesia and cognitive dysfunction in CRS.
Collapse
Affiliation(s)
- Ning Yang
- Department of Anesthesiology, Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Wang
- Department of Anesthesiology, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Luo
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gaofeng Zhan
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
13
|
Meftahi GH, Aboutaleb N. Gallic acid ameliorates behavioral dysfunction, oxidative damage, and neuronal loss in the prefrontal cortex and hippocampus in stressed rats. J Chem Neuroanat 2023; 134:102364. [PMID: 38016595 DOI: 10.1016/j.jchemneu.2023.102364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 11/30/2023]
Abstract
Gallic acid (GA) is known to be a natural phenolic compound with antioxidant and neuroprotective effects. This study aims to investigate the impact of GA against restraint stress-induced oxidative damage, anxiety-like behavior, neuronal loss, and spatial learning and memory impairment in male Wistar rats. The animals were divided into four groups (n = 8) and subjected to restraint stress for 4 h per day for 14 consecutive days or left undisturbed (control without inducing stress). In the treatment group, the animals were treated with 2 mL normal saline plus 100 mg/kg GA per day for 14 consecutive days (STR + GA group). The animals received the drug or normal saline by gavage 2 h before inducing restraint stress. ELISA assay measured oxidative stress factors. Elevated-plus maze and Morris water maze tests assessed anxiety-like behavior and spatial learning and memory, respectively. Also, neuronal density was determined using Nissl staining. Restraint stress significantly increased MDA and reduced the activities of GPX and SOD in the stressed rats, which were reserved by treatment with 100 mg/kg GA. Restraint stress markedly enhanced the anxiety-like behavior and spatial learning and memory impairment that were reserved by GA. In addition, treatment with GA reduced the neuronal loss in the stressed rats in the hippocampus and prefrontal cortex (PFC) regions. Taken together, our findings suggest that GA has the potential to be used as a good candidate to attenuate neurobehavioral disorders as well as neuronal loss in the hippocampus and PFC induced by restraint stress via reducing oxidative damage.
Collapse
Affiliation(s)
- Gholam Hossein Meftahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Nahid Aboutaleb
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Physiology Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Tang M, Liu T, Shen Y, Wang L, Xue Y, Zhao T, Xie K, Gong Z, Yin T. Potential antidepressant-like effects of N-3 polyunsaturated fatty acids through inhibition of endoplasmic reticulum stress. Psychopharmacology (Berl) 2023; 240:1877-1889. [PMID: 37612456 DOI: 10.1007/s00213-023-06377-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/25/2023] [Indexed: 08/25/2023]
Abstract
RATIONALE The growing evidence has demonstrated the importance of endoplasmic reticulum stress (ERS) in the pathophysiology of depression. ERS genes were considered to be potential novel therapeutic targets for depression. OBJECTIVES To clarify the mechanisms of the chronic unpredictable mild stress (CUMS)-induced ERS response and the potential contributing pathways in depression, and further investigate the potential link between N-3 polyunsaturated fatty acids (PUFAs) and stress-induced ERS disturbances. METHODS This study analyzed the expression of ERS-related genes including GRP78, ATF-4, ATF-6, XBP-1, and CHOP, and sigma-1R with real-time PCR in peripheral blood mononuclear cell (PBMC) RNA samples from participants. All of the rats except for those in the control groups were subjected to 5 consecutive weeks of CUMS to establish the depression model, and the antidepressant effects of N-3 PUFAs were observed by behavior tests. Moreover, the effect of diet and stress on the ERS pathways was also investigated using the western blot. RESULTS Blood CHOP, ATF-4, and XBP-1 levels were notably elevated in depressed patients relative to healthy individuals. Moreover, increased sigma-1R and decreased ATF-6 implied the protective role of sigma-1R through modulating ERS in patients with depression. Animal studies disclosed the novel findings that supplementary N-3 PUFAs in rats alleviated CUMS-induced disturbance of ERS through the ATF-4/XBP-1/CHOP pathway, implying its potential strategy for depression. CONCLUSION CUMS-induced depressive-like behaviors are related to the disturbance of ERS. Furthermore, supplementary N-3 PUFAs might be an effective way to alleviate ERS.
Collapse
Affiliation(s)
- Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Ting Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Yanmei Shen
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Lu Wang
- Mental Health Institute of the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ying Xue
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Tingyu Zhao
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Kaiqiang Xie
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
- Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
| | - Tao Yin
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
- Institute of Hospital Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
| |
Collapse
|
15
|
Zhang H, Zhang C, Xu D, Wang Q, Xu D. Effects of subchronic exposure of perfluorooctane sulfonate on cognitive function of mice and its mechanism. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 329:121650. [PMID: 37062406 DOI: 10.1016/j.envpol.2023.121650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is an emerging persistent organic pollutant, and its potential impact on cognitive function remains unclear. We adopted the C57BL/6J mouse model to investigate the effect of PFOS on cognitive function, as well as the underlying mechanisms. Subchronic exposure was performed by administering PFOS via drinking water for 6 months (at doses of 0, 0.2, and 2.0 mg/kg/day), starting from 10.5 months old. The object recognition ability was tested at 2, 4, and 6 months of exposure, and spatial learning and memory were assessed at endpoint. The apoptosis of neurons and astrocytes in the cortex and hippocampus was analyzed, as well as the potential apoptotic signaling pathways. Our results showed that exposure to PFOS for 6 months caused a decrease in object recognition ability and a decline in learning and spatial memory. PFOS selectively increased apoptosis in neurons of the cerebral cortex and specifically activated the endoplasmic reticulum stress PERK/CHOP signaling pathway. In conclusion, our results confirmed that subchronic exposure to PFOS can lead to cognitive impairment in mice, which might be closely associated with the specific activation of an endoplasmic reticulum stress-induced pro-apoptosis pathway in the cerebral cortex neurons.
Collapse
Affiliation(s)
- Haijing Zhang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China
| | - Chao Zhang
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Donggang Xu
- Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Qin Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China
| | - Dongqun Xu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, 100021, China.
| |
Collapse
|
16
|
Zhu W, Li Y, Ma X, Yang H, Wang Z, Shi R, Shi W, Cong B. Bibliometric analysis of post-traumatic stress disorder in forensic medicine: Research trends, hot spots, and prospects. Front Psychol 2023; 13:1074999. [PMID: 36726521 PMCID: PMC9884826 DOI: 10.3389/fpsyg.2022.1074999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/22/2022] [Indexed: 01/17/2023] Open
Abstract
Background Post-traumatic stress disorder (PTSD) has various risk factors, complex pathogenesis, and diverse symptoms, and is often comorbid with other injuries and diseases, making forensic diagnosis difficult. Methods To explore the current research status and trends of PTSD, we used the Web of Science Core Collection databases to screen PTSD-related literature published between 2010 and 2021 and CiteSpace to perform bibliometric analysis. Results In recent years, PTSD-related research has grown steadily. The countries and institutions with the most research results were the United States and England, and King's College London and Boston University, respectively. Publications were identified from 2,821 different journals, including 13 forensic-related journals, but the journal distribution was relatively scattered and there was a lack of professional core journals. Keyword co-occurrence and clustering identified many hot topics; "rat model," "mental health," and "satisfaction" were the topics most likely to have a clear effect on future research. Analysis extracted nine turning points from the literature that suggested that neural network centers, the hypothalamic-pituitary-adrenal axis, and biomarkers were new research directions. It was found that COVID-19 can cause severe psychological stress and induce PTSD, but the relationship needs further study. The literature on stress response areas and biomarkers has gradually increased over time, but specific systemic neural brain circuits and biomarkers remain to be determined. Conclusion There is a need to expand the collection of different types of biological tissue samples from patients with different backgrounds, screen PTSD biomarkers and molecular targets using multi-omics and molecular biology techniques, and establish PTSD-related molecular networks. This may promote a systematic understanding of the abnormal activation of neural circuits in patients with PTSD and help to establish a personalized, accurate, and objective forensic diagnostic standard.
Collapse
|
17
|
Zhang N, Shentu Y, Zhu M, Wang H, Yin X, Du C, Xue F, Fan J, Gong Y, Fan X. Role of Ero1α in cognitive impairment induced by chronic hypoxia. Brain Res 2022; 1797:148117. [PMID: 36220374 DOI: 10.1016/j.brainres.2022.148117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 09/17/2022] [Accepted: 10/05/2022] [Indexed: 11/02/2022]
Abstract
Recent reports suggested the endoplasmic reticulum stress (ERS)-associated pathway is involved with cognitive impairment in hypoxia condition. ERO1-like protein alpha (Ero1α), an endoplasmic reticulum membrane-bound N-glycoprotein, has been reported to promote oxidative protein folding. However, no studies have reported whether the Ero1α is trapped in hypoxia-induced neuronal loss through the ERS-associated pathways. In our study, this effect of Ero1α was investigated using C57BL/6J mice, the HT22 cells and primary rat neurons. C57BL/6J mice were modeled in a hypoxic chamber for 4 weeks. Behavioral tests were then carried out to test cognitive functions, including the Morris water maze and fear conditioning test. Proteomics showed that Ero1α distinctly upregulated compared with normoxia group and verified using western blotting. Flow cytometry and immunofluorescence were used to analyze the neuroprotective effect of inhibitor EN460 of Ero1α in the HT22 cells. In C57BL/6J mice, hypoxia significantly caused cognitive decline. Brain slice staining results were also used to confirm this effect. Western blot analysis demonstrated that Ero1α, ERS-associated proteins and apoptosis-associated proteins significantly increased in the hypoxia treated groups, further proliferation-related marker protein decreased. EN460, a selective endoplasmic reticulum oxidation 1 (ERO1) inhibitor, counteracted neuronal apoptosis and ameliorated neuronal cell proliferation in the HT22 cells. Taken together, our data indicate that hypoxia induces cognitive impairment, at least in part, by upregulating Ero1α which contributes to neuronal apoptosis through ERS signaling pathway, providing preliminary experimental evidence that the Ero1α is a promising therapeutic target in hypoxia-induced cognitive deficits.
Collapse
Affiliation(s)
- Nan Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yangping Shentu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Zhu
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hui Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xianghong Yin
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Congkuo Du
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Feng Xue
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
18
|
Gao H, Lei X, Ye S, Ye T, Hua R, Wang G, Song H, Zhou P, Wang Y, Cai B. Genistein attenuates memory impairment in Alzheimer's disease via ERS-mediated apoptotic pathway in vivo and in vitro. J Nutr Biochem 2022; 109:109118. [PMID: 35933022 DOI: 10.1016/j.jnutbio.2022.109118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/04/2022] [Accepted: 06/24/2022] [Indexed: 02/06/2023]
Abstract
Genistein (GS), an isoflavone compound found in soybean, plays a neuroprotective role in Alzheimer's disease (AD). However, the mechanism of its action remains unclear. Herein, binding ability between GS and GRP78 was predicted by molecular docking, and the effect of GS in vivo and vitro were further studied. In this study, the effects of GS on learning and memory ability, changes of hippocampal neurons and ultrastructure of hippocampal CA3 region in AD rats were investigated. Besides, the protein or mRNA levels of the related proteins were detected. The results showed GS could effectively improve the learning and the memory ability, reduce the damage of hippocampal neurons, and decrease the protein or mRNA expression levels of GRP78, CHOP, Caspase-12, Cle-Caspase-9, Cle-Caspase-3, PERK, and p-PERK. Taken together, our data reveal GS has a neuroprotective effect by inhibiting the ERS-mediated apoptotic pathway, which may be a new therapeutic target for the treatment of AD.
Collapse
Affiliation(s)
- Huawu Gao
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Xin Lei
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Shu Ye
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China
| | - Ting Ye
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Rupeng Hua
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Guoquan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Peng Zhou
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.
| | - Yan Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.
| | - Biao Cai
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China; Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, China; Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, China.
| |
Collapse
|
19
|
Fang X, Han Q, Li S, Luo A. Melatonin attenuates spatial learning and memory dysfunction in developing rats by suppressing isoflurane-induced endoplasmic reticulum stress via the SIRT1/Mfn2/PERK signaling pathway. Heliyon 2022; 8:e10326. [PMID: 36091956 PMCID: PMC9459431 DOI: 10.1016/j.heliyon.2022.e10326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/03/2022] [Accepted: 08/12/2022] [Indexed: 11/29/2022] Open
Abstract
Use of the inhalation anesthetic isoflurane may increase the risk of cognitive deficiency and neurotoxicity after birth. A growing body of evidence suggests that melatonin is an effective treatment for various types of oxidative stress damage and neurodegenerative disease. In this study, we aimed to examine the effects of melatonin on isoflurane-induced endoplasmic reticulum (ER) stress, spatial learning and memory impairment during development. The rats were grouped according to whether the rats were exposed to isoflurane or a control gas and whether they were administered melatonin or phosphate buffered saline (PBS). We administered isoflurane to 7-day-old Sprague-Dawley rat pups with intraperitoneal injections of melatonin (20 mg/kg) 15 min before and 3 h after the initiation of anesthesia. Twelve hours after isoflurane anesthesia, rats were randomly selected from each group and sacrificed. The hippocampal tissue and serum were collected to determine the levels of SIRT1, Mfn2, PERK, and other proteins or cytokines related to ER stress, apoptosis, and neuroinflammation. Subsequently, all remaining rats were assessed for spatial learning and memory deficiency 31 days after birth using the Morris water maze test. We found that melatonin attenuated isoflurane-induced ER stress and neuroapoptosis in the hippocampus and decreased the level of neuroinflammatory markers in the serum of newborn rats, resulting in improved spatial learning and memory. In addition, the neuroprotective effect of melatonin was weakened after the SIRT1/Mfn2/PERK signaling pathway was suppressed by lentivirus transfection. Therefore, our findings demonstrate that melatonin ameliorates spatial learning and memory impairment after isoflurane exposure, and these beneficial effects are associated with a reduction in ER stress, neuroapoptosis, and neuroinflammation via the SIRT1/Mfn2/PERK signaling pathway.
Collapse
Affiliation(s)
- Xi Fang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Qiang Han
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan 430030, Hubei, China
| |
Collapse
|
20
|
Topol IA, Polyakova IS, Elykova AV, Kamyshny AM. Peculiarities of Endoplasmic Reticulum Stress Regulator XBP1 Expression in the Gut-Associated Lymphoid Tissue of Wistar Rats under Chronic Stress. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s002209302205026x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
21
|
Upregulation of PGC-1 α Attenuates Oxygen-Glucose Deprivation-Induced Hippocampal Neuronal Injury. Neural Plast 2022; 2022:9682999. [PMID: 35719138 PMCID: PMC9203239 DOI: 10.1155/2022/9682999] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/25/2022] [Indexed: 11/30/2022] Open
Abstract
Hippocampal neuronal damage likely underlies cognitive impairment in vascular dementia (VaD). PPARγ coactivator-1α (PGC-1α) is a master regulator of mitochondrial biogenesis. However, the role and the precise mechanism of how PGC-1α alleviates hippocampal neuronal injury remain unknown. To address this question, HT-22 cells, an immortalized hippocampal neuron cell line, with or without PGC-1α overexpression were subjected to oxygen-glucose deprivation (OGD), which mimics the circumstance of chronic cerebral hypoperfusion in VaD. After OGD, cell viability was assessed using the MTS assay. The mitochondrial function and reactive oxygen species (ROS) were both detected. ChIP-Seq analysis was employed to discover the underlying molecular mechanism of PGC-1α-mediated neuroprotective effects. Our results showed that mitochondrial membrane potentials were increased and ROS production was decreased in PGC-1α overexpressing cells, which increased cell viability. The further bioinformatics analysis from ChIP-Seq data indicated that PGC-1α may participate in the regulation of apoptosis, autophagy, and mitophagy pathways in HT-22 cells. We found that PGC-1α promoted the LC3-II formation and reduced the neuronal apoptosis determined by TUNEL staining. In addition, PGC-1α upregulated the expressions of mitochondrial antioxidants, including SOD2, Trx2, and Prx3. In summary, our findings indicate that PGC-1α may attenuate OGD-induced hippocampal neuronal damage by regulating multiple mechanisms, like autophagy and mitochondrial function. Thus, PGC-1α may be a potential therapeutic target for hippocampal damage associated with cognitive impairment.
Collapse
|
22
|
Neurobiological Links between Stress, Brain Injury, and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8111022. [PMID: 35663199 PMCID: PMC9159819 DOI: 10.1155/2022/8111022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 12/13/2022]
Abstract
Stress, which refers to a combination of physiological, neuroendocrine, behavioral, and emotional responses to novel or threatening stimuli, is essentially a defensive adaptation under physiological conditions. However, strong and long-lasting stress can lead to psychological and pathological damage. Growing evidence suggests that patients suffering from mild and moderate brain injuries and diseases often show severe neurological dysfunction and experience severe and persistent stressful events or environmental stimuli, whether in the acute, subacute, or recovery stage. Previous studies have shown that stress has a remarkable influence on key brain regions and brain diseases. The mechanisms through which stress affects the brain are diverse, including activation of endoplasmic reticulum stress (ERS), apoptosis, oxidative stress, and excitatory/inhibitory neuron imbalance, and may lead to behavioral and cognitive deficits. The impact of stress on brain diseases is complex and involves impediment of recovery, aggravation of cognitive impairment, and neurodegeneration. This review summarizes various stress models and their applications and then discusses the effects and mechanisms of stress on key brain regions—including the hippocampus, hypothalamus, amygdala, and prefrontal cortex—and in brain injuries and diseases—including Alzheimer’s disease, stroke, traumatic brain injury, and epilepsy. Lastly, this review highlights psychological interventions and potential therapeutic targets for patients with brain injuries and diseases who experience severe and persistent stressful events.
Collapse
|
23
|
The Importance of Endoplasmic Reticulum Stress as a Novel Antidepressant Drug Target and Its Potential Impact on CNS Disorders. Pharmaceutics 2022; 14:pharmaceutics14040846. [PMID: 35456680 PMCID: PMC9032101 DOI: 10.3390/pharmaceutics14040846] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 04/07/2022] [Accepted: 04/10/2022] [Indexed: 11/24/2022] Open
Abstract
Many central nervous system (CNS) diseases, including major depressive disorder (MDD), are underpinned by the unfolded protein response (UPR) activated under endoplasmic reticulum (ER) stress. New, more efficient, therapeutic options for MDD are needed to avoid adverse effects and drug resistance. Therefore, the aim of the work was to determine whether UPR signalling pathway activation in astrocytes may serve as a novel target for antidepressant drugs. Among the tested antidepressants (escitalopram, amitriptyline, S-ketamine and R-ketamine), only S-ketamine, and to a lesser extent R-ketamine, induced the expression of most ER stress-responsive genes in astrocytes. Furthermore, cell viability and apoptosis measuring assays showed that (R-)S-ketamine did not affect cell survival under ER stress. Under normal conditions, S-ketamine played the key role in increasing the release of brain-derived neurotrophic factor (BDNF), indicating that the drug has a complex mechanism of action in astrocytes, which may contribute to its therapeutic effects. Our findings are the first to shed light on the relationship between old astrocyte specifically induced substance (OASIS) stabilized by ER stress and (R-)S-ketamine; however, the possible involvement of OASIS in the mechanism of therapeutic ketamine action requires further study.
Collapse
|
24
|
Acute stress and alcohol exposure during adolescence result in an anxious phenotype in adulthood: Role of altered glutamate/endocannabinoid transmission mechanisms. Prog Neuropsychopharmacol Biol Psychiatry 2022; 113:110460. [PMID: 34695542 DOI: 10.1016/j.pnpbp.2021.110460] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 12/31/2022]
Abstract
BACKGROUND Stressful episodes and high alcohol consumption during adolescence are considered major risk factors for the development of psychiatric disorders in adulthood. Identification of mechanisms underlying these early events, which enhanced vulnerability to mental illness, is essential for both their prevention and treatment. METHODS Male Wistar rats were used to investigate the long-term effects of early restraint stress and intermittent alcohol exposure (intragastric administration of 3 g/kg ethanol; 4 days/week for 4 weeks during adolescence) on anxiety-like behavior and the expression of signaling systems associated with emotional behaviors [e.g., corticosterone, fatty acid-derived molecules and endocannabinoid enzymes, glutamate receptor subunits, corticotropin releasing hormone receptors (CRHR1 and CRHR2) and neuropeptide Y receptors (NPY1R and NPYR2)] in the blood and amygdala. RESULTS Overall, both stress and alcohol exposure during adolescence induced anxiogenic-like behaviors, increased plasma levels of corticosterone and increases in the amygdalar expression of the cannabinoid CB2 receptor and certain subunits of glutamate receptors (i.e., mGluR1, mGluR5 and NMDAR1) in young adult rats. In addition, there were specific main effects of alcohol exposure on the expression of the cannabinoid CB1 receptor, monoacylglycerol lipase (MAGL) and NPY2R in the amygdala, and significant increases were observed in rats exposed to alcohol. Interestingly, there were significant interaction effects between restraint stress and alcohol exposure on the expression of plasma 2-arachidonoyl glycerol (2-AG), and both CRHR1,2 and NPY1R in the amygdala. Thus, the restraint stress was associated with increased 2-AG levels, which was not observed in rats exposed to alcohol. The alcohol exposure was associated with an increased expression of CRHR1,2 but the restraint stress prevented these increases (stress alcohol rats). In contrast, NPY1R was only increased in rats exposed to stress and alcohol. Finally, we did not observe any potentiation of the behavioral and molecular effects by the combination of stress and alcohol, which is concordant with an overall ceiling effect on some of the variables. CONCLUSION Separate and combined early stress and alcohol induced a common anxious phenotype with increased corticosterone in adulthood. However, there were differences in the amygdalar expression of signaling systems involved in maladaptive changes in emotional behavior. Therefore, our results suggest the existence of partially different mechanisms for stress and alcohol exposures.
Collapse
|
25
|
Abstract
Klotho gene was originally recognized as a putative aging-suppressor and its prominent age-regulating effects are mostly attributed to the modulation of mineral homeostasis in the kidney. However, recent studies link alterations in hippocampal Klotho expression with cognitive impairment and neurodegenerative diseases. This suggests that hippocampal neurons require Klotho for health and proper functionality. Klotho protects against neuronal dysfunction and regulates several intracellular signaling pathways including oxidative stress response, inflammation, DNA damage, autophagy, endoplasmic reticulum stress response, and multiple types of cell death. Specifically, this chapter covers the current knowledge as to how Klotho protein affects the hippocampal neuronal cells, with special attention paid to underlying molecular mechanisms, and thus influences hippocampal development, hippocampal-dependent cognition, behavior, and motor skills as well as mediates neurodegenerative processes.
Collapse
Affiliation(s)
- Jennifer Mytych
- Department of Biotechnology, Institute of Biology and Biotechnology, Collegium Scientarium Naturalium, University of Rzeszow, Werynia, Poland.
| |
Collapse
|
26
|
Gao C, Chen X, Xu H, Guo H, Zheng L, Yan Y, Ren Z, Luo C, Gao Y, Wang Z, Tao L, Wang T. Restraint Stress Delays the Recovery of Neurological Impairments and Exacerbates Brain Damages through Activating Endoplasmic Reticulum Stress-mediated Neurodegeneration/Autophagy/Apopotosis post Moderate Traumatic Brain Injury. Mol Neurobiol 2022; 59:1560-1576. [PMID: 35001355 DOI: 10.1007/s12035-022-02735-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 12/30/2021] [Indexed: 11/26/2022]
Abstract
Based on accumulating evidence, patients recovering from mild and moderate traumatic brain injury (TBI) often experience increased sensitivity to stressful events. However, few studies have assessed on the effects and pathophysiological mechanisms of stress on TBI. In the current study, using a mouse model of moderate TBI, we investigated whether restraint stress (RS) regulates secondary neurodegeneration and neuronal cell death, which are commonly associated with neurological dysfunctions. Our data showed that RS significantly reduced body weight recovery, delayed the recovery of neurological functions (motor function, cognitive function and anxiety-like behavior) and exacerbated the brain lesion volume after moderate TBI. Immunofluorescence results indicated that moderate TBI-induced cell insults and blood-brain barrier leakage were aggravated by RS. Further Western blotting experiments showed that RS activated endoplasmic reticulum (ER) stress excessively after moderate TBI and decreased the number of NeuN-positive cells, but increased the number of CHOP/NeuN-co-positive cells by performing immunostaining in the injured cortex after moderate TBI. Moreover, RS increased the ratios of CHOP/Aβ and CHOP/p-Tau co-positive cells in the injured cortex after moderate TBI. However, blocking ER stress with the classic ER stress inhibitor salubrinal remarkably decreased apoptosis and the levels of autophagy-related proteins in the mouse model of moderate TBI plus RS. Collectively, RS delays the recovery of neurological function and deteriorates morphological damage by excessively activating ER stress-mediated neurodegeneration, apoptosis and autophagy after moderate TBI. Thus, monitoring stress levels in patients recovering from non-severe TBI may merit consideration in the future.
Collapse
Affiliation(s)
- Cheng Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Xueshi Chen
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Heng Xu
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Hanmu Guo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Lexin Zheng
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Ya'nan Yan
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Zhiyang Ren
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Chengliang Luo
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Yuan Gao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Zufeng Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China
| | - Luyang Tao
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China.
| | - Tao Wang
- Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Affiliated Guangji Hospital, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
27
|
Cellular Response to Unfolded Proteins in Depression. Life (Basel) 2021; 11:life11121376. [PMID: 34947907 PMCID: PMC8707777 DOI: 10.3390/life11121376] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/05/2021] [Accepted: 12/07/2021] [Indexed: 02/07/2023] Open
Abstract
Despite many scientific studies on depression, there is no clear conception explaining the causes and mechanisms of depression development. Research conducted in recent years has shown that there is a strong relationship between depression and the endoplasmic reticulum (ER) stress. In order to restore ER homeostasis, the adaptive unfolded protein response (UPR) mechanism is activated. Research suggests that ER stress response pathways are continuously activated in patients with major depressive disorders (MDD). Therefore, it seems that the recommended drugs should reduce ER stress. A search is currently underway for drugs that will be both effective in reducing ER stress and relieving symptoms of depression.
Collapse
|
28
|
Zhang P, Yu Y, Wang P, Shen H, Ling X, Xue X, Yang Q, Zhang Y, Xiao J, Wang Z. Role of Hydrogen Sulfide in Myocardial Ischemia-Reperfusion Injury. J Cardiovasc Pharmacol 2021; 77:130-141. [PMID: 33165141 DOI: 10.1097/fjc.0000000000000943] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 10/13/2020] [Indexed: 12/28/2022]
Abstract
ABSTRACT Hydrogen sulfide (H2S), generally known as a new gas signal molecule after nitric oxide and carbon monoxide, has been found as an important endogenous gasotransmitter in the last few decades, and it plays a significant role in the cardiovascular system both pathologically and physiologically. In recent years, there is growing evidence that H2S provides myocardial protection against myocardial ischemia-reperfusion injury (MIRI), which resulted in an ongoing focus on the possible mechanisms of action accounting for the H2S cardioprotective effect. At present, lots of mechanisms of action have been verified through in vitro and in vivo models of I/R injury, such as S-sulfhydrated modification, antiapoptosis, effects on microRNA, bidirectional effect on autophagy, antioxidant stress, or interaction with NO and CO. With advances in understanding of the molecular pathogenesis of MIRI and pharmacology studies, the design, the development, and the pharmacological characterization of H2S donor drugs have made great important progress. This review summarizes the latest research progress on the role of H2S in MIRI, systematically explains the molecular mechanism of H2S affecting MIRI, and provides a new idea for the formulation of a myocardial protection strategy in the future.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Yue Yu
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Pei Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Hua Shen
- Department of Cardiovascular Surgery, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Ling
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Xiaofei Xue
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Qian Yang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Yufeng Zhang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Jian Xiao
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| | - Zhinong Wang
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China; and
| |
Collapse
|
29
|
Melatonin ameliorated CUMS-induced depression-like behavior via restoring endoplasmic reticulum stress in rat hippocampus. Neuroreport 2020; 32:8-15. [PMID: 33165196 DOI: 10.1097/wnr.0000000000001554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Melatonin is a hormone synthesized and secreted by the pineal gland with the effect of regulating sleep rhythm. Circadian and sleep disturbances may be central for understanding the pathophysiology and treatment of depression. Recently, the melatonergic system has been implicated in the pathophysiology and treatment of depression. In this study, we observed the effects of melatonin on depression-like behavior induced by chronic unpredictable mild stress (CUMS) in rats, and its molecular mechanism was explored. Adult male Sprague-Dawley rats were exposed to CUMS for 4 weeks. Melatonin or saline was injected intraperitoneally. Behavioral changes of Sprague-Dawley rats were detected by the open field test, sugar preference test, elevated O maze test and forced swimming test. In addition, the plasma corticosterone level and the expression of endoplasmic reticulum stress-related protein in the hippocampus of rats were measured. Compared with the control group, the CUMS-exposed Sprague-Dawley rats showed depression-like behavior, which was significantly improved by melatonin treatment. Moreover, CUMS induced endoplasmic reticulum stress and JNK phosphorylation in the hippocampus. Melatonin treatment could significantly reverse the endoplasmic reticulum stress and JNK phosphorylation induced by CUMS. These results suggest that melatonin improves depression-like behavior by inhibiting endoplasmic reticulum stress induced by CUMS. This study demonstrates that melatonin can improve depression-like behavior induced by CUMS, which may be related to the inhibition of endoplasmic reticulum stress and JNK phosphorylation in rat hippocampus.
Collapse
|
30
|
K V A, Madhana RM, Bais AK, Singh VB, Malik A, Sinha S, Lahkar M, Kumar P, Samudrala PK. Cognitive Improvement by Vorinostat through Modulation of Endoplasmic Reticulum Stress in a Corticosterone-Induced Chronic Stress Model in Mice. ACS Chem Neurosci 2020; 11:2649-2657. [PMID: 32673474 DOI: 10.1021/acschemneuro.0c00315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic stress is the leading cause of memory impairment today. Various stress-based models are being developed for studying cognitive impairment. Repurposing of existing drugs in a new pharmacology class is the safest and cheapest option for treatment instead of new drug discovery. Vorinostat (VOR) is the first histone deacetylase (HDAC) inhibitor approved for the treatment of cutaneous T-cell lymphoma by the U.S. FDA. VOR follows the rule of five and is reported to cross the blood-brain barrier. Therefore, we aimed to evaluate the procognitive potential of VOR (25 mg/kg) administered by intraperitoneal (ip) route in a stress-based model of chronic corticosterone (CORT) injections (20 mg/kg, subcutaneously (sc)). The study comprised six groups. Normal mice were administered vehicle (VEH) (days 1-21, sc) in the first group, VOR (days 8-21, 25 mg/kg, ip) in the second group, and fluoxetine (FLX) (days 8-21, 15 mg/kg, oral) in the third group. Mice in the remaining three groups were given 20 mg/kg (sc) CORT for 21 days, and VOR (days 8-21, 25 mg/kg, ip) or FLX (days 8-21, 15 mg/kg, oral) was additionally administered to the treatment groups. Behavioral tests such as Morris water maze test, novel object recognition test, and object in place test were performed at the end of the dosing schedule to assess cognition. After behavior tests, mice were sacrificed, and hippocampus was separated from brain tissue for reverse transcriptase polymerase chain reaction (RT-PCR), Western blot, and immunohistochemistry studies. VOR treatment attenuated endoplasmic reticulum (ER) stress in CORT mice as evident from the reduction in DNA damage-inducible transcript 3 (Ddit3) (gene encoding CHOP), caspase 12 (Casp12), and calpain-2 (Capn2) mRNA levels, and cleaved caspase 3 (CASP3) protein expression. Bax inhibitor-1 (BI-1) was significantly increased in VOR-treated CORT mice. VOR also reversed CORT induced increase in HDAC2 level in the CA3 region. The protective effects of VOR were comparable to that of FLX in CORT mice. Thus, VOR has the potential to reverse cognitive dysfunction via modulation of ER stress markers and HDAC2.
Collapse
Affiliation(s)
- Athira K V
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Mirza, Kamrup, 781125 Assam, India
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, 682041 Kerala, India
| | - Rajaram Mohanrao Madhana
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Mirza, Kamrup, 781125 Assam, India
| | - Akhilesh Kumar Bais
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Mirza, Kamrup, 781125 Assam, India
| | - Vijay Bahadur Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Mirza, Kamrup, 781125 Assam, India
| | - Arpit Malik
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Mirza, Kamrup, 781125 Assam, India
| | - Swapnil Sinha
- DST WOS-A Scientist, Department of Biotechnology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Mirza, Kamrup, 781125 Assam, India
| | - Mangala Lahkar
- Department of Pharmacology, Gauhati Medical College, Guwahati, 781032 Assam, India
| | - Pramod Kumar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, 781125 Assam, India
| | - Pavan Kumar Samudrala
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Guwahati, Mirza, Kamrup, 781125 Assam, India
| |
Collapse
|
31
|
Gao C, Meng Y, Chen G, Chen W, Chen XS, Luo CL, Zhang MY, Wang ZF, Wang T, Tao LY. Chronic restraint stress exacerbates neurological deficits and disrupts the remodeling of the neurovascular unit in a mouse intracerebral hemorrhage model. Stress 2020; 23:338-348. [PMID: 31591949 DOI: 10.1080/10253890.2019.1678023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Growing evidences have shown that patients recovering from stroke experience high and unremitting stress. Chronic restraint stress (CRS) has been found to exacerbate neurological impairments in an experimental focal cortical ischemia model. However, there have been no studies reporting the effect and mechanism of CRS on intracerebral hemorrhage (ICH). This study aimed to evaluate the effect of CRS on a mouse ICH model. Adult male C57BL mice were subjected to infusion of collagenase IV (to induce ICH) or saline (for sham) into the left striatum. After ICH, animals were stressed with application of CRS protocol for 21 days. Our results showed that CRS significantly exacerbated neurological deficits (Garcia test, corner turn test, and wire grip test) and the ipsilateral brain atrophy and reduced body weight gain after ICH. Immunofluorescence staining indicated that CRS exerted significant suppressive effects on neuron, astrocyte, vascular endothelial cell and pericyte and excessively activated microglia post ICH. All of the key cellular components mentioned above are involved in the neurovascular unit (NVU) remodeling in the peri-hemorrhagic region after ICH. Western blot results showed that matrix metalloproteinase (MMP)-9 and tight junction (TJ) proteins including zonula occludens-1, occludin and claudin-5 were increased after ICH, but MMP-9 protein was further up-regulated and TJ-related proteins were down-regulated by CRS. In addition, ICH-induced activation of endoplasmic reticulum stress and apoptosis were further strengthened by CRS. Collectively, CRS exacerbates neurological deficits and disrupts the remodeling of the peri-hemorrhagic NVU after ICH, which may be associated with TJ proteins degradation and excessive activation of MMP-9 and endoplasmic reticulum stress-apoptosis.LAY SUMMARYCRS exacerbates neurological deficits and disrupts the remodeling of the NVU in the recovery stage after ICH, which suggest that monitoring chronic stress levels in patients recovering from ICH may merit consideration in the future.
Collapse
Affiliation(s)
- Cheng Gao
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai, China
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Ying Meng
- Community Health Center, Suzhou Western Eco-City, Suzhou, China
| | - Guang Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Wei Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Xue-Shi Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Cheng-Liang Luo
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Ming-Yang Zhang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Zu-Feng Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| | - Tao Wang
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Science, Shanghai, China
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
- School of Pharmacy, Soochow University, Suzhou, China
| | - Lu-Yang Tao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou, China
| |
Collapse
|
32
|
Amin SN, Hassan SS, Khashaba AS, Youakim MF, Latif NSA, Rashed LA, Yassa HD. Hippocampal and Cerebellar Changes in Acute Restraint Stress and the Impact of Pretreatment with Ceftriaxone. Brain Sci 2020; 10:E193. [PMID: 32218213 PMCID: PMC7225952 DOI: 10.3390/brainsci10040193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/15/2020] [Accepted: 03/22/2020] [Indexed: 12/12/2022] Open
Abstract
Acute restraint stress (ARS) is an unavoidable stress situation and may be encountered in different clinical situations. The aim of the current study was to investigate the effects of ARS on the hippocampus and cerebellum, assess the impact of these effects on the behavior and cognitive function, and determine whether pretreatment with ceftriaxone would attenuate the damages produced by ARS on the hippocampus and cerebellum. Four groups of male mice were included in this study: The control group, ARS group, ceftriaxone group, and ARS + ceftriaxone group. Tail suspension test, Y-maze task, and open field tests were used to assess depression, working spatial memory, and anxiety. The biochemical analyses included measurements of serum cortisol, tumor necrotic factor (TNF), interleukin-6, hippocampal expression of bone morphogenetic protein 9 (BMP9), lysosomal-associated membrane protein 1 (LAMP1), glutamate transporter 1 (GLT1), heat shock protein 90, cerebellar expression of S100 protein, glutamic acid decarboxylase (GAD), and carbon anhydrase. Histopathological examination of the brain sections was conducted on the hippocampus and cerebellum by hematoxylin and eosin stains in addition to ultrastructure evaluation using electron microscopy. Our results suggested that ceftriaxone had neuroprotective properties by attenuating the effects of ARS on the hippocampus and cerebellum in mice. This effect was demonstrated by the improvement in the cognitive and behavioral tests as well as by the preservation of the hippocampal and cerebellar architecture.
Collapse
Affiliation(s)
- Shaimaa N. Amin
- Department of Basic Medical Sciences, Faculty of Medicine, Hashemite University, Zarqa 13133, Jordan
- Department of Medical Physiology, Faculty of Medicine, Cairo University, Cairo 11451, Egypt
| | - Sherif S. Hassan
- Department of Medical Education, School of Medicine, California University of Science & Medicine, San Bernardino, CA 82408, USA
- Department of Anatomy, Faculty of Medicine, Cairo University, Cairo 11451, Egypt;
| | - Ahmed S. Khashaba
- Department of Basic Sciences, Riyadh Elm University, Riyadh 12734, Saudi Arabia;
| | - Magdy F. Youakim
- Department of Anatomy, Faculty of Medicine, Cairo University, Cairo 11451, Egypt;
| | - Noha S. Abdel Latif
- Department of Medical Pharmacology, Faculty of Medicine, Cairo University Cairo 11451, Egypt;
| | - Laila A. Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo 11451, Egypt;
| | - Hanan D. Yassa
- Department of Anatomy and Embryology, Faculty of Medicine, Beni-Suef University, Beni Suef 62511, Egypt;
| |
Collapse
|
33
|
MANF Ablation Causes Prolonged Activation of the UPR without Neurodegeneration in the Mouse Midbrain Dopamine System. eNeuro 2020; 7:ENEURO.0477-19.2019. [PMID: 32005751 PMCID: PMC7053174 DOI: 10.1523/eneuro.0477-19.2019] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 01/08/2023] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER) localized protein that regulates ER homeostasis and unfolded protein response (UPR). The biology of endogenous MANF in the mammalian brain is unknown and therefore we studied the brain phenotype of MANF-deficient female and male mice at different ages focusing on the midbrain dopamine system and cortical neurons. We show that a lack of MANF from the brain led to the chronic activation of UPR by upregulation of the endoribonuclease activity of the inositol-requiring enzyme 1α (IRE1α) pathway. Furthermore, in the aged MANF-deficient mouse brain in addition the protein kinase-like ER kinase (PERK) and activating transcription factor 6 (ATF6) branches of the UPR pathways were activated. Neuronal loss in neurodegenerative diseases has been associated with chronic ER stress. In our mouse model, increased UPR activation did not lead to neuronal cell loss in the substantia nigra (SN), decrease of striatal dopamine or behavioral changes of MANF-deficient mice. However, cortical neurons lacking MANF were more vulnerable to chemical induction of additional ER stress in vitro. We conclude that embryonic neuronal deletion of MANF does not cause the loss of midbrain dopamine neurons in mice. However, endogenous MANF is needed for maintenance of neuronal ER homeostasis both in vivo and in vitro.
Collapse
|
34
|
Yoshino Y, Dwivedi Y. Elevated expression of unfolded protein response genes in the prefrontal cortex of depressed subjects: Effect of suicide. J Affect Disord 2020; 262:229-236. [PMID: 31727394 PMCID: PMC6917852 DOI: 10.1016/j.jad.2019.11.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/01/2019] [Accepted: 11/02/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Major Depressive Disorder (MDD) is a leading cause of mental disability worldwide. Despite many studies, the pathophysiology associated with MDD brain is not very clear. It is reported that cellular stress is related to depressive symptoms. Under stressful conditions, intracellular homeostasis processes can be disrupted, which can induce a process of unfolded protein response (UPR) in the subcellular lumen of endoplasmic reticulum (ER). The purpose of this study is to elucidate whether UPR is active in the depressed brain. METHODS The dorsolateral prefrontal cortex (dlPFC) was used from 23 non-psychiatric controls and 43 MDD subjects. The expression levels of UPR associated genes (GRP78, GRP94, XBP-1, CHOP, ATF4C, and ATF6C) were measured by qRT-PCR. RESULTS The level of mRNA expression in MDD subjects was significantly higher for GRP78 (p = 0.008), GRP94 (p = 0.018), and ATF4C (p = 0.03) compared to non-psychiatric controls. Further analysis suggested that changes in the expression of these genes were specifically higher only in those MDD subjects who died by suicide but not in those who died by causes other than suicide when compared with non-psychiatric controls (GRP78, p = 0.007; GRP94, p = 0.041; ATF4C, p = 0.037). LIMITATIONS This study was performed only in MDD subjects who had died by suicide. Suicide subjects with other psychiatric illnesses need to be included. CONCLUSIONS Given that UPR is involved in many physiological processes in the brain, including inflammatory response as well as apoptosis, increased expression of UPR genes indicates that ER stress and mediated UPR may be critical factors in suicidality among depressed patients.
Collapse
Affiliation(s)
- Yuta Yoshino
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294, United States.
| |
Collapse
|
35
|
Effects of Limonene on Chronic Restraint Stress-Induced Memory Impairment and Anxiety in Male Rats. NEUROPHYSIOLOGY+ 2019. [DOI: 10.1007/s11062-019-09800-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
36
|
Timberlake Ii M, Roy B, Dwivedi Y. A Novel Animal Model for Studying Depression Featuring the Induction of the Unfolded Protein Response in Hippocampus. Mol Neurobiol 2019; 56:8524-8536. [PMID: 31267370 DOI: 10.1007/s12035-019-01687-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/21/2019] [Indexed: 02/08/2023]
Abstract
Depression is the leading cause of disability worldwide with global distribution of 322 million people suffering from the disease. While much is understood about depression, the underlying pathophysiology is yet to be fully characterized. Recently, the unfolded protein response (UPR) has been shown to be involved in regulating key aspects like inflammation, cell death, and behavioral depression. The UPR is an evolutionarily conserved ancient response system that reacts to the stressful environmental impact on a cell; the net effect of stress to a cell is that the quality of protein folding is diminished. The UPR responds by repairing and removing misfolded proteins and, if necessary, initiates apoptosis. Here, we demonstrate that the UPR is not only involved in depression, but that its activation causes a depressive phenotype. The hippocampi of rats were directly infused with 500 ng of tunicamycin (TM), an agent that initiates the UPR by blocking N-terminal glycosylation. Three to 8 days post-surgery, the rats showed depressive behavior in escape latency, forced swim despair, sucrose preference anhedonia, and also physiological signs of depression like decreased weight. Further, these behavioral changes were associated with enhanced expression of key UPR genes and proteins in the hippocampus. We propose that this model will make an excellent tool for studying depression and for understanding pathways that are affected by the UPR which directly causes depressive behavior.
Collapse
Affiliation(s)
- Matthew Timberlake Ii
- Department of Psychiatry and Behavioral Neurobiology, SC711 Sparks Center, University of Alabama at Birmingham, 1720 7th Avenue South, Birmingham, AL, 35294, USA
| | - Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, SC711 Sparks Center, University of Alabama at Birmingham, 1720 7th Avenue South, Birmingham, AL, 35294, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, SC711 Sparks Center, University of Alabama at Birmingham, 1720 7th Avenue South, Birmingham, AL, 35294, USA.
| |
Collapse
|
37
|
Restraint stress exacerbates cell degeneration induced by acute binge ethanol in the adolescent, but not in the adult or middle-aged, brain. Behav Brain Res 2019; 364:317-327. [PMID: 30797854 DOI: 10.1016/j.bbr.2019.02.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/12/2019] [Accepted: 02/20/2019] [Indexed: 12/22/2022]
Abstract
Restraint stress (RS) induces neurotoxicity in the hippocampus, yet most of the studies have employed protracted RS (i.e., ≈ 21 days). Binge ethanol can induce brain toxicity, an effect affected by age. It could be postulated that RS may facilitate ethanol-induced neurotoxicity, perhaps to a greater extent in adolescent vs. older subjects. We analyzed whether adolescent, adult or middle-aged male rats exposed to five episodes of RS followed, 72h later, by binge ethanol (i.e., two administrations of 2.5 g/kg ethanol) exhibited hippocampal neurotoxicity. Adolescents, but not adult or middle-aged rats, exhibited sensitivity to the neurotoxic effects of ethanol at dorsal CA2, ventral CA3 and ventral DG, and a neurotoxic effect of stress at dorsal CA1. Moreover, the combination of ethanol and stress exerted a synergistic effect upon cell degeneration at ventral CA1 and CA2, which was restricted to adolescents. Ethanol also increased cell degeneration, irrespective of age or stress, in dorsal CA3 and in dorsal DG; and ethanol and stress had, across all ages, a synergistic effect upon cell degeneration at the dorsal CA1. The greater neurotoxic response of adolescents to ethanol, stress, or ethanol+stress can put them at risk for the development of alcohol problems.
Collapse
|
38
|
Yao M, Zhao Z, Wei L, Zhou D, Xue Z, Ge S. HSF1/HSP pathway in the hippocampus is involved in SIRT1-mediated caloric restriction-induced neuroprotection after surgery in aged mice. Exp Gerontol 2019; 119:184-192. [PMID: 30772489 DOI: 10.1016/j.exger.2019.02.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 02/09/2019] [Accepted: 02/12/2019] [Indexed: 10/27/2022]
Abstract
Postoperative cognitive dysfunction is common in the elderly. Endoplasmic reticulum stress (ER-stress) increases neuronal apoptosis after surgery, and chaperone molecules, such as heat shock proteins (HSPs), help reduce unfolded protein reactions, thereby promoting protein homeostasis. Mammal sirtuin1 (SIRT1)-mediated deacetylation of heat shock factor 1 (HSF1) upregulates HSF1 binding to the HSP70 promoter. Caloric restriction (CR) improves cognition in many neurodegenerative models. In this study, we evaluated whether CR improves impaired learning and memory after surgery by attenuating ER-stress in an SIRT1-dependent manner. Male 18-month-old C57BL/6J mice receiving a 12-week CR or an ad libitum (AL) diet pre-intervention were challenged with tibial open fracture surgery and anesthesia or no treatment. We found a significant protective effect of CR on memory in contextual fear conditioning test after surgery compared with the AL group. CR alleviated ER-stress and neuronal apoptosis in the hippocampus induced by surgery. CR increased HSP70 expression through the HSF1/HSP pathway in a SIRT1-mediated manner, and inhibition of SIRT1 in the hippocampus by lentivirus injection partially reduced the benefits of CR (increased HSP70, deacetylated HSF1, reduced ER-stress, and improved memory). Taken together, our results showed that CR alleviates memory impairment postoperatively via attenuation of ER-stress in the hippocampus in an SIRT1-dependent manner, and the SIRT1/HSF1/HSP70 pathway is involved in this process.
Collapse
Affiliation(s)
- Minmin Yao
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Zhimeng Zhao
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lan Wei
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Di Zhou
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Zhanggang Xue
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Shengjin Ge
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
39
|
Linking unfolded protein response to inflammation and depression: potential pathologic and therapeutic implications. Mol Psychiatry 2019; 24:987-994. [PMID: 30214045 PMCID: PMC6416085 DOI: 10.1038/s41380-018-0241-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/26/2018] [Accepted: 08/16/2018] [Indexed: 11/21/2022]
Abstract
Depression is a devastating mental disorder that affects millions of people worldwide. Inflammation has been shown to be a key factor involved in the underlying pathophysiology of depression and has been shown in a substantial proportion of cases of depression. Changes attributed with morphological deformities and immunomodulation in susceptible regions of the depressed brain raised the possibility of altered cellular homeostasis transduced by the intracellular stress response. How emotional stressors can lead to an inflamed brain that directly affects physiology and activity is yet to be fully understood. The unfolded protein response (UPR) has been shown to be active in both models of depression as well as in postmortem brain of depressed individuals. The UPR is the cellular response to stress which results in misfolded proteins. Interestingly, UPR activation is directly linked to both inflammatory cytokine production and Toll-like receptor (TLR) expression. The TLRs are part of the innate immune response which typically reacts to "classic invasions" such as bacteria or viruses as well as trauma. TLRs have also been shown to be upregulated in depression, thus solidifying the connection between inflammation and depression. In this review, we aim to tie the UPR-TLR response and depression, and describe the implications of such an association. We also propose future directions for their role in treatment for depression.
Collapse
|
40
|
Akimoto H, Oshima S, Michiyama Y, Negishi A, Nemoto T, Kobayashi D. Metabolic Profiling of the Hippocampus of Rats Experiencing Nicotine-Withdrawal Symptoms. Biol Pharm Bull 2018; 41:1879-1884. [DOI: 10.1248/bpb.b18-00486] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hayato Akimoto
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| | - Shinji Oshima
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| | - Yuichi Michiyama
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| | - Akio Negishi
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University
| | - Tadashi Nemoto
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST)
| | | |
Collapse
|
41
|
Wang B, Ge S, Xiong W, Xue Z. Effects of resveratrol pretreatment on endoplasmic reticulum stress and cognitive function after surgery in aged mice. BMC Anesthesiol 2018; 18:141. [PMID: 30305045 PMCID: PMC6180510 DOI: 10.1186/s12871-018-0606-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/01/2018] [Indexed: 12/12/2022] Open
Abstract
Background Postoperative cognitive dysfunction (POCD) seriously reduces quality of life and is associated with increased morbidity and mortality. The causes and neuropathogenesis of POCD remain largely unknown. Resveratrol, a sirtuin 1 (Sirt1) activator, is a polyphenol compound found in red wine that has protective functions in neuropathology paradigms. Endoplasmic reticulum stress (ERS) is a primary cellular response that activates the unfolded protein response (UPR). ERS and UPR mediate molecular and biochemical mechanisms related to neurodegeneration; however, the roles of ERS and Sirt1 in POCD remain unclear. The properties of resveratrol might be useful in the setting of POCD. Methods In the present study, we investigated learning and memory function and ERS pathways in aged mice after surgery under local anesthesia, and we evaluated the effects of resveratrol pretreatment. Results We found that resveratrol attenuated postoperative learning and memory impairment in aged mice postoperatively but did not alter locomotor activity. Resveratrol significantly decreased postoperative expression of ERS pathway UPR-related proteins and inflammatory mediators including nuclear factor-κB (NF-κB) in the hippocampus. This was accompanied by higher Sirt1 protein expression levels. Pretreatment with resveratrol did not affect the number of hippocampal neurons in aged mice after surgery. Conclusion Overall, resveratrol pretreatment attenuated short-term learning and memory impairment and the ERS pathway UPR in aged mice after surgery under local anesthesia.
Collapse
Affiliation(s)
- Bei Wang
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shengjin Ge
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Wanxia Xiong
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhanggang Xue
- Department of Anesthesia, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| |
Collapse
|
42
|
Inkster B, Simmons A, Cole J, Schoof E, Linding R, Nichols T, Muglia P, Holsboer F, Saemann P, McGuffin P, Fu C, Miskowiak K, Matthews PM, Zai G, Nicodemus K. Unravelling the GSK3β-related genotypic interaction network influencing hippocampal volume in recurrent major depressive disorder. Psychiatr Genet 2018; 28:77-84. [PMID: 30080747 PMCID: PMC6531290 DOI: 10.1097/ypg.0000000000000203] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Glycogen synthase kinase 3β (GSK3β) has been implicated in mood disorders. We previously reported associations between a GSK3β polymorphism and hippocampal volume in major depressive disorder (MDD). We then reported similar associations for a subset of GSK3β-regulated genes. We now investigate an algorithm-derived comprehensive list of genes encoding proteins that directly interact with GSK3β to identify a genotypic network influencing hippocampal volume in MDD. PARTICIPANTS AND METHODS We used discovery (N=141) and replication (N=77) recurrent MDD samples. Our gene list was generated from the NetworKIN database. Hippocampal measures were derived using an optimized Freesurfer protocol. We identified interacting single nucleotide polymorphisms using the machine learning algorithm Random Forest and verified interactions using likelihood ratio tests between nested linear regression models. RESULTS The discovery sample showed multiple two-single nucleotide polymorphism interactions with hippocampal volume. The replication sample showed a replicable interaction (likelihood ratio test: P=0.0088, replication sample; P=0.017, discovery sample; Stouffer's combined P=0.0007) between genes associated previously with endoplasmic reticulum stress, calcium regulation and histone modifications. CONCLUSION Our results provide genetic evidence supporting associations between hippocampal volume and MDD, which may reflect underlying cellular stress responses. Our study provides evidence of biological mechanisms that should be further explored in the search for disease-modifying therapeutic targets for depression.
Collapse
Affiliation(s)
- Becky Inkster
- Department of Psychiatry, University of Cambridge, UK
- Wolfson College, University of Cambridge, UK
- Cambridgeshire and Peterborough NHS Foundation Trust, UK
| | - Andy Simmons
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - James Cole
- The Computational, Cognitive & Clinical Neuroimaging Lab, Department of Medicine, Imperial College London, UK
| | - Erwin Schoof
- Biotech Research & Innovation Centre, University of Copenhagen
| | - Rune Linding
- Biotech Research & Innovation Centre, University of Copenhagen
| | - Tom Nichols
- Department of Statistics, Warwick University, UK
| | - Pierandrea Muglia
- Genetics Division, Drug Discovery, Medicine Development Centre, GlaxoSmithKline, R&D, Verona, Italy
| | | | | | - Peter McGuffin
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Cynthia Fu
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, UK
| | - Kamilla Miskowiak
- Department of Psychiatry, Psychiatric Centre Copenhagen, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Paul M Matthews
- Department of Medicine, Imperial College London and UK Dementia Research Institute
| | - Gwyneth Zai
- Neurogenetics Section, Molecular Brain Science Department, Campbell Family Mental Health Research Institute, and Mood & Anxiety Division, Centre for Addiction and Mental Health, Toronto, Canada
- Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Kristin Nicodemus
- Centre for Genomics and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, UK
| |
Collapse
|
43
|
Neuroprotective Effects of dl-3-n-Butylphthalide against Doxorubicin-Induced Neuroinflammation, Oxidative Stress, Endoplasmic Reticulum Stress, and Behavioral Changes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9125601. [PMID: 30186550 PMCID: PMC6116408 DOI: 10.1155/2018/9125601] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 05/16/2018] [Accepted: 06/06/2018] [Indexed: 11/24/2022]
Abstract
Doxorubicin (DOX) is a broad-spectrum antitumor drug while its use is limited due to its neurobiological side effects associated with depression. We investigated the neuroprotective efficacy of dl-3-n-butylphthalide (dl-NBP) against DOX-induced anxiety- and depression-like behaviors in rats. dl-NBP was given (30 mg/kg) daily by gavage over three weeks starting seven days before DOX administration. Elevated plus maze (EPM) test, forced swimming test (FST), and sucrose preference test (SPT) were performed to assess anxiety- and depression-like behaviors. Our study showed that the supplementation of dl-NBP significantly mitigated the behavioral changes induced by DOX. To further explore the mechanism of neuroprotection induced by dl-NBP, several biomarkers including oxidative stress markers, endoplasmic reticulum (ER) stress markers, and neuroinflammatory cytokines in the hippocampus were quantified. The results showed that dl-NBP treatment alleviated DOX-induced neural apoptosis. Meanwhile, DOX-induced oxidative stress and ER stress in the hippocampus were significantly ameliorated in dl-NBP pretreatment group. Our study found that dl-NBP alleviated the upregulation of malondialdehyde (MDA), nitric oxide (NO), CHOP, glucose-regulated protein 78 kD (GRP-78), and caspase-12 and increased the levels of reduced glutathione (GSH) and activities of catalase (CAT), glutathione reductase (GR), and glutathione peroxidase (GPx) in the hippocampus of rats exposed to DOX. Additionally, the gene expression of interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-alpha (TNF-α) and protein levels of inducible nitric oxide synthase (iNOS) were significantly increased in DOX-treated group, whereas DOX-induced neuroinflammation was significantly attenuated in dl-NBP supplementation group. In conclusion, dl-NBP could alleviate DOX-induced anxiety- and depression-like behaviors via attenuating oxidative stress, ER stress, inflammatory reaction, and neural apoptosis, providing a basis as a therapeutic potential against DOX-induced neurotoxicity.
Collapse
|
44
|
Timberlake M, Prall K, Roy B, Dwivedi Y. Unfolded protein response and associated alterations in toll-like receptor expression and interaction in the hippocampus of restraint rats. Psychoneuroendocrinology 2018; 89:185-193. [PMID: 29414031 PMCID: PMC5878717 DOI: 10.1016/j.psyneuen.2018.01.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 01/08/2018] [Accepted: 01/18/2018] [Indexed: 12/12/2022]
Abstract
Recent evidence suggests that the cellular response to stress often elicits the unfolded protein response (UPR), which has an active role in major depression in emotionally relevant regions of the brain, such as the hippocampus. Much of the UPR activity has been found to be coalesced with the pro-inflammatory environment of the depressed brain. Specifically, downstream transcriptions of pro-inflammatory cytokines and increased regulation of candidate inflammatory mediators, such as toll-like receptors (TLRs), are promoted by the UPR. The present study examined the hippocampus associated expression profile of Tlr genes and their interaction with the UPR chaperone GRP94 in stress-induced rodent model of depression (restraint stress model). Also, the expression status of UPR related genes was evaluated in hippocampus using the same model. mRNA and protein levels of Tlr and UPR associated genes were examined by qRT-PCR and Western blot, respectively. Co-immunoprecipitation (Co-IP) method was used to determine the direct interaction between TLRs with GRP94 in depressed rat brain. The results showed that both UPR (Xbp-1, its spliced variant sXbp-1, Atf-6, Chop, and Grp94) and Tlr (2, 3, 4, 7 and 9) genes were significantly upregulated in the hippocampi of rats who were exposed to restraint stress. Similar upregulation was observed in the protein levels of the above-mentioned TLRs and the UPR chaperone protein GRP94 as well as total and phosphorylated forms of sensor proteins IRE1α and PERK. Further, a significantly increased interaction was observed between GRP94 and the activated TLR proteins. Since, increased inflammatory activity in vulnerable areas like hippocampus is coherently associated with depressed brain; our present data suggest that the UPR may be an integral part of increased activity of inflammatory regulations in depression.
Collapse
Affiliation(s)
- Matthew Timberlake
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Kevin Prall
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Bhaskar Roy
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, Alabama, 35294, USA
| | - Yogesh Dwivedi
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
45
|
Guo QH, Tong QH, Lu N, Cao H, Yang L, Zhang YQ. Proteomic Analysis of the Hippocampus in Mouse Models of Trigeminal Neuralgia and Inescapable Shock-Induced Depression. Neurosci Bull 2018; 34:74-84. [PMID: 28424990 PMCID: PMC5799118 DOI: 10.1007/s12264-017-0131-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/06/2017] [Indexed: 12/31/2022] Open
Abstract
To investigate the behavioral and biomolecular similarity between neuralgia and depression, a trigeminal neuralgia (TN) mouse model was established by constriction of the infraorbital nerve (CION) to mimic clinical trigeminal neuropathic pain. A mouse learned helplessness (LH) model was developed to investigate inescapable foot-shock-induced psychiatric disorders like depression in humans. Mass spectrometry was used to assess changes in the biomolecules and signaling pathways in the hippocampus from TN or LH mice. TN mice developed not only significant mechanical allodynia but also depressive-like behaviors (mainly behavioral despair) at 2 weeks after CION, similar to LH mice. MS analysis demonstrated common and distinctive protein changes in the hippocampus between groups. Many protein function families (such as cell-to-cell signaling and interaction, and cell assembly and organization,) and signaling pathways (e.g., the Huntington's disease pathway) were involved in chronic neuralgia and depression. Together, these results demonstrated that the LH and TN models both develop depressive-like behaviors, and revealed the involvement of many psychiatric disorder-related biomolecules/pathways in the pathogenesis of TN and LH.
Collapse
Affiliation(s)
- Qing-Huan Guo
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Qing-He Tong
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ning Lu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Hong Cao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| | - Liu Yang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China
| | - Yu-Qiu Zhang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
46
|
Tang YY, Wang AP, Wei HJ, Li MH, Zou W, Li X, Wang CY, Zhang P, Tang XQ. Role of silent information regulator 1 in the protective effect of hydrogen sulfide on homocysteine-induced cognitive dysfunction: Involving reduction of hippocampal ER stress. Behav Brain Res 2018; 342:35-42. [PMID: 29307666 DOI: 10.1016/j.bbr.2017.12.040] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 12/15/2017] [Accepted: 12/31/2017] [Indexed: 12/23/2022]
Abstract
Homocysteine (Hcy) causes cognitive deficits and hippocampal endoplasmic reticulum (ER) stress. Our previous study has confirmed that Hydrogen sulfide (H2S) attenuates Hcy-induced cognitive dysfunction and hippocampal ER stress. Silent information regulator 1 (Sirt-1) is indispensable in the formation of learning and memory. Therefore, the aim of this study was to explore the role of Sirt-1 in the protective effect of H2S against Hcy-induced cognitive dysfunction. We found that NaHS (a donor of H2S) markedly up-regulated the expression of Sirt-1 in the hippocampus of Hcy-exposed rats. Sirtinol, a specific inhibitor of Sirt-1, reversed the improving role of NaHS in the cognitive function of Hcy-exposed rats, as evidenced by that sirtinol increased the escape latency and the swim distance in the acquisition trial of morris water maze (MWM) test, decreased the times crossed through and the time spent in the target quadrant in the probe trail of MWM test, and reduced the discrimination index in the novel object recognition test (NORT) in the rats cotreated with NaHS and Hcy. We also found that sirtinol reversed the protection of NaHS against Hcy-induced hippocampal ER-stress, as evidenced by up-regulating the expressions of GRP78, CHOP, and cleaved caspase-12 in the hippocampus of rats cotreated with NaHS and Hcy. These results suggested the contribution of upregulation of hippocampal Sirt-1 to the improving role of H2S in the cognitive function of Hcy-exposed rats, which involves suppression of hippocampal ER stress. Our finding provides a new insight into the mechanism underlying the inhibitory role of H2S in Hcy-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Yi-Yun Tang
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Physiology, Medical College, University of South China, Hengyang, 421001, Hunan, PR China
| | - Ai-Ping Wang
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Anatomy, Medical College, University of South China, Hengyang, 421001, Hunan, PR China
| | - Hai-Jun Wei
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Physiology, Medical College, University of South China, Hengyang, 421001, Hunan, PR China
| | - Man-Hong Li
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, PR China
| | - Wei Zou
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, PR China.
| | - Xiang Li
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Anaesthesiology, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, PR China
| | - Chun-Yan Wang
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Pathophysiology, Medical College, University of South China, Hengyang, 421001, Hunan, PR China
| | - Ping Zhang
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, PR China
| | - Xiao-Qing Tang
- Institute of Neuroscience, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Physiology, Medical College, University of South China, Hengyang, 421001, Hunan, PR China; Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, PR China.
| |
Collapse
|
47
|
Sadeghi M, Reisi P, Radahmadi M. The effects of CCK-8S on spatial memory and long-term potentiation at CA1 during induction of stress in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2017; 20:1368-1376. [PMID: 29238473 PMCID: PMC5722998 DOI: 10.22038/ijbms.2017.9619] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Objectives Cholecystokinin (CCK) has been proposed as a mediator in stress. However, it is still not fully documented what are its effects. We aimed to evaluate the effects of systemic administration of CCK exactly before induction of stress on spatial memory and synaptic plasticity at CA1 in rats. Materials and Methods Male Wistar rats were divided into 4 groups: the control, the control-CCK, the stress and the stress-CCK. Restraint stress was induced 6 hr per day, for 24 days. Cholecystokinin sulfated octapeptide (CCK-8S) was injected (1.6 µg/kg, IP) before each session of stress induction. Spatial memory was evaluated by Morris water maze test. Long-term potentiation (LTP) in Schaffer collateral-CA1 synapses was assessed (by 100 Hz tetanization) in order to investigate synaptic plasticity. Results Stress impaired spatial memory significantly (P<0.01). CCK in the control rats improved memory (P<0.05), and prevented the impairments in the stress group. With respect to the control group, both fEPSP amplitude and slope were significantly (P<0.05) decreased in the stress group. However, there were no differences between responses of the control-CCK and Stress-CCK groups compared to the control group. Conclusion The present results suggest that high levels of CCK-8S during induction of stress can modulate the destructive effects of stress on hippocampal synaptic plasticity and memory. Therefore, the mediatory effects of CCK in stress are likely as compensatory responses.
Collapse
Affiliation(s)
- Malihe Sadeghi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parham Reisi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Radahmadi
- Department of Physiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
48
|
Othman H, Ammari M, Sakly M, Abdelmelek H. Effects of repeated restraint stress and WiFi signal exposure on behavior and oxidative stress in rats. Metab Brain Dis 2017; 32:1459-1469. [PMID: 28451780 DOI: 10.1007/s11011-017-0016-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/17/2017] [Indexed: 12/14/2022]
Abstract
Today, due to technology development and aversive events of daily life, Human exposure to both radiofrequency and stress is unavoidable. This study investigated the co-exposure to repeated restraint stress and WiFi signal on cognitive function and oxidative stress in brain of male rats. Animals were divided into four groups: Control, WiFi-exposed, restrained and both WiFi-exposed and restrained groups. Each of WiFi exposure and restraint stress occurred 2 h (h)/day during 20 days. Subsequently, various tests were carried out for each group, such as anxiety in elevated plus maze, spatial learning abilities in the water maze, cerebral oxidative stress response and cholinesterase activity in brain and serum. Results showed that WiFi exposure and restraint stress, alone and especially if combined, induced an anxiety-like behavior without impairing spatial learning and memory abilities in rats. At cerebral level, we found an oxidative stress response triggered by WiFi and restraint, per se and especially when combined as well as WiFi-induced increase in acetylcholinesterase activity. Our results reveal that there is an impact of WiFi signal and restraint stress on the brain and cognitive processes especially in elevated plus maze task. In contrast, there are no synergistic effects between WiFi signal and restraint stress on the brain.
Collapse
Affiliation(s)
- Haifa Othman
- Faculty of Sciences of Bizerte, Laboratory of Integrative Physiology, University of Carthage, 7021, Jarzouna, Tunisia
| | - Mohamed Ammari
- Faculty of Sciences of Bizerte, Laboratory of Integrative Physiology, University of Carthage, 7021, Jarzouna, Tunisia.
- Higher Institute of Applied Biological Sciences of Tunis, University of Tunis El Manar, 9, Rue Zouhair Essafi, 1006, Tunis, Tunisia.
| | - Mohsen Sakly
- Faculty of Sciences of Bizerte, Laboratory of Integrative Physiology, University of Carthage, 7021, Jarzouna, Tunisia
| | - Hafedh Abdelmelek
- Faculty of Sciences of Bizerte, Laboratory of Integrative Physiology, University of Carthage, 7021, Jarzouna, Tunisia
| |
Collapse
|
49
|
Dong L, Wang Y, Lv J, Zhang H, Jiang N, Lu C, Xu P, Liu X. Memory enhancement of fresh ginseng on deficits induced by chronic restraint stress in mice. Nutr Neurosci 2017; 22:235-242. [DOI: 10.1080/1028415x.2017.1373928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Liming Dong
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yi Wang
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingwei Lv
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongxia Zhang
- Division of Stem Cell Regulation and Application, Hunan University of Chinese Medicine, Changsha, China
| | - Ning Jiang
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cong Lu
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pan Xu
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xinmin Liu
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
50
|
Qie X, Wen D, Guo H, Xu G, Liu S, Shen Q, Liu Y, Zhang W, Cong B, Ma C. Endoplasmic Reticulum Stress Mediates Methamphetamine-Induced Blood-Brain Barrier Damage. Front Pharmacol 2017; 8:639. [PMID: 28959203 PMCID: PMC5603670 DOI: 10.3389/fphar.2017.00639] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/29/2017] [Indexed: 12/22/2022] Open
Abstract
Methamphetamine (METH) abuse causes serious health problems worldwide, and long-term use of METH disrupts the blood-brain barrier (BBB). Herein, we explored the potential mechanism of endoplasmic reticulum (ER) stress in METH-induced BBB endothelial cell damage in vitro and the therapeutic potential of endoplasmic reticulum stress inhibitors for METH-induced BBB disruption in C57BL/6J mice. Exposure of immortalized BMVEC (bEnd.3) cells to METH significantly decreased cell viability, induced apoptosis, and diminished the tightness of cell monolayers. METH activated ER stress sensor proteins, including PERK, ATF6, and IRE1, and upregulated the pro-apoptotic protein CHOP. The ER stress inhibitors significantly blocked the upregulation of CHOP. Knockdown of CHOP protected bEnd.3 cells from METH-induced cytotoxicity. Furthermore, METH elevated the production of reactive oxygen species (ROS) and induced the dysfunction of mitochondrial characterized by a Bcl2/Bax ratio decrease, mitochondrial membrane potential collapse, and cytochrome c. ER stress release was partially reversed by ROS inhibition, and cytochrome c release was partially blocked by knockdown of CHOP. Finally, PBA significantly attenuated METH-induced sodium fluorescein (NaFluo) and Evans Blue leakage, as well as tight junction protein loss, in C57BL/6J mice. These data suggest that BBB endothelial cell damage was caused by METH-induced endoplasmic reticulum stress, which further induced mitochondrial dysfunction, and that PBA was an effective treatment for METH-induced BBB disruption.
Collapse
Affiliation(s)
- Xiaojuan Qie
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China.,Department of Anesthesiology, The Third Hospital of Hebei Medical UniversityShijiazhuang, China
| | - Di Wen
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Hongyan Guo
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Guanjie Xu
- Department of Anesthesiology, The Third Hospital of Hebei Medical UniversityShijiazhuang, China
| | - Shuai Liu
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Qianchao Shen
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Yi Liu
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Wenfang Zhang
- The 8th Brigade of General Division of Criminal Investigation, Beijing Municipal Public Security BureauBeijing, China
| | - Bin Cong
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| | - Chunling Ma
- Hebei Key Laboratory of Forensic Medicine, Department of Forensic Medicine, Hebei Medical UniversityShijiazhuang, China
| |
Collapse
|