1
|
Grizzell JA, Clarity TT, Rodriguez RM, Marshall ZQ, Cooper MA. Effects of social dominance and acute social stress on morphology of microglia and structural integrity of the medial prefrontal cortex. Brain Behav Immun 2024; 122:353-367. [PMID: 39187049 PMCID: PMC11402560 DOI: 10.1016/j.bbi.2024.08.043] [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: 11/28/2023] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 08/28/2024] Open
Abstract
Chronic stress increases activity of the brain's innate immune system and impairs function of the medial prefrontal cortex (mPFC). However, whether acute stress triggers similar neuroimmune mechanisms is poorly understood. Across four studies, we used a Syrian hamster model to investigate whether acute stress drives changes in mPFC microglia in a time-, subregion-, and social status-dependent manner. We found that acute social defeat increased expression of ionized calcium binding adapter molecule 1 (Iba1) in the infralimbic (IL) and prelimbic (PL) and altered the morphology Iba1+ cells 1, 2, and 7 days after social defeat. We also investigated whether acute defeat induced tissue degeneration and reductions of synaptic plasticity 2 days post-defeat. We found that while social defeat increased deposition of cellular debris and reduced synaptophysin immunoreactivity in the PL and IL, treatment with minocycline protected against these cellular changes. Finally, we tested whether a reduced conditioned defeat response in dominant compared to subordinate hamsters was associated with changes in microglia reactivity in the IL and PL. We found that while subordinate hamsters and those without an established dominance relationships showed defeat-induced changes in morphology of Iba1+ cells and cellular degeneration, dominant hamsters showed resistance to these effects of social defeat. Taken together, these findings indicate that acute social defeat alters microglial morphology, increases markers of tissue degradation, and impairs structural integrity in the IL and PL, and that experience winning competitive interactions can specifically protect the IL and reduce stress vulnerability.
Collapse
Affiliation(s)
- J Alex Grizzell
- Neuroscience and Behavioral Biology Program, Emory University, United States; Department of Psychology, University of Tennessee Knoxville, United States; Department of Psychology and Neurosciences, University of Colorado Boulder, United States
| | - Thomas T Clarity
- Department of Psychology, University of Tennessee Knoxville, United States
| | - R Mason Rodriguez
- Department of Psychology, University of Tennessee Knoxville, United States
| | - Zachary Q Marshall
- Department of Psychology and Neurosciences, University of Colorado Boulder, United States
| | - Matthew A Cooper
- Department of Psychology, University of Tennessee Knoxville, United States.
| |
Collapse
|
2
|
Yang L, Xing W, Shi Y, Hu M, Li B, Hu Y, Zhang G. Stress-induced NLRP3 inflammasome activation and myelin alterations in the hippocampus of PTSD rats. Neuroscience 2024; 555:156-166. [PMID: 39043314 DOI: 10.1016/j.neuroscience.2024.07.028] [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: 01/31/2024] [Revised: 07/14/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024]
Abstract
Inflammatory and myelin changes may contribute to the pathophysiology of post-traumatic stress disorder (PTSD). The NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3), a brain inflammasome, is activated in the hippocampus of mice with PTSD. In other psychiatric disorders, NLRP3 expression has been associated with axonal myelination and demyelination. However, the association between NLRP3 and myelin in rats with PTSD remains unclear. Therefore, this study aims to investigate the relationship between the NLRP3 inflammasome and myelin in the hippocampus of rats with PTSD. A rat model of post-traumatic stress disorder was established using the single-prolonged stress (SPS) approach. Hippocampal tissues were collected for the detection of NLRP3 inflammasome-associated proteins and myelin basic protein at 3, 7, and 14 days after SPS. To further explore the relationship between NLRP3 and myelin, the NLRP3-specific inhibitor MCC950 was administered intraperitoneally to rats starting 72 h before SPS, and then alterations in NLRP3 inflammasome-associated proteins and myelin were observed in the PTSD and control groups. We found that NLRP3 and downstream related proteins were activated in the hippocampus of rats 3 days after SPS, and the myelin content in the hippocampus increased after SPS stress. MCC950 reduced the expression of NLRP3-related pathway proteins, improved anxiety behaviour and spatial learning memory impairment, and inhibited the increase in myelin content in the hippocampal region of rats after SPS. In conclusion the study indicates that NLRP3 has a significant role in the hippocampal region of rats with PTSD. Inhibition of the NLRP3 inflammasome could be a potential target for treating PTSD.
Collapse
Affiliation(s)
- Luodong Yang
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Wenlong Xing
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Yan Shi
- Shihezi University, Shihezi, China
| | - Min Hu
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Bin Li
- Shihezi University, Shihezi, China
| | - Yuanyuan Hu
- First Affiliated Hospital of Shihezi University, Shihezi, China
| | - Guiqing Zhang
- First Affiliated Hospital of Shihezi University, Shihezi, China.
| |
Collapse
|
3
|
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
|
4
|
Jyonouchi H. Autism spectrum disorder and a possible role of anti-inflammatory treatments: experience in the pediatric allergy/immunology clinic. Front Psychiatry 2024; 15:1333717. [PMID: 38979496 PMCID: PMC11228311 DOI: 10.3389/fpsyt.2024.1333717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 06/03/2024] [Indexed: 07/10/2024] Open
Abstract
Autism spectrum disorder (ASD1) is a behaviorally defined syndrome encompassing a markedly heterogeneous patient population. Many ASD subjects fail to respond to the 1st line behavioral and pharmacological interventions, leaving parents to seek out other treatment options. Evidence supports that neuroinflammation plays a role in ASD pathogenesis. However, the underlying mechanisms likely vary for each ASD patient, influenced by genetic, epigenetic, and environmental factors. Although anti-inflammatory treatment measures, mainly based on metabolic changes and oxidative stress, have provided promising results in some ASD subjects, the use of such measures requires the careful selection of ASD subjects based on clinical and laboratory findings. Recent progress in neuroscience and molecular immunology has made it possible to allow re-purposing of currently available anti-inflammatory medications, used for autoimmune and other chronic inflammatory conditions, as treatment options for ASD subjects. On the other hand, emerging anti-inflammatory medications, including biologic and gate-keeper blockers, exert powerful anti-inflammatory effects on specific mediators or signaling pathways. It will require both a keen understanding of the mechanisms of action of such agents and the careful selection of ASD patients suitable for each treatment. This review will attempt to summarize the use of anti-inflammatory agents already used in targeting ASD patients, and then emerging anti-inflammatory measures applicable for ASD subjects based on scientific rationale and clinical trial data, if available. In our experience, some ASD patients were treated under diagnoses of autoimmune/autoinflammatory conditions and/or post-infectious neuroinflammation. However, there are little clinical trial data specifically for ASD subjects. Therefore, these emerging immunomodulating agents for potential use for ASD subjects will be discussed based on preclinical data, case reports, or data generated in patients with other medical conditions. This review will hopefully highlight the expanding scope of immunomodulating agents for treating neuroinflammation in ASD subjects.
Collapse
Affiliation(s)
- Harumi Jyonouchi
- Department of Pediatrics, Saint Peter's University Hospital, New Brunswick, NJ, United States
- Department of Pediatrics, Rutgers University-Robert Wood Johnson School of Medicine, New Brunswick, NJ, United States
| |
Collapse
|
5
|
Iglesias LP, Soares N, Asth L, Moreira FA, Aguiar DC. Minocycline as a potential anxiolytic drug: systematic review and meta-analysis of evidence in murine models. Behav Pharmacol 2024; 35:4-13. [PMID: 38375658 DOI: 10.1097/fbp.0000000000000754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
Minocycline is a tetracycline antibiotic with off-label use as an anti-inflammatory drug. Because it can cross the blood-brain barrier, minocycline has been proposed as an alternative treatment for psychiatric disorders, in which inflammation plays an important role. However, its beneficial effects on anxiety disorders are unclear. Therefore, we performed a systematic review and meta-analysis to evaluate the efficacy of minocycline as an anxiolytic drug in preclinical models. We performed a PubMed search according to the PRISMA guidelines and PICOS strategy. The risk of bias was evaluated using the SYRCLE tool. We included studies that determined the efficacy of minocycline in animal models of anxiety that may involve exposures (e.g. stressors, immunomodulators, injury). Data extracted included treatment effect, dose range, route of administration, and potential mechanisms for the anxiolytic effect. Meta-analysis of twenty studies showed that minocycline reduced anxiety-like behavior in rodents previously exposed to stress or immunostimulants but not in exposure-naïve animals. This effect was not associated with the dose administered or treatment duration. The mechanism for the anxiolytic activity of minocycline may depend on its anti-inflammatory effects in the brain regions involving anxiety. These suggest that minocycline could be repurposed as a treatment for anxiety and related disorders and warrants further evaluation.
Collapse
Affiliation(s)
- Lia P Iglesias
- Graduate School in Neuroscience, Universidade Federal de Minas Gerais (UFMG)
| | - Nicia Soares
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Laila Asth
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Fabricio A Moreira
- Graduate School in Neuroscience, Universidade Federal de Minas Gerais (UFMG)
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Daniele C Aguiar
- Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
6
|
Burback L, Brémault-Phillips S, Nijdam MJ, McFarlane A, Vermetten E. Treatment of Posttraumatic Stress Disorder: A State-of-the-art Review. Curr Neuropharmacol 2024; 22:557-635. [PMID: 37132142 PMCID: PMC10845104 DOI: 10.2174/1570159x21666230428091433] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/19/2023] [Accepted: 02/23/2023] [Indexed: 05/04/2023] Open
Abstract
This narrative state-of-the-art review paper describes the progress in the understanding and treatment of Posttraumatic Stress Disorder (PTSD). Over the last four decades, the scientific landscape has matured, with many interdisciplinary contributions to understanding its diagnosis, etiology, and epidemiology. Advances in genetics, neurobiology, stress pathophysiology, and brain imaging have made it apparent that chronic PTSD is a systemic disorder with high allostatic load. The current state of PTSD treatment includes a wide variety of pharmacological and psychotherapeutic approaches, of which many are evidence-based. However, the myriad challenges inherent in the disorder, such as individual and systemic barriers to good treatment outcome, comorbidity, emotional dysregulation, suicidality, dissociation, substance use, and trauma-related guilt and shame, often render treatment response suboptimal. These challenges are discussed as drivers for emerging novel treatment approaches, including early interventions in the Golden Hours, pharmacological and psychotherapeutic interventions, medication augmentation interventions, the use of psychedelics, as well as interventions targeting the brain and nervous system. All of this aims to improve symptom relief and clinical outcomes. Finally, a phase orientation to treatment is recognized as a tool to strategize treatment of the disorder, and position interventions in step with the progression of the pathophysiology. Revisions to guidelines and systems of care will be needed to incorporate innovative treatments as evidence emerges and they become mainstream. This generation is well-positioned to address the devastating and often chronic disabling impact of traumatic stress events through holistic, cutting-edge clinical efforts and interdisciplinary research.
Collapse
Affiliation(s)
- Lisa Burback
- Department of Psychiatry, University of Alberta, Edmonton, Canada
| | | | - Mirjam J. Nijdam
- ARQ National Psychotrauma Center, Diemen, The Netherlands
- Department of Psychiatry, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | | | - Eric Vermetten
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
- Department of Psychiatry, New York University Grossman School of Medicine, New York, USA
| |
Collapse
|
7
|
Chen XD, Wei JX, Wang HY, Peng YY, Tang C, Ding Y, Li S, Long ZY, Lu XM, Wang YT. Effects and mechanisms of salidroside on the behavior of SPS-induced PTSD rats. Neuropharmacology 2023; 240:109728. [PMID: 37742716 DOI: 10.1016/j.neuropharm.2023.109728] [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: 08/08/2023] [Revised: 09/13/2023] [Accepted: 09/22/2023] [Indexed: 09/26/2023]
Abstract
Post-traumatic stress disorder (PTSD) is a complex mental disorder, closely associated with stress and traumatic events. Salidroside (Sal) has been reported to possess neuroprotective effects. However, the behavioral effects and mechanisms of Sal on PTSD remain unknown. In this study, we utilized a rat model of PTSD induced by single prolonged stress (SPS) and administered Sal intraperitoneally (25, 50, 75 mg/kg/d) for 14 days. We then examined the behavioral effects and underlying mechanisms of Sal on SPS-induced PTSD rats. Our findings demonstrated that Sal alleviated anxiety-like behavior and spatial learning and memory impairment in SPS-induced PTSD rats. Furthermore, Sal treatment preserved the histomorphology of the hippocampal region. It was observed that Sal protected against hippocampal neuronal apoptosis in PTSD rats by reducing the number of TUNEL-positive cells and modulating apoptosis-related proteins (Bcl-2 and Bax). Additionally, Sal inhibited the activation of the NF-κB/iNOS/COX-2 signaling pathway in the hippocampus of PTSD rats, thereby suppressing the release of inflammatory factors (TNF-α and IL-1β) and the activation of microglia. Notably, Sal increased the expression of synapse-associated proteins PSD95 and Synapsin I in the hippocampus, while also enhancing dendritic density in the region. In conclusion, our results demonstrated that Sal could attenuate SPS-induced PTSD-like behaviors by inhibiting hippocampal neuronal apoptosis, enhancing hippocampal synaptic plasticity, and reducing neuroinflammatory responses. These findings may provide a foundation for the potential clinical application of Sal in the treatment of PTSD.
Collapse
Affiliation(s)
- Xing-Dong Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China; College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jing-Xiang Wei
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yu-Yuan Peng
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Can Tang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yang Ding
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Sen Li
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zai-Yun Long
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
| | - Yong-Tang Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
8
|
Zhang Z, Guo Z, Jin P, Yang H, Hu M, Zhang Y, Tu Z, Hou S. Transcriptome Profiling of Hippocampus After Cerebral Hypoperfusion in Mice. J Mol Neurosci 2023; 73:423-436. [PMID: 37266840 PMCID: PMC10432347 DOI: 10.1007/s12031-023-02123-0] [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: 02/10/2023] [Accepted: 05/18/2023] [Indexed: 06/03/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) is considered to be one of the major mechanism in the pathogenesis of vascular cognitive impairment (VCI). Increased inflammatory cells, particularly microglia, often parallel hypoperfusion-induced gray matter damage such as hippocampal lesions, but the exact mechanism remains largely unknown. To understand the pathological mechanisms, we analyzed hippocampus-specific transcriptome profiles after cerebral hypoperfusion. The mouse hypoperfusion model was induced by employing the 0.16/0.18 mm bilateral common carotid artery stenosis (BCAS) procedure. Cerebral blood flow (CBF) was assessed after 3-week hypoperfusion. Pathological changes were evaluated via hematoxylin staining and immunofluorescence staining. RNA-sequencing (RNA-seq) was performed using RNA samples of sham- or BCAS-operated mice, followed by quantitative real-time PCR (qRT-PCR) validation. We found that the 0.16/0.18 mm BCAS induced decreased CBF, hippocampal neuronal loss, and microglial activation. Furthermore, GSEA between sham and BCAS mice showed activation of interferon-beta signaling along with inflammatory immune responses. In addition, integrative analysis with published single-cell RNA-seq revealed that up-regulated differentially expressed genes (DEGs) were enriched in a distinct cell type of "microglia," and down-regulated DEGs were enriched in "CA1 pyramidal," not in "interneurons" or "S1 pyramidal." This database of transcriptomic profiles of BCAS-hypoperfusion will be useful for future studies to explore potential targets for vascular cognitive dysfunction.
Collapse
Affiliation(s)
- Zengyu Zhang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zimin Guo
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Pengpeng Jin
- Department of Chronic Disease Management, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Hualan Yang
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Mengting Hu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Yuan Zhang
- Department of Vascular Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, 201399, Shanghai, China
| | - Zhilan Tu
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| | - Shuangxing Hou
- Department of Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
9
|
Li J, Tong L, Schock BC, Ji LL. Post-traumatic Stress Disorder: Focus on Neuroinflammation. Mol Neurobiol 2023; 60:3963-3978. [PMID: 37004607 DOI: 10.1007/s12035-023-03320-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 03/09/2023] [Indexed: 04/04/2023]
Abstract
Post-traumatic stress disorder (PTSD), gaining increasing attention, is a multifaceted psychiatric disorder that occurs following a stressful or traumatic event or series of events. Recently, several studies showed a close relationship between PTSD and neuroinflammation. Neuroinflammation, a defense response of the nervous system, is associated with the activation of neuroimmune cells such as microglia and astrocytes and with changes in inflammatory markers. In this review, we first analyzed the relationship between neuroinflammation and PTSD: the effect of stress-derived activation of the hypothalamic-pituitary-adrenal (HPA) axis on the main immune cells in the brain and the effect of stimulated immune cells in the brain on the HPA axis. We then summarize the alteration of inflammatory markers in brain regions related to PTSD. Astrocytes are neural parenchymal cells that protect neurons by regulating the ionic microenvironment around neurons. Microglia are macrophages of the brain that coordinate the immunological response. Recent studies on these two cell types provided new insight into neuroinflammation in PTSD. These contribute to promoting comprehension of neuroinflammation, which plays a pivotal role in the pathogenesis of PTSD.
Collapse
Affiliation(s)
- Jimeng Li
- Department of 2nd Clinical College, China Medical University, Shenyang, Liaoning, China
| | - Lei Tong
- Department of Anatomy, College of Basic Sciences, China Medical University, Shenyang, Liaoning, China
| | - Bettina C Schock
- The Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast Faculty of Medicine Health and Life Sciences, Belfast, UK
| | - Li-Li Ji
- Department of Anatomy, College of Basic Sciences, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
10
|
Li N, Yao CY, Diao J, Liu XL, Tang EJ, Huang QS, Zhou YM, Hu YG, Li XK, Long JY, Xiao H, Li DW, Du N, Li YF, Luo P, Cai TJ. The role of MAPK/NF-κB-associated microglial activation in T-2 toxin-induced mouse learning and memory impairment. Food Chem Toxicol 2023; 174:113663. [PMID: 36775139 DOI: 10.1016/j.fct.2023.113663] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 12/10/2022] [Accepted: 02/09/2023] [Indexed: 02/12/2023]
Abstract
T-2 toxin is a mycotoxin with multiple toxic effects and has emerged as an important food pollutant. Microglia play a significant role in the toxicity of various neurotoxins. However, whether they participate in the neurotoxicity of T-2 toxin has not been reported. To clarify this point, an in vivo mouse model of T-2 toxin (4 mg/kg) poisoning was established. The results of Morris water maze and open-field showed that T-2 toxin induced learning and memory impairment and locomotor inhibition. Meanwhile, T-2 toxin induced microglial activation, while inhibiting microglia activation by minocycline (50 mg/kg) suppressed the toxic effect of the T-2 toxin. To further unveil the potential mechanisms involved in T-2 toxin-induced microglial activation, an in vitro model of T-2 toxin (0, 2.5, 5, 10 ng/mL) poisoning was established using BV-2 cells. Transcriptomic sequencing revealed lots of differentially expressed genes related to MAPK/NF-κB pathway. Western blotting results further confirmed that T-2 toxin (5 ng/mL) induced the activation of MAPKs and their downstream NF-κB. Moreover, the addition of inhibitors of NF-κB and MAPKs reversed the microglial activation induced by T-2 toxin. Overall, microglial activation may contribute a considerable role in T-2 toxin-induced behavioral abnormalities, which could be MAPK/NF-κB pathway dependent.
Collapse
Affiliation(s)
- Na Li
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China; Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Chun-Yan Yao
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jun Diao
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Chongqing Jiulongpo District Center for Disease Control and Prevention, Chongqing, 400050, China
| | - Xiao-Ling Liu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - En-Jie Tang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Qing-Song Huang
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China; Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yu-Meng Zhou
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yue-Gu Hu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiu-Kuan Li
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China; Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jin-Yun Long
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China; Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Hua Xiao
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Da-Wei Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Ning Du
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Ya-Fei Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Peng Luo
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China.
| | - Tong-Jian Cai
- School of Public Health, Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China; Department of Epidemiology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
11
|
Pate BS, Bouknight SJ, Harrington EN, Mott SE, Augenblick LM, Smiley CE, Morgan CG, Calatayud BM, Martínez-Muñiz GA, Thayer JF, Wood SK. Site-Specific knockdown of microglia in the locus coeruleus regulates hypervigilant responses to social stress in female rats. Brain Behav Immun 2023; 109:190-203. [PMID: 36682513 PMCID: PMC11195023 DOI: 10.1016/j.bbi.2023.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Women are at increased risk for psychosocial stress-related anxiety disorders, yet mechanisms regulating this risk are unknown. Psychosocial stressors activate microglia, and the resulting neuroimmune responses that females exhibit heightened sensitivity to may serve as an etiological factor in their elevated risk. However, studies examining the role of microglia during stress in females are lacking. METHODS Microglia were manipulated in the stress-sensitive locus coeruleus (LC) of female rats in the context of social stress in two ways. First, intra-LC lipopolysaccharide (LPS; 0 or 3 μg/side, n = 5-6/group), a potent TLR4 agonist and microglial activator, was administered. One hour later, rats were exposed to control or an aggressive social defeat encounter between two males (WS, 15-min). In a separate study, females were treated with intra-LC or intra-central amygdala mannosylated liposomes containing clodronate (m-CLD; 0 or 25 μg/side, n = 13-14/group), a compound toxic to microglia. WS-evoked burying, cardiovascular responses, and sucrose preference were measured. Brain and plasma cytokines were quantified, and cardiovascular telemetry assessed autonomic balance. RESULTS Intra-LC LPS augmented the WS-induced burying response and increased plasma corticosterone and interleukin-1β (IL-1β). Further, the efficacy and selectivity of microinjected m-CLD was fully characterized. In the context of WS, intra-LC m-CLD attenuated the hypervigilant burying response during WS as well as the accumulation of intra-LC IL-1β. Intra-central amygdala m-CLD had no effect on WS-evoked behavior. CONCLUSIONS These studies highlight an innovative method for depleting microglia in a brain region specific manner and indicate that microglia in the LC differentially regulate hypervigilant WS-evoked behavioral and autonomic responses.
Collapse
Affiliation(s)
- Brittany S Pate
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA; Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Samantha J Bouknight
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Evelynn N Harrington
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Sarah E Mott
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Lee M Augenblick
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Cora E Smiley
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA; WJB Dorn VA Medical Center, Columbia, SC, USA
| | - Christopher G Morgan
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Brittney M Calatayud
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Gustavo A Martínez-Muñiz
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, CA, USA
| | - Susan K Wood
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA; WJB Dorn VA Medical Center, Columbia, SC, USA.
| |
Collapse
|
12
|
Jiang H, Chen L, Li Y, Gao X, Yang X, Zhao B, Li Y, Wang Y, Yu X, Zhang X, Feng S, Chai Y, Meng H, Ren X, Bao T. Effects of acupuncture on regulating the hippocampal inflammatory response in rats exposed to post-traumatic stress disorder. Neurosci Lett 2023; 796:137056. [PMID: 36621587 DOI: 10.1016/j.neulet.2023.137056] [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: 09/16/2022] [Revised: 12/18/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
Data from clinical and experimental studies have verified the efficacy and safety of acupuncture in the treatment of post-traumatic stress disorder (PTSD). However, the concrete mechanism has not been well elucidated. The stress-induced activation of inflammatory response is involved in the development and pathogenesis of PTSD. Here, we aimed to investigate the effects of acupuncture on regulating the hippocampal inflammatory response in rats exposed to PTSD. Forty male rats were randomly divided into control, model, acupuncture and sertraline group. Within 1 day after adaptive feeding, all rats were exposed to single prolonged stress (SPS), except for the rats in the control group. Rats in acupuncture group were exposed to acupuncture intervention at the acupoints of Baihui (GV20) and Yintang (GV29), 20 min once per day for 15 days. Rats in sertraline group were exposed to a suspension of sertraline and distilled water (0.2 mg/ml), once per day for 15 days continuously. Body weight and elevated plus maze experiment were detected at different time-points to evaluate the behavioral changes of rats. HE staining method was used to observe the basic pathological morphological changes in hippocampus. Immunofluorescence staining method was used to observe the activation of hippocampal microglia. The content of IL-6 and IL-1β in serum were detected by ELISA method. Compared with the control group, the body weight of rats in model group significantly decreased on 8 days, and the percentage of time in open arms and open arm entries decreased significantly on 15 days after SPS procedures, which indicated that SPS induced PTSD-like behavior in rats. Acupuncture exerted therapeutic effect. Simultaneously, the result of HE staining confirmed that SPS induced hippocampal morphological changes in SPS rats. Notably, acupuncture reversed the reduction and pathological injury to some extent. The results have also shown that acupuncture intervention effectively reversed the activated microglia of the hippocampus in rats. Moreover, the expression of IL-1β in serum was significantly decreased by acupuncture intervention. In summary, the present study demonstrated that the role of acupuncture in eliminating PTSD-like behavior might be connected with reversing the pathological process of the inflammatory response mediated by the activation of microglia induced by SPS.
Collapse
Affiliation(s)
- Huili Jiang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.
| | - Lu Chen
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yufei Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xingzhou Gao
- Beijing ChangPing District Hospital, Beijing, China
| | - Xinjing Yang
- Department of Traditional Chinese Medicine, South China Hospital of Shenzhen University, Shenzhen, China
| | - Bingcong Zhao
- Beijing Key Laboratory of Acupuncture Neuromodulation, Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yahuan Li
- Beijing Increase Biomedical Company Limited, Beijing, China
| | - Yu Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xue Yu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | | | - Shixing Feng
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yemao Chai
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Hong Meng
- School of Science, Beijing Technology and Business University, Beijing, China
| | - Xiujun Ren
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Tuya Bao
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
13
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
14
|
Motaghinejad M, Motevalian M. Neuroprotective Properties of Minocycline Against Methylphenidate-Induced Neurodegeneration: Possible Role of CREB/BDNF and Akt/GSK3 Signaling Pathways in Rat Hippocampus. Neurotox Res 2022; 40:689-713. [PMID: 35446003 DOI: 10.1007/s12640-021-00454-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/17/2021] [Accepted: 11/29/2021] [Indexed: 12/19/2022]
Abstract
Neurodegeneration is a side effect of methylphenidate (MPH), and minocycline possesses neuroprotective properties. This study aimed to investigate the neuroprotective effects of minocycline against methylphenidate-induced neurodegeneration mediated by signaling pathways of CREB/BDNF and Akt/GSK3. Seven groups of seventy male rats were randomly distributed in seven groups (n = 10). Group 1 received 0.7 ml/rat of normal saline (i.p.), and group 2 was treated with MPH (10 mg/kg, i.p.). Groups 3, 4, 5, and 6 were simultaneously administered MPH (10 mg/kg) and minocycline (10, 20, 30, and 40 mg/kg, i.p.) for 21 days. Minocycline alone (40 mg/kg, i.p.) was administrated to group 7. Open field test (OFT) (on day 22), forced swim test (FST) (on day 24), and elevated plus maze (on day 26) were conducted to analyze the mood-related behaviors; hippocampal oxidative stress, inflammatory, and apoptotic parameters, as well as the levels of protein kinase B (Akt-1), glycogen synthase kinase 3 (GSK3), cAMP response element-binding protein (CREB), and brain-derived neurotrophic factor (BDNF), were also assessed. Furthermore, localization of total CREB, Akt, and GSK3 in the DG and CA1 areas of the hippocampus were measured using immunohistochemistry (IHC). Histological changes in the mentioned areas were also evaluated. Minocycline treatment inhibited MPH-induced mood disorders and decreased lipid peroxidation, oxidized form of glutathione (GSSG), interleukin 1 beta (IL-1β), alpha tumor necrosis factor (TNF-α), Bax, and GSK3 levels. In the contrary, it increased the levels of reduced form of glutathione (GSH), Bcl-2, CREB, BDNF, and Akt-1 and superoxide dismutase (SOD), glutathione peroxidase (GPx), and glutathione reductase (GR) activities in the experimental animals' hippocampus. IHC data showed that minocycline also improved the localization and expression of CREB and Akt positive cells and decreased the GSK3 positive cells in the DG and CA1 regions of the hippocampus of MPH-treated rats. Minocycline also inhibited MPH-induced changes of hippocampal cells' density and shape in both DG and CA1 areas of the hippocampus. According to obtained data, it can be concluded that minocycline probably via activation of the P-CREB/BDNF or Akt/GSK3 signaling pathway can confer its neuroprotective effects against MPH-induced neurodegeneration.
Collapse
Affiliation(s)
- Majid Motaghinejad
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
| | - Manijeh Motevalian
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
- Department of Pharmacology, School of Medicine, Tehran, Iran University of Medical Sciences, Shahid Hemmat High way, Iran Univ. Med. Sci., P.O. Box 14496-14525, Tehran, Iran.
| |
Collapse
|
15
|
Avalos MP, Guzman AS, Rigoni D, Gorostiza EA, Sanchez MA, Mongi-Bragato B, Garcia-Keller C, Perassi EM, Virgolini MB, Peralta Ramos JM, Iribarren P, Calfa GD, Bollati FA, Cancela LM. Minocycline prevents chronic restraint stress-induced vulnerability to developing cocaine self-administration and associated glutamatergic mechanisms: a potential role of microglia. Brain Behav Immun 2022; 101:359-376. [PMID: 35065197 DOI: 10.1016/j.bbi.2022.01.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/24/2021] [Accepted: 01/16/2022] [Indexed: 12/14/2022] Open
Abstract
Stressful experience-induced cocaine-related behaviors are associated with a significant impairment of glutamatergic mechanisms in the Nucleus Accumbens core (NAcore). The hallmarks of disrupted glutamate homeostasis following restraint stress are the enduring imbalance of glutamate efflux after a cocaine stimulus and increased basal concentrations of extracellular glutamate attributed to GLT-1 downregulation in the NAcore. Glutamate transmission is tightly linked to microglia functioning. However, the role of microglia in the biological basis of stress-induced addictive behaviors is still unknown. By using minocycline, a potent inhibitor of microglia activation with anti-inflammatory properties, we determined whether microglia could aid chronic restraint stress (CRS)-induced glutamate homeostasis disruption in the NAcore, underpinning stress-induced cocaine self-administration. In this study, adult male rats were restrained for 2 h/day for seven days (day 1-7). From day 16 until completing the experimental protocol, animals received a vehicle or minocycline treatment (30 mg/Kg/12h i.p.). On day 21, animals were assigned to microscopic, biochemical, neurochemical or behavioral studies. We confirm that the CRS-induced facilitation of cocaine self-administration is associated with enduring GLT-1 downregulation, an increase of basal extracellular glutamate and postsynaptic structural plasticity in the NAcore. These alterations were strongly related to the CRS-induced reactive microglia and increased TNF-α mRNA and protein expression, since by administering minocycline, the impaired glutamate homeostasis and the facilitation of cocaine self-administration were prevented. Our findings are the first to demonstrate that minocycline suppresses the CRS-induced facilitation of cocaine self-administration and glutamate homeostasis disruption in the NAcore. A role of microglia is proposed for the development of glutamatergic mechanisms underpinning stress-induced vulnerability to cocaine addiction.
Collapse
Affiliation(s)
- María Paula Avalos
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Andrea Susana Guzman
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Daiana Rigoni
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Ezequiel Axel Gorostiza
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Marianela Adela Sanchez
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Bethania Mongi-Bragato
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Constanza Garcia-Keller
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Eduardo Marcelo Perassi
- Instituto de Investigaciones en Fisicoquímica de Córdoba (INFIQC-CONICET), Departamento de Química Teórica y Computacional, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Miriam Beatriz Virgolini
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Javier María Peralta Ramos
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Pablo Iribarren
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Gastón Diego Calfa
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina
| | - Flavia Andrea Bollati
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| | - Liliana Marina Cancela
- Instituto de Farmacología Experimental de Córdoba (IFEC-CONICET), Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, X5000HUA, Córdoba, Argentina.
| |
Collapse
|
16
|
Ni K, Zhu J, Xu X, Liu Y, Yang S, Huang Y, Xu R, Jiang L, Zhang J, Zhang W, Ma Z. Hippocampal Activated Microglia May Contribute to Blood–Brain Barrier Impairment and Cognitive Dysfunction in Post-Traumatic Stress Disorder-Like Rats. J Mol Neurosci 2022; 72:975-982. [PMID: 35167061 PMCID: PMC8852956 DOI: 10.1007/s12031-022-01981-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/02/2022] [Indexed: 12/20/2022]
Abstract
Post-traumatic stress disorder (PTSD)-associated cognitive dysfunction significantly disturbs patients’ quality of life and will to live. However, its underlying mechanism is as yet unknown. Recent researches indicate that blood–brain barrier (BBB) breakdown is responsible for early cognitive dysfunction. Microglia might participate in remodeling of BBB-associated tight junction and regulating BBB integrity. Nevertheless, it is unclear whether microglia activation and BBB injury involve in PTSD-associated cognitive dysfunction. Hence, we established an animal model of PTSD, single prolonged stress (SPS), and investigated permeability changes in the hippocampus and further explored the effects of microglia on BBB remodeling. The Y maze was used to assess the changes of cognitive function. The sodium fluorescein (NaFlu) assay and western blotting analysis were employed to detect BBB integrity changes. Minocycline was administered to inhibit microglial activation. Immunofluorescence stains were used to assess the activation states in microglia. The results showed that SPS-exposed rats exhibited poorer cognitive performance, higher passage of NaFlu, and lower expression of tight junction proteins (occludin and claudin 5) in the hippocampus on the day after SPS, but no difference on the 7th day. Inhibition of microglial activation by minocycline attenuated poor cognitive performance and BBB impairment including the extravasation of NaFlu and protein levels of the tight junction. Taken together, the present study indicates that BBB impairment may underlie the shared pathological basis of PTSD and cognitive dysfunction. Microglial activation may involve in BBB remodeling at the early stage of SPS.
Collapse
Affiliation(s)
- Kun Ni
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Jixiang Zhu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Xuan Xu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yue Liu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Shuai Yang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Yulin Huang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Rui Xu
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Li Jiang
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, China
| | - Juan Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Wei Zhang
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
| | - Zhengliang Ma
- Department of Anesthesiology, Drum Tower Hospital, Medical School of Nanjing University, 321 Zhongshan Road, Nanjing, 210008, China.
- Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, 321 Zhongshan Road, Nanjing, 210008, China.
| |
Collapse
|
17
|
Lee B, Sur B, Oh S. Neuroprotective effect of Korean red ginseng against single prolonged stress-induced memory impairments and inflammation in the rat brain associated with BDNF expression. J Ginseng Res 2021; 46:435-443. [PMID: 35600771 PMCID: PMC9120622 DOI: 10.1016/j.jgr.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/04/2021] [Accepted: 08/05/2021] [Indexed: 11/25/2022] Open
Abstract
Background Post-traumatic stress disorder (PTSD) is a psychiatric disease that develops following exposure to a traumatic event and is a stress-associated mental disorder characterized by an imbalance of neuroinflammation. Korean Red Ginseng (KRG) is the herbal supplement that is known to be involved in a variety of pharmacological activities. We aimed to investigate the effects of KRG on neuroinflammation as a potential mechanism involved in single prolonged stress (SPS) that negatively influences memory formation and consolidation and leads to cognitive and spatial impairment by regulating BDNF signaling, synaptic proteins, and the activation of NF-kB. Methods We analyzed the cognitive and spatial memory, and inflammatory cytokine levels during the SPS procedure. SPS model rats were injected intraperitoneally with 20, 50, or 100 mg/kg/day KRG for 14 days. Results KRG administration significantly attenuated the cognitive and spatial memory deficits, as well as the inflammatory reaction in the hippocampus associated with activation of NF-κB in the hippocampus induced by SPS. Moreover, the effects of KRG were equivalent to those exerted by paroxetine. In addition, KRG improved the expression of BDNF mRNA and the synaptic protein PSD-95 in the hippocampus. Taken together, these findings demonstrate that KRG exerts memory-improving actions by regulating anti-inflammatory activities and the NF-κB and neurotrophic pathway. Conclusion Our findings suggest that KRG is a potential functional ingredient for protecting against memory deficits in mental diseases, such as PTSD.
Collapse
|
18
|
Patankar PS, Joshi S, Mane A, Manjesh PS, Kokate D. Anxiolytic effect of minocycline in posttraumatic stress disorder model of Syrian hamsters. Tzu Chi Med J 2021; 33:263-269. [PMID: 34386364 PMCID: PMC8323640 DOI: 10.4103/tcmj.tcmj_243_20] [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: 09/19/2020] [Revised: 11/13/2020] [Accepted: 12/07/2020] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE The objective was to study the anxiolytic effect of minocycline in resident-intruder social conflict in submissive hamsters post resident intrusion model using open field test (OFT) and elevated plus maze (EPM) and serum cortisol levels. MATERIALS AND METHODS Fifty-two singly housed male Syrian hamsters were used, post standardization of an animal model. Resident intrusion was done (5 min), in which smaller hamsters were placed in the cage of larger hamster, and the behavior of smaller hamster was noted. Eight submissive hamsters per group (disease control, lorazepam group as a positive control, and the test drug was minocycline) were used, and the drug was administered immediately post resident intrusion, intraperitoneally. Behavioral tests, namely OFT and EPM, were done followed by retro-orbital blood collection for serum cortisol estimation. The level of significance was set at P < 0.05. RESULTS The minocycline group showed a statistically significant decrease in serum cortisol levels compared to the disease control group. Among all the variables pertaining to both the behavioral tests, namely EPM and OFT, the results indicated an anxiolytic effect, which was statistically significant compared to the disease control group. CONCLUSION As per the biochemical test using serum cortisol levels and behavioral tests in the form of EPM and OFT, the study concluded that the anxiolytic effect of minocycline is at least comparable to the positive control, lorazepam.
Collapse
Affiliation(s)
- Panini Shrikant Patankar
- Department of Pharmacology and Therapeutics, Seth GSMC and KEM Hospital, Mumbai, Maharashtra, India
| | - Shirish Joshi
- Department of Pharmacology and Therapeutics, Seth GSMC and KEM Hospital, Mumbai, Maharashtra, India
| | - Abhishek Mane
- Department of Pharmacology and Therapeutics, Seth GSMC and KEM Hospital, Mumbai, Maharashtra, India
| | - P. S. Manjesh
- Department of Pharmacology and Therapeutics, Seth GSMC and KEM Hospital, Mumbai, Maharashtra, India
| | - Dhananjay Kokate
- Department of Pharmacology and Therapeutics, Seth GSMC and KEM Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
19
|
Wang W, Wang R, Jiang Z, Li H, Zhu Z, Khalid A, Liu D, Pan F. Inhibiting Brd4 alleviated PTSD-like behaviors and fear memory through regulating immediate early genes expression and neuroinflammation in rats. J Neurochem 2021; 158:912-927. [PMID: 34050937 DOI: 10.1111/jnc.15439] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 05/11/2021] [Accepted: 05/18/2021] [Indexed: 12/30/2022]
Abstract
Post-traumatic stress disorder (PTSD) is characterized by depression/anxiety and memory failure, primarily fear memory. According to the reports, neuroinflammation and synaptic plasticity can play a role in the neurophysiological mechanisms underlying PTSD. Bromodomain-containing protein 4 (Brd4) intriguingly affects regulating of inflammatory responses and learning and memory. This study aimed to explore the effect of inhibiting Brd4 on depression/anxiety-like behaviors, spatial and fear memory, and underlying mechanisms in a model of PTSD. Inescapable foot shocks (IFS) with a sound reminder in 6 days were used to induce PTSD-like behaviors which were tested using contextual and cue fear tests, sucrose preference test, open-field test, elevated plus maze test, and Y-maze test. Meanwhile, the Brd4 inhibitor JQ1 was used as an intervention. The results found that IFS induced PTSD-like behaviors and indicated obvious Brd4 expression in microglia of the prefrontal cortex (PFC), hippocampus, and amygdala, pro-inflammatory cytokines over-expression, microglial activation, and nuclear factor-kappa B over-expression in PFC and hippocampus but not in amygdala. Meanwhile, the alterations of immediate early genes (IEGs) were found in PFC, hippocampus, and amygdala. Besides, dendritic spine density was reduced in PFC and hippocampus but was elevated in amygdala of rats with IFS. In addition, treatment with JQ1 significantly reduced freezing time in the contextual and cue fear test, reversed the behavioral impairment, decreased the elevated neuroinflammation, and normalized the alteration in IEGs and dendritic spine densities. The results suggested that Brd4 was involved in IFS-induced PTSD-like behaviors through regulating neuroinflammation, dynamics of IEGs, and synaptic plasticity.
Collapse
Affiliation(s)
- Wei Wang
- Department of Medical Psychology and Ethics, School of Basic Medical Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Rui Wang
- Department of Medical Psychology and Ethics, School of Basic Medical Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Zhijun Jiang
- Department of Medical Psychology and Ethics, School of Basic Medical Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Haonan Li
- Department of Medical Psychology and Ethics, School of Basic Medical Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Zemeng Zhu
- Department of Medical Psychology and Ethics, School of Basic Medical Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Arslan Khalid
- Department of Medical Psychology and Ethics, School of Basic Medical Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Dexiang Liu
- Department of Medical Psychology and Ethics, School of Basic Medical Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Fang Pan
- Department of Medical Psychology and Ethics, School of Basic Medical Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| |
Collapse
|
20
|
Wang YL, Wu HR, Zhang SS, Xiao HL, Yu J, Ma YY, Zhang YD, Liu Q. Catalpol ameliorates depressive-like behaviors in CUMS mice via oxidative stress-mediated NLRP3 inflammasome and neuroinflammation. Transl Psychiatry 2021; 11:353. [PMID: 34103482 PMCID: PMC8187638 DOI: 10.1038/s41398-021-01468-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
The purpose of the present study was to investigate whether catalpol exhibited neuroprotective effects in chronic unpredictable mild stress (CUMS) mice through oxidative stress-mediated nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin-domain-containing 3 (NLRP3) inflammasome and neuroinflammation. Deficits in behavioral tests, including open field test (OFT), forced swim test (FST), and elevated plus-maze test (EPM), were ameliorated following catalpol administration. To study the potential mechanism, western blots, quantitative real-time PCR (qRT-PCR) analysis and immunofluorescence imaging were performed on the hippocampus samples. We found that the defects of behavioral tests induced by CUMS could be reversed by the absence of NLRP3 and NLRP3 inflammasome might be involved in the antidepressant effects of catalpol on CUMS mice. Similar to the NLRP3 inflammasome, the expression of interleukin-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), and inducible nitride oxide synthase (iNOS) were increased after CUMS. The current study demonstrated that catalpol possessed anti-inflammatory effect on CUMS mice and inhibited microglial polarization to the M1 phenotype. In addition, the activity of mitochondrial oxidative stress might be involved in the NLRP3 activation, which was proved by the downregulation of NLRP3, apoptosis-associated speck-like protein containing a CARD (ASC), and cleaved IL-1β, after the administration of mitochondrion-targeted antioxidant peptide SS31. Taken together, we provided evidence that catalpol exhibited antidepressive effects on CUMS mice possibly via the oxidative stress-mediated regulation of NLRP3 and neuroinflammation.
Collapse
Affiliation(s)
- Ya-lin Wang
- grid.8547.e0000 0001 0125 2443Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Hao-ran Wu
- grid.8547.e0000 0001 0125 2443Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Shan-shan Zhang
- grid.8547.e0000 0001 0125 2443Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Hong-lei Xiao
- grid.8547.e0000 0001 0125 2443Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Jin Yu
- grid.8547.e0000 0001 0125 2443Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Yuan-yuan Ma
- grid.8547.e0000 0001 0125 2443State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China
| | - Yao-dong Zhang
- grid.207374.50000 0001 2189 3846Children’s Hospital Affiliated to Zhengzhou University, Henan Neural Development Engineering Research Center, Henan, China
| | - Qiong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, 200032, China.
| |
Collapse
|
21
|
Ahmed A, Misrani A, Tabassum S, Yang L, Long C. Minocycline inhibits sleep deprivation-induced aberrant microglial activation and Keap1-Nrf2 expression in mouse hippocampus. Brain Res Bull 2021; 174:41-52. [PMID: 34087360 DOI: 10.1016/j.brainresbull.2021.05.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 12/26/2022]
Abstract
Sleep deprivation (SD) is a hallmark of modern society and associated with many neuropsychiatric disorders, including depression and anxiety. However, the cellular and molecular mechanisms underlying SD-associated depression and anxiety remain elusive. Does the neuroinflammation play a role in mediating the effects of SD? In this study, we investigated SD-induced cellular and molecular alterations in the hippocampus and asked whether treatment with an anti-inflammatory drug, minocycline, could attenuate these alterations. We found that SD animals exhibit activated microglia and decreased levels of Keap1 and Nrf2 (antioxidant and anti-inflammatory factors) in the hippocampus. In vivo local field potential recordings show decreased theta and beta oscillations, but increased high gamma oscillations, as a result of SD. Behavioral analysis revealed increased immobility time in the forced swim and tail suspension tests, and decreased sucrose intake in SD mice, all indicative of depressive-like behavior. Moreover, open field test and elevated plus maze test results indicated that SD increases anxiety-like behavior. Interestingly, treatment with the microglial modulator minocycline prevented SD-induced microglial activation, restored Keap1 and Nrf2 levels, normalized neuronal oscillations, and alleviated depressive-like and anxiety-like behavior. The present study reveals that microglial activation and Keap1-Nrf2 signaling play a crucial role in SD-induced behavioral alteration, and that minocycline treatment has a protective effect on these alterations.
Collapse
Affiliation(s)
- Adeel Ahmed
- School of Life Sciences, South China Normal University, Guangzhou, 510631, PR China
| | - Afzal Misrani
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, 510006, PR China; South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, 511400, PR China
| | - Sidra Tabassum
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, 510006, PR China; South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, 511400, PR China
| | - Li Yang
- Precise Genome Engineering Center, School of Life Sciences, Guangzhou University, Guangzhou, 510006, PR China
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou, 510631, PR China; South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou, 511400, PR China.
| |
Collapse
|
22
|
A multispecies probiotic accelerates fear extinction and inhibits relapse in mice: Role of microglia. Neuropharmacology 2021; 193:108613. [PMID: 34022177 DOI: 10.1016/j.neuropharm.2021.108613] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/12/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022]
Abstract
The relapse of fear memory remains a clinical challenge in treatment of fear-related disorders. Here we tested the effects and underlying mechanisms of probiotics treatment after fear conditioning on fear extinction. We found that fear conditioning induced synapse loss, microglial activation, and synaptic phagocytosis of activated microglial cells in hippocampal dentate gyrus of mice. And probiotics treatment (1 capsule/day/mice) after fear conditioning for 27 days inhibited these changes, promoted fear extinction, and inhibited the recovery of fear memory even 7 days after extinction. 16S rRNA gene sequencing demonstrated that probiotics supplement after fear conditioning partially normalized fear conditioning-induced dysbiosis of gut microbiota. In addition, we also found that repopulation of microglial cells in fear conditioning mice via PLX3397 treatment promoted long-term extinction of fear memory. Probiotics treatment after fear conditioning inhibited microglial activation and had similar therapeutic effects as the microglial cell repopulation induced by PLX3397 treatment. These data showed that (1) probiotics treatment after fear conditioning might promote long-term fear extinction which could be associated with the mitigation of synaptic pruning of activated microglial cells; (2) probiotics may be applicable as therapeutic strategy to inhibit microglial activation and treat fear-related disorders.
Collapse
|
23
|
Cannabinoid receptor 1 signalling modulates stress susceptibility and microglial responses to chronic social defeat stress. Transl Psychiatry 2021; 11:164. [PMID: 33723234 PMCID: PMC7961142 DOI: 10.1038/s41398-021-01283-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/28/2021] [Accepted: 02/18/2021] [Indexed: 01/31/2023] Open
Abstract
Psychosocial stress is one of the main environmental factors contributing to the development of psychiatric disorders. In humans and rodents, chronic stress is associated with elevated inflammatory responses, indicated by increased numbers of circulating myeloid cells and activation of microglia, the brain-resident immune cells. The endocannabinoid system (ECS) regulates neuronal and endocrine stress responses via the cannabinoid receptor 1 (CB1). CB1-deficient mice (Cnr1-/-) are highly sensitive to stress, but if this involves altered inflammatory responses is not known. To test this, we exposed Cnr1+/+ and Cnr1-/- mice to chronic social defeat stress (CSDS). Cnr1-/- mice were extremely sensitive to a standard protocol of CSDS, indicated by an increased mortality rate. Therefore, a mild CSDS protocol was established, which still induced a behavioural phenotype in susceptible Cnr1-/- mice. These mice also showed altered glucocorticoid levels after mild CSDS, suggesting dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis. Mild CSDS induced weak myelopoiesis in the periphery, but no recruitment of myeloid cells to the brain. In contrast, mild CSDS altered microglial activation marker expression and morphology in Cnr1-/- mice. These microglial changes correlated with the severity of the behavioural phenotype. Furthermore, microglia of Cnr1-/- mice showed increased expression of Fkbp5, an important regulator of glucocorticoid signalling. Overall, the results confirm that CB1 signalling protects the organism from the physical and emotional harm of social stress and implicate endocannabinoid-mediated modulation of microglia in the development of stress-related pathologies.
Collapse
|
24
|
Schultebraucks K, Sijbrandij M, Galatzer-Levy I, Mouthaan J, Olff M, van Zuiden M. Forecasting individual risk for long-term Posttraumatic Stress Disorder in emergency medical settings using biomedical data: A machine learning multicenter cohort study. Neurobiol Stress 2021; 14:100297. [PMID: 33553513 PMCID: PMC7843920 DOI: 10.1016/j.ynstr.2021.100297] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/22/2020] [Accepted: 01/12/2021] [Indexed: 02/06/2023] Open
Abstract
The necessary requirement of a traumatic event preceding the development of Posttraumatic Stress Disorder, theoretically allows for administering preventive and early interventions in the early aftermath of such events. Machine learning models including biomedical data to forecast PTSD outcome after trauma are highly promising for detection of individuals most in need of such interventions. In the current study, machine learning was applied on biomedical data collected within 48 h post-trauma to forecast individual risk for long-term PTSD, using a multinominal approach including the full spectrum of common PTSD symptom courses within one prognostic model for the first time. N = 417 patients (37.2% females; mean age 46.09 ± 15.88) admitted with (suspected) serious injury to two urban Academic Level-1 Trauma Centers were included. Routinely collected biomedical information (endocrine measures, vital signs, pharmacotherapy, demographics, injury and trauma characteristics) upon ED admission and subsequent 48 h was used. Cross-validated multi-nominal classification of longitudinal self-reported symptom severity (IES-R) over 12 months and bimodal classification of clinician-rated PTSD diagnosis (CAPS-IV) at 12 months post-trauma was performed using extreme Gradient Boosting and evaluated on hold-out sets. SHapley Additive exPlanations (SHAP) values were used to explain the derived models in human-interpretable form. Good prediction of longitudinal PTSD symptom trajectories (multiclass AUC = 0.89) and clinician-rated PTSD at 12 months (AUC = 0.89) was achieved. Most relevant prognostic variables to forecast both multinominal and dichotomous PTSD outcomes included acute endocrine and psychophysiological measures and hospital-prescribed pharmacotherapy. Thus, individual risk for long-term PTSD was accurately forecasted from biomedical information routinely collected within 48 h post-trauma. These results facilitate future targeted preventive interventions by enabling future early risk detection and provide further insights into the complex etiology of PTSD.
Collapse
Affiliation(s)
- Katharina Schultebraucks
- Vagelos School of Physicians and Surgeons, Department of Emergency Medicine, Columbia University Medical Center, New York, NY, United States of America; Data Science Institute, Columbia University, New York, New York, USA
| | - Marit Sijbrandij
- Vrije Universiteit, Department of Clinical, Neuro- and Developmental Psychology; Amsterdam Public Health Research Institute, World Health Organization Collaborating Centre for Research and Dissemination of Psychological Interventions, Amsterdam, the Netherlands
| | - Isaac Galatzer-Levy
- Department of Psychiatry, New York University School of Medicine, New York, New York, USA
| | - Joanne Mouthaan
- Department of Clinical Psychology, Institute of Psychology, Faculty of Social and Behavioural Sciences, Leiden University, Leiden, the Netherlands
| | - Miranda Olff
- ARQ National Psychotrauma Centre, Diemen, the Netherlands.,Department of Psychiatry, Amsterdam University Medical Centers, Location Amsterdam Medical Center, University of Amsterdam, Amsterdam Public Health Research Institute and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
| | - Mirjam van Zuiden
- Department of Psychiatry, Amsterdam University Medical Centers, Location Amsterdam Medical Center, University of Amsterdam, Amsterdam Public Health Research Institute and Amsterdam Neuroscience Research Institute, Amsterdam, the Netherlands
| |
Collapse
|
25
|
Li S, Liao Y, Dong Y, Li X, Li J, Cheng Y, Cheng J, Yuan Z. Microglial deletion and inhibition alleviate behavior of post-traumatic stress disorder in mice. J Neuroinflammation 2021; 18:7. [PMID: 33402212 PMCID: PMC7786489 DOI: 10.1186/s12974-020-02069-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 12/23/2020] [Indexed: 12/11/2022] Open
Abstract
Background Alteration of immune status in the central nervous system (CNS) has been implicated in the development of post-traumatic stress disorder (PTSD). However, the nature of overall changes in brain immunocyte landscape in PTSD condition remains unclear. Methods We constructed a mouse PTSD model by electric foot-shocks followed by contextual reminders and verified the PTSD-related symptoms by behavior test (including contextual freezing test, open-field test, and elevated plus maze test). We examined the immunocyte panorama in the brains of the naïve or PTSD mice by using single-cell mass cytometry. Microglia number and morphological changes in the hippocampus, prefrontal cortex, and amygdala were analyzed by histopathological methods. The gene expression changes of those microglia were detected by quantitative real-time PCR. Genetic/pharmacological depletion of microglia or minocycline treatment before foot-shocks exposure was performed to study the role of microglia in PTSD development and progress. Results We found microglia are the major brain immune cells that respond to PTSD. The number of microglia and ratio of microglia to immunocytes was significantly increased on the fifth day of foot-shock exposure. Furthermore, morphological analysis and gene expression profiling revealed temporal patterns of microglial activation in the hippocampus of the PTSD brains. Importantly, we found that genetic/pharmacological depletion of microglia or minocycline treatment before foot-shock exposure alleviated PTSD-associated anxiety and contextual fear. Conclusion Our results demonstrated a critical role for microglial activation in PTSD development and a potential therapeutic strategy for the clinical treatment of PTSD in the form of microglial inhibition. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-020-02069-9.
Collapse
Affiliation(s)
- Shuoshuo Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Yajin Liao
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China
| | - Yuan Dong
- Department of Biochemistry, Medical College, Qingdao University, Qingdao, 266071, Shandong, China
| | - Xiaoheng Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Jun Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China
| | - Jinbo Cheng
- Center on Translational Neuroscience, College of Life & Environmental Science, Minzu University of China, Beijing, 100081, China.
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing, 100850, China. .,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
26
|
Han YY, Jin K, Pan QS, Li B, Wu ZQ, Gan L, Yang L, Long C. Microglial activation in the dorsal striatum participates in anxiety-like behavior in Cyld knockout mice. Brain Behav Immun 2020; 89:326-338. [PMID: 32688031 DOI: 10.1016/j.bbi.2020.07.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/26/2022] Open
Abstract
CYLD lysine 63 deubiquitinase (CYLD), that is mainly involved in immune responses and inflammation, is expressed at high levels in the brain, especially in the dorsal striatum, but its physiological function of CYLD in the brain remains unexplored. The present study investigated the effect of Cyld gene knockout on behavior relevant to the dorsal striatum, such as motor activity and depression-like and anxiety-like behavior. Microglia and the pro-inflammatory cytokines including interleukin (IL)-1 β and tumor necrosis factor (TNF)- α were evaluated in the dorsal striatum to elucidate the underlying mechanism. Cyld knockout (Cyld-/-) mice exhibited anxiety-like behavior, but not motor deficits or depression-like behavior. Microglia were activated and the mRNA levels of IL-1 β and TNF- α were increased in the dorsal striatum of Cyld-/- mice compared to Cyld+/+ mice. The microglial modulator minocycline partially reversed the anxiety-like behavior, microglial activation and increase in IL-1 β and TNF- α mRNA and protein levels in the dorsal striatum of Cyld-/- mice. Collectively, these results suggest that Cyld knockout leading to microglial activation promotes IL-1 β and TNF- α expression and acts as a critical pathway in the pathophysiology of anxiety.
Collapse
Affiliation(s)
- Yuan-Yuan Han
- School of Life Sciences, South China Normal University, Guangzhou 510631, PR China
| | - Kai Jin
- School of Life Sciences, South China Normal University, Guangzhou 510631, PR China
| | - Qi-Sheng Pan
- School of Life Sciences, South China Normal University, Guangzhou 510631, PR China
| | - Bo Li
- School of Life Sciences, South China Normal University, Guangzhou 510631, PR China
| | - Zhuo-Qing Wu
- School of Life Sciences, South China Normal University, Guangzhou 510631, PR China
| | - Lin Gan
- School of Life Sciences, South China Normal University, Guangzhou 510631, PR China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou 510006, PR China.
| | - Cheng Long
- School of Life Sciences, South China Normal University, Guangzhou 510631, PR China; South China Normal University-Panyu Central Hospital Joint Laboratory of Translational Medical Research, Panyu Central Hospital, Guangzhou 511400, PR China.
| |
Collapse
|
27
|
Lycopene ameliorates PTSD-like behaviors in mice and rebalances the neuroinflammatory response and oxidative stress in the brain. Physiol Behav 2020; 224:113026. [DOI: 10.1016/j.physbeh.2020.113026] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/19/2020] [Accepted: 06/22/2020] [Indexed: 12/14/2022]
|
28
|
Zhao M, Wang W, Jiang Z, Zhu Z, Liu D, Pan F. Long-Term Effect of Post-traumatic Stress in Adolescence on Dendrite Development and H3K9me2/BDNF Expression in Male Rat Hippocampus and Prefrontal Cortex. Front Cell Dev Biol 2020; 8:682. [PMID: 32850808 PMCID: PMC7412801 DOI: 10.3389/fcell.2020.00682] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 07/06/2020] [Indexed: 01/10/2023] Open
Abstract
Exposure to a harsh environment in early life increases in the risk of post-traumatic stress disorder (PTSD) of an individual. Brain derived neurotrophic factor (BDNF) plays an important role in neurodevelopment in developmental stages. Both chronic and traumatic stresses induce a decrease in the level of BDNF and reduce neural plasticity, which is linked to the pathogenesis of PTSD. Also, studies have shown that stress alters the epigenetic marker H3K9me2, which can bind to the promoter region of the Bdnf gene and reduce BDNF protein level. However, the long-term effects of traumatic stress during adolescence on H3K9me2, BDNF expression and dendrite development are not well-known. The present study established a model of PTSD in adolescent rats using an inescapable foot shock (IFS) procedure. Anxiety-like behaviors, social interaction behavior and memory function were assessed by the open field test, elevated plus maze test, three-chamber sociability test and Morris water maze test. In addition, neuronal development and H3K9me2/BDNF expression in hippocampus (HIP) and prefrontal cortex (PFC) were evaluated by Golgi staining, western blotting, qRT-PCR analysis and CHIP-qPCR analysis. Additionally, the Unc0642, a small molecule inhibitor of histone methyltransferase (EHMT2) was used for intervention. The results showed that the IFS procedure induced the PTSD-like behaviors in rats, resulted in fewer dendrite branches and shorter dendrite length in CA1 of HIP and PFC, increased H3K9me2 level and decreased BDNF expression in HIP and PFC. Also, although all the changes can persist to adulthood, Unc0642 administration relieved most of alterations. Our study suggests that traumatic stress in adolescence leads to immediate and long-term mental disorders, neuronal morphological changes, lower BDNF level and increased H3K9me2 level in the HIP and PFC, indicating that H3K9me2/BDNF dysfunction plays a key role in pathogenesis of PTSD.
Collapse
Affiliation(s)
- Mingyue Zhao
- Department of Medical Psychology and Medical Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Wang
- Department of Medical Psychology and Medical Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zhijun Jiang
- Department of Medical Psychology and Medical Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zemeng Zhu
- Department of Medical Psychology and Medical Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dexiang Liu
- Department of Medical Psychology and Medical Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fang Pan
- Department of Medical Psychology and Medical Ethics, School of Basic Medicine Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
29
|
Rooney S, Sah A, Unger MS, Kharitonova M, Sartori SB, Schwarzer C, Aigner L, Kettenmann H, Wolf SA, Singewald N. Neuroinflammatory alterations in trait anxiety: modulatory effects of minocycline. Transl Psychiatry 2020; 10:256. [PMID: 32732969 PMCID: PMC7393101 DOI: 10.1038/s41398-020-00942-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 07/07/2020] [Accepted: 07/15/2020] [Indexed: 02/04/2023] Open
Abstract
High trait anxiety is a substantial risk factor for developing anxiety disorders and depression. While neuroinflammation has been identified to contribute to stress-induced anxiety, little is known about potential dysregulation in the neuroinflammatory system of genetically determined pathological anxiety or high trait anxiety individuals. We report microglial alterations in various brain regions in a mouse model of high trait anxiety (HAB). In particular, the dentate gyrus (DG) of the hippocampus of HABs exhibited enhanced density and average cell area of Iba1+, and density of phagocytic (CD68+/Iba1+) microglia compared to normal anxiety (NAB) controls. Minocycline was used to assess the capacity of a putative microglia 'inhibitor' in modulating hyperanxiety behavior of HABs. Chronic oral minocycline indeed reduced HAB hyperanxiety, which was associated with significant decreases in Iba1+ and CD68+Iba1+ cell densities in the DG. Addressing causality, it was demonstrated that longer (10 days), but not shorter (5 days), periods of minocycline microinfusions locally into the DG of HAB reduced Iba-1+ cell density and attenuated hyperanxiety-related behavior, indicating that neuroinflammation in the DG is at least partially involved in the maintenance of pathological anxiety. The present data reveal evidence of disturbances in the microglial system of individuals with high trait anxiety. Minocycline attenuated HAB hyperanxiety, likely by modulation of microglial activity within the DG. Thus, the present data suggest that drugs with microglia-targeted anti-inflammatory properties could be promising as novel alternative or complimentary anxiolytic therapeutic approaches in specific subgroups of individuals genetically predisposed to hyperanxiety.
Collapse
Affiliation(s)
- Sinead Rooney
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Anupam Sah
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Michael S Unger
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Maria Kharitonova
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Simone B Sartori
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria
| | - Christoph Schwarzer
- Department of Pharmacology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Paracelsus Medical University, Salzburg, Austria
| | - Helmut Kettenmann
- Department of Cellular Neurosciences, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Susanne A Wolf
- Department of Cellular Neurosciences, Max Delbrück Center for Molecular Medicine, Berlin, Germany
- Department of Ophthalmology, Charité Universitätsmedizin, Berlin, Germany
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and Center for Molecular Biosciences Innsbruck (CMBI), University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
30
|
Yang L, Wang J, Wang D, Hu G, Liu Z, Yan D, Serikuly N, Alpyshov ET, Demin KA, Strekalova T, de Abreu MS, Song C, Kalueff AV. Delayed behavioral and genomic responses to acute combined stress in zebrafish, potentially relevant to PTSD and other stress-related disorders: Focus on neuroglia, neuroinflammation, apoptosis and epigenetic modulation. Behav Brain Res 2020; 389:112644. [PMID: 32344037 DOI: 10.1016/j.bbr.2020.112644] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/22/2020] [Accepted: 04/05/2020] [Indexed: 12/30/2022]
Abstract
Stress is a common trigger of stress-related illnesses, such as anxiety, phobias, depression and post-traumatic stress disorder (PTSD). Various animal models successfully reproduce core behaviors of these clinical conditions. Here, we develop a novel zebrafish model of stress (potentially relevant to human stress-related disorders), based on delayed persistent behavioral, endocrine and genomic responses to an acute severe 'combined' stressor. Specifically, one week after adult zebrafish were exposed to a complex combined 90-min stress, we assessed their behaviors in the novel tank and the light-dark box tests, as well as whole-body cortisol and brain gene expression, focusing on genomic biomarkers of microglia, astrocytes, neuroinflammation, apoptosis and epigenetic modulation. Overall, stressed fish displayed persistent anxiety-like behavior, elevated whole-body cortisol, as well as upregulated brain mRNA expression of genes encoding the glucocorticoid receptor, neurotrophin BDNF and its receptors (TrkB and P75), CD11b (a general microglial biomarker), COX-2 (an M1-microglial biomarker), CD206 (an M2-microglial biomarker), GFAP (a general astrocytal biomarker), C3 (an A1-astrocytal biomarker), S100α10 (an A2-astrocytal biomarker), as well as pro-inflammatory cytokines IL-6, IL-1β, IFN-γ and TNF-α. Stress exposure also persistently upregulated the brain expression of several key apoptotic (Bax, Caspase-3, Bcl-2) and epigenetic genes (DNMT3a, DNMT3b, HAT1, HDAC4) in these fish. Collectively, the present model not only successfully recapitulates lasting behavioral and endocrine symptoms of clinical stress-related disorders, but also implicates changes in neuroglia, neuroinflammation, apoptosis and epigenetic modulation in long-term effects of stress pathogenesis in vivo.
Collapse
Affiliation(s)
- LongEn Yang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Jingtao Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Dongmei Wang
- School of Pharmacy, Southwest University, Chongqing, China
| | - Guojun Hu
- School of Pharmacy, Southwest University, Chongqing, China
| | - ZiYuan Liu
- School of Pharmacy, Southwest University, Chongqing, China
| | - Dongni Yan
- School of Pharmacy, Southwest University, Chongqing, China
| | - Nazar Serikuly
- School of Pharmacy, Southwest University, Chongqing, China
| | - Erik T Alpyshov
- School of Pharmacy, Southwest University, Chongqing, China; Granov Russian Scientific Center of Radiology and Surgical Technologies, Ministry of Healthcare of Russian Federation, St. Petersburg, Russia; Scientific Research Institute of Physiology and Basic Medicine, Novosibirsk, Russia; Institute of Medicine and Psychology, Novosibirsk State University, Novosibirsk, Russia
| | - Konstantin A Demin
- Institute of Experimental Medicine, Almazov Medical Research Center, Ministy of Healthcare of Russian Federation, St. Petersburg, Russia
| | - Tatyana Strekalova
- I.M. Sechenov First Moscow State Medical University, Moscow, Russia; Maastricht University, Maastricht, the Netherlands; Research Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Murilo S de Abreu
- Bioscience Institute, University of Passo Fundo, Passo Fundo, Brazil
| | - Cai Song
- Institute for Marine Drugs and Nutrition, Marine Medicine Development Center, Shenzhen Institute, Guangdong Ocean University, Zhanjiang, China
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China; Ural Federal University, Ekaterinburg, Russia.
| |
Collapse
|
31
|
Camargos QM, Silva BC, Silva DG, Toscano ECDB, Oliveira BDS, Bellozi PMQ, Jardim BLDO, Vieira ÉLM, de Oliveira ACP, Sousa LP, Teixeira AL, de Miranda AS, Rachid MA. Minocycline treatment prevents depression and anxiety-like behaviors and promotes neuroprotection after experimental ischemic stroke. Brain Res Bull 2019; 155:1-10. [PMID: 31756420 DOI: 10.1016/j.brainresbull.2019.11.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/28/2019] [Accepted: 11/17/2019] [Indexed: 11/26/2022]
Abstract
Depression and anxiety have been reported as the major neuropsychiatric consequences following stroke. Minocycline, a neuroprotective drug has minimized depressive symptoms in patients with major depressive disorders and anxiety-like symptoms. In addition, minocycline demonstrated efficacy and seemed a promising neuroprotective agent in acute stroke patients. The present studied evaluated the effects of minocycline treatment on the depression and anxiety-like behaviors, brain damage and expression of inflammatory and neuroprotective mediators after transient global cerebral ischemia in C57BL/6 mice. Brain ischemia was induced by bilateral occlusion of the common carotids (BCCAo) for 25 min and subsequent reperfusion. Sham and BCCAo animals received minocycline at a dose of 30 mg/kg by intraperitoneal injection during 14 days. The locomotor activity, depression and anxiety-like behaviors were assessed by open field, forced swim and elevated plus maze tests, respectively. Then, the brains were removed and processed to evaluate brain damage by histological and morphometric analysis, hippocampal neurodegeneration using Fluoro-Jade C histochemistry, microglial activity using iba-1 immunohistochemistry, brain levels of TNF, IFN-γ, IL-6, IL-10, IL-12p70 and CCL2 by CBA, CX3CL1 and BDNF by ELISA assays. The animals developed depression and anxiety-like behaviors post-stroke and minocycline treatment prevented those neurobehavioral changes. Moreover, minocycline-treated BCCAo animals showed less intense brain damage in the cerebral cortex, brainstem and cerebellum as well as significantly reduced hippocampal neurodegeneration. BCCAo groups exhibited up-regulation of some cytokines at day 14 after ischemia and brain levels of CX3CL1 and BDNF remained unaltered. Our data indicate that the depression and anxiety-like behavioral improvements promoted by minocycline treatment might be related to its neuroprotective effect after brain ischemia in mice.
Collapse
Affiliation(s)
- Quezya Mendes Camargos
- Laboratório de Patologia Celular e Molecular do Departamento de Patologia Geral, Instituto de Ciências Biológicas, UFMG, Brazil
| | - Bruno Costa Silva
- Laboratório de Patologia Celular e Molecular do Departamento de Patologia Geral, Instituto de Ciências Biológicas, UFMG, Brazil
| | - Daniele Gonçalves Silva
- Laboratório de Patologia Celular e Molecular do Departamento de Patologia Geral, Instituto de Ciências Biológicas, UFMG, Brazil
| | | | | | | | | | - Érica Leandro Marciano Vieira
- Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, MG, Brazil
| | | | - Lirlândia Pires Sousa
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, UFMG, Brazil
| | - Antônio Lúcio Teixeira
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, School of Medicine, University of Texas Health Science Center at Houston, TX, USA
| | - Aline Silva de Miranda
- Departamento de Morfologia, Instituto de Ciências Biológicas, UFMG, Brazil; Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, MG, Brazil.
| | - Milene Alvarenga Rachid
- Laboratório de Patologia Celular e Molecular do Departamento de Patologia Geral, Instituto de Ciências Biológicas, UFMG, Brazil; Laboratório Interdisciplinar de Investigação Médica, Faculdade de Medicina, Universidade Federal de Minas Gerais, MG, Brazil.
| |
Collapse
|
32
|
Shin J, Yin Y, Kim DK, Lee SY, Lee W, Kang JW, Kim DW, Hong J. Foxp3 plasmid-encapsulated PLGA nanoparticles attenuate pain behavior in rats with spinal nerve ligation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 18:90-100. [DOI: 10.1016/j.nano.2019.02.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/11/2019] [Accepted: 02/24/2019] [Indexed: 11/29/2022]
|
33
|
Johnson JD, Barnard DF, Kulp AC, Mehta DM. Neuroendocrine Regulation of Brain Cytokines After Psychological Stress. J Endocr Soc 2019; 3:1302-1320. [PMID: 31259292 PMCID: PMC6595533 DOI: 10.1210/js.2019-00053] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 05/06/2019] [Indexed: 12/22/2022] Open
Abstract
There is growing evidence that stress-induced brain cytokines are important in the etiology of depression and anxiety. Here, we review how the neuroendocrine responses to psychological stressors affect the immediate and long-term regulation of inflammatory cytokines within the brain and highlight how the regulation changes across time with repeated stress exposure. In doing so, we report on the percentage of studies in the literature that observed increases in either IL-1β, TNF-α, or IL-6 within the hypothalamus, hippocampus, or prefrontal cortex after either acute or chronic stress exposure. The key takeaway is that catecholamines and glucocorticoids play critical roles in the regulation of brain cytokines after psychological stress exposure. Central catecholamines stimulate the release of IL-1β from microglia, which is a key factor in the further activation of microglia and recruitment of monocytes into the brain. Meanwhile, the acute elevation of glucocorticoids inhibits the production of brain cytokines via two mechanisms: the suppression of noradrenergic locus coeruleus neurons and inhibition of the NFκB signaling pathway. However, glucocorticoids and peripheral catecholamines facilitate inflammatory responses to future stimuli by stimulating monocytes to leave the bone marrow, downregulating inhibitory receptors on microglia, and priming inflammatory responses mediated by peripheral monocytes or macrophages. The activation of microglia and the elevation of peripheral glucocorticoid and catecholamine levels are both necessary during times of stress exposure for the development of psychopathologies.
Collapse
Affiliation(s)
- John D Johnson
- Kent State University, Biological Sciences Department, School of Biomedical Sciences, Kent, Ohio
| | - David F Barnard
- Kent State University, Biological Sciences Department, School of Biomedical Sciences, Kent, Ohio
| | - Adam C Kulp
- Kent State University, Biological Sciences Department, School of Biomedical Sciences, Kent, Ohio
| | - Devanshi M Mehta
- Kent State University, Biological Sciences Department, School of Biomedical Sciences, Kent, Ohio
| |
Collapse
|
34
|
Abizaid A, Merali Z, Anisman H. Cannabis: A potential efficacious intervention for PTSD or simply snake oil? J Psychiatry Neurosci 2019; 44:75-78. [PMID: 30810022 PMCID: PMC6397040 DOI: 10.1503/jpn.190021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Alfonso Abizaid
- From the Department of Neuroscience, Carleton University (Abizaid, Anisman); and the Royal’s Institute of Mental Health (Merali), Ottawa, Ont., Canada
| | - Zul Merali
- From the Department of Neuroscience, Carleton University (Abizaid, Anisman); and the Royal’s Institute of Mental Health (Merali), Ottawa, Ont., Canada
| | - Hymie Anisman
- From the Department of Neuroscience, Carleton University (Abizaid, Anisman); and the Royal’s Institute of Mental Health (Merali), Ottawa, Ont., Canada
| |
Collapse
|