1
|
Tao L, Li XX, Tu XR, Liu R, Xu JW, Lv YL, Yao YY. Hippocampal Crhr1 conditional gene knockout ameliorated the depression-like behavior and pathological damage in male offspring mice caused by chronic stress during pregnancy. Behav Brain Res 2024; 472:115139. [PMID: 38969017 DOI: 10.1016/j.bbr.2024.115139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/24/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Numerous studies have demonstrated that chronic stress during pregnancy (CSDP) can induce depression and hippocampal damage in offspring. It has also been observed that high levels of corticotropin-releasing hormone (CRH) can damage hippocampal neurons, and intraperitoneal injection of a corticotropin releasing hormone receptor 1 (CRHR1) antagonist decreases depression-like behavior and hippocampal neuronal damage in a mouse depression model. However, whether CSDP causes hippocampal damage and depression in offspring through the interaction of CRH and hippocampal CRHR1 remains unknown and warrants further investigation. Therefore, hippocampal Crhr1 conditional gene knockout mice and C57/BL6J mice were used to study these questions. Depression-related indexs in male offspring mice were examined using the forced swim test (FST), sucrose preference test (SPT), tail suspension test (TST) and open field test (OFT). Serum CRH levels were measured by enzyme-linked immunosorbent assay (ELISA). Golgi-Cox staining was used to examine the morphological changes of hippocampal neuronal dendrites. Neuronal apoptosis in the hippocampal CA3 regions was detected by terminal deoxynucleotidy transferase dUTP nick end labeling (TUNEL) staining. The levels of mammalian target of rapamycin (mTOR), phosphorylated mTOR (p-mTOR) and protein kinase B (AKT) proteins were measured by Western blot analysis. This study showed that CSDP induces depression-like behavior, hippocampal neuronal dendrite damage and apoptosis in male offspring mice. Conditional gene knockout of hippocampal Crhr1 in mice reduced CSDP-induced depression-like behavior, hippocampal neuronal dendrite damage and apoptosis in male offspring, and counteracted the CSDP-induced decreased expression of p-Akt and mTOR activity in male offspring hippocampus. These findings demonstrated that CSDP might inhibit the Akt/mTOR pathway by increasing the levels of CRH, leading to increased CRH-mediated activation of hippocampal CRHR1, thereby inducing synaptic impairment and apoptosis in hippocampal neurons, which in turn leads to depression-like behavior in offspring.
Collapse
Affiliation(s)
- Long Tao
- School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Nanjing Jiangning District Center for Disease Control and Prevention, Nanjing 211100, China
| | - Xiao-Xiao Li
- School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Xin-Ru Tu
- School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Rui Liu
- School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Jia-Wen Xu
- School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yi-Li Lv
- Anhui Provincial Center for Disease Control and Prevention, Hefei, Anhui 230601, China
| | - Yu-You Yao
- School of Public Health, Anhui Medical University, Hefei, Anhui 230032, China; Anhui Provincial Key Laboratory of Population Health & Aristogenics, Hefei, Anhui 230032, China.
| |
Collapse
|
2
|
Wang Z, Lai C, Shen B, Li B, Chen J, Shen X, Huang Z, Yang C, Gao Y. Effects of Evodiamine on Behavior and Hippocampal Neurons through Inhibition of Angiotensin-Converting Enzyme and Modulation of the Renin Angiotensin Pathway in a Mouse Model of Post-Traumatic Stress Disorder. Nutrients 2024; 16:1957. [PMID: 38931311 PMCID: PMC11207023 DOI: 10.3390/nu16121957] [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: 04/25/2024] [Revised: 06/12/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Post-traumatic stress disorder (PTSD) is a persistent psychiatric condition that arises following exposure to traumatic events such as warfare, natural disasters, or other catastrophic incidents, typically characterized by heightened anxiety, depressive symptoms, and cognitive dysfunction. In this study, animals subjected to single prolonged stress (SPS) were administered evodiamine (EVO) and compared to a positive control group receiving sertraline. The animals were then assessed for alterations in anxiety, depression, and cognitive function. Histological analysis was conducted to examine neuronal changes in the hippocampus. In order to predict the core targets and related mechanisms of evodiamine intervention in PTSD, network pharmacology was used. The metabolic markers pre- and post-drug administration were identified using nontargeted serum metabolomics techniques, and the intersecting Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were screened. Finally, the core targets were validated through molecular docking, enzyme-linked immunosorbent assays, and immunofluorescence staining to confirm the anti-PTSD effects and mechanisms of these targets. As well as improving cognitive impairment, evodiamine reversed anxiety- and depression-like behaviors. It also inhibited the reduction in the number of hippocampal neuronal cells and Nissl bodies in SPS mice inhibited angiotensin converting enzyme (ACE) levels in the hippocampus of SPS mice, and modulated the renin angiotensin pathway and its associated serum metabolites in brain tissue. Evodiamine shows promise as a potential candidate for alleviating the symptoms of post-traumatic stress disorder.
Collapse
Affiliation(s)
- Zhixing Wang
- Medical College, Qinghai University, Xining 810016, China; (Z.W.); (C.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (B.S.); (B.L.); (J.C.); (X.S.); (C.Y.)
| | - Chengcai Lai
- Medical College, Qinghai University, Xining 810016, China; (Z.W.); (C.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (B.S.); (B.L.); (J.C.); (X.S.); (C.Y.)
| | - Baoying Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (B.S.); (B.L.); (J.C.); (X.S.); (C.Y.)
| | - Bowei Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (B.S.); (B.L.); (J.C.); (X.S.); (C.Y.)
| | - Junru Chen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (B.S.); (B.L.); (J.C.); (X.S.); (C.Y.)
| | - Xin Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (B.S.); (B.L.); (J.C.); (X.S.); (C.Y.)
| | - Zhengping Huang
- Department of Neurology, Fujian Medical University, Quanzhou 362000, China;
| | - Chunqi Yang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (B.S.); (B.L.); (J.C.); (X.S.); (C.Y.)
| | - Yue Gao
- Medical College, Qinghai University, Xining 810016, China; (Z.W.); (C.L.)
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing 100850, China; (B.S.); (B.L.); (J.C.); (X.S.); (C.Y.)
| |
Collapse
|
3
|
Li X, Liu N, Wu D, Li SC, Wang Q, Zhang DW, Song LL, Huang M, Chen X, Li W. Hippocampal transcriptomic analyses reveal the potential antiapoptotic mechanism of a novel anticonvulsant agent Q808 on pentylenetetrazol-induced epilepsy in rats. Biomed Pharmacother 2024; 175:116746. [PMID: 38739991 DOI: 10.1016/j.biopha.2024.116746] [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: 01/29/2024] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/16/2024] Open
Abstract
Brain apoptosis is one of the main causes of epileptogenesis. The antiapoptotic effect and potential mechanism of Q808, an innovative anticonvulsant chemical, have never been reported. In this study, the seizure stage and latency to reach stage 2 of pentylenetetrazol (PTZ) seizure rat model treated with Q808 were investigated. The morphological change and neuronal apoptosis in the hippocampus were detected by hematoxylin and eosin (HE) and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining, respectively. The hippocampal transcriptomic changes were observed using RNA sequencing (RNA-seq). The expression levels of hub genes were verified by quantitative reverse-transcription PCR (qRT-PCR). Results revealed that Q808 could allay the seizure score and prolong the stage 2 latency in seizure rats. The morphological changes of neurons and the number of apoptotic cells in the DG area were diminished by Q808 treatment. RNA-seq analysis revealed eight hub genes, including Map2k3, Nfs1, Chchd4, Hdac6, Siglec5, Slc35d3, Entpd1, and LOC103690108, and nine hub pathways among the control, PTZ, and Q808 groups. Hub gene Nfs1 was involved in the hub pathway sulfur relay system, and Map2k3 was involved in the eight remaining hub pathways, including Amyotrophic lateral sclerosis, Cellular senescence, Fc epsilon RI signaling pathway, GnRH signaling pathway, Influenza A, Rap1 signaling pathway, TNF signaling pathway, and Toll-like receptor signaling pathway. qRT-PCR confirmed that the mRNA levels of these hub genes were consistent with the RNA-seq results. Our findings might contribute to further studies exploring the new apoptosis mechanism and actions of Q808.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Ning Liu
- Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin 130015, China
| | - Di Wu
- Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin 130015, China
| | - Shu Chang Li
- Jilin Cancer Hospital, ChangChun, Jilin 130012, China
| | - Qing Wang
- Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin 130015, China
| | - Dian-Wen Zhang
- Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin 130015, China
| | - Lian-Lian Song
- Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin 130015, China
| | - Min Huang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China.
| | - Wei Li
- Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin 130015, China.
| |
Collapse
|
4
|
Balan I, Boero G, Chéry SL, McFarland MH, Lopez AG, Morrow AL. Neuroactive Steroids, Toll-like Receptors, and Neuroimmune Regulation: Insights into Their Impact on Neuropsychiatric Disorders. Life (Basel) 2024; 14:582. [PMID: 38792602 PMCID: PMC11122352 DOI: 10.3390/life14050582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/18/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Pregnane neuroactive steroids, notably allopregnanolone and pregnenolone, exhibit efficacy in mitigating inflammatory signals triggered by toll-like receptor (TLR) activation, thus attenuating the production of inflammatory factors. Clinical studies highlight their therapeutic potential, particularly in conditions like postpartum depression (PPD), where the FDA-approved compound brexanolone, an intravenous formulation of allopregnanolone, effectively suppresses TLR-mediated inflammatory pathways, predicting symptom improvement. Additionally, pregnane neurosteroids exhibit trophic and anti-inflammatory properties, stimulating the production of vital trophic proteins and anti-inflammatory factors. Androstane neuroactive steroids, including estrogens and androgens, along with dehydroepiandrosterone (DHEA), display diverse effects on TLR expression and activation. Notably, androstenediol (ADIOL), an androstane neurosteroid, emerges as a potent anti-inflammatory agent, promising for therapeutic interventions. The dysregulation of immune responses via TLR signaling alongside reduced levels of endogenous neurosteroids significantly contributes to symptom severity across various neuropsychiatric disorders. Neuroactive steroids, such as allopregnanolone, demonstrate efficacy in alleviating symptoms of various neuropsychiatric disorders and modulating neuroimmune responses, offering potential intervention avenues. This review emphasizes the significant therapeutic potential of neuroactive steroids in modulating TLR signaling pathways, particularly in addressing inflammatory processes associated with neuropsychiatric disorders. It advances our understanding of the complex interplay between neuroactive steroids and immune responses, paving the way for personalized treatment strategies tailored to individual needs and providing insights for future research aimed at unraveling the intricacies of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Irina Balan
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Giorgia Boero
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC 27710, USA;
| | - Samantha Lucenell Chéry
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minna H. McFarland
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Neuroscience Curriculum, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alejandro G. Lopez
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - A. Leslie Morrow
- Bowles Center for Alcohol Studies, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (I.B.); (S.L.C.); (M.H.M.); (A.G.L.)
- Department of Psychiatry, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
5
|
Jyothi AK, Thotakura B, Priyadarshini C S, Subramanian M, Rajila HS. Evidence of alterations in the learning and memory in offspring of stress-induced male rats. J Basic Clin Physiol Pharmacol 2023; 34:473-487. [PMID: 34428362 DOI: 10.1515/jbcpp-2020-0183] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 04/20/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES There is extensive data pointing to offspring outcomes related to maternal life incidents, but there is less research concerning the association between paternal life events and progeny brain development and behaviour. As male gametogenesis is a continuous process, the incidences happening in life can modify the epigenetic regulation, altering the offspring's development and behaviour. The present study evaluates the effects of paternal stress during different life periods on their offspring's learning ability, memory, morphological and biochemical changes in the prefrontal cortex and hippocampus in the rat model. METHODS Four weeks' old male rats were subjected to five variable stressors at the rate of one per day. Stress received male rats were bred with naive female rats for 1 to 3 nights. The offspring's learning and memory were assessed by the Morris water maze test and automated Y maze. Following behavioural studies, offspring were euthanized to examine global DNA methylation, neurotransmitter levels, namely acetylcholine, glutamate in the hippocampus and frontal cortex. RESULTS The offspring of stress-induced animals exhibited a delay in acquiring learning and defect in memory and altered global DNA methylation in the hippocampus (p=0.000124). There was significant reduction of acetylcholine and glutamate levels in hippocampus (p=0.000018, p=0.00001, respectively) and in prefrontal cortex (p=0.00001, p=0.00001, respectively). HPA axis of offspring was altered considerably (p=0.00001). The histomorphometry of the prefrontal cortex and different hippocampal regions revealed a statistically significant (p<0.05) reduction in neuronal numbers in the offspring of stressed animals compared to that of control. These impacts were markedly high in the offspring of fathers who received stress during both pubertal and adult periods. CONCLUSIONS The findings of this study demonstrate that paternal stress can impact offspring learning and memory.
Collapse
Affiliation(s)
- Ashok Kumar Jyothi
- Department of Anatomy, Basaveshwara Medical College and Hospital, Chitradurga, Karnataka, India
- Department of Anatomy, Tagore Medical College & Hospital, Chennai, Tamil Nadu, India
| | - Balaji Thotakura
- Department of Anatomy, Chettinad Academy of Research and Education, Chennai, Chennai, Tamil Nadu, India
| | | | - Manickam Subramanian
- Department of Anatomy, Chettinad Academy of Research and Education, Chennai, Tamil Nadu, India
| | | |
Collapse
|
6
|
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: 4.0] [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
|
7
|
Gu T, Xu C, Meng X, Gao D, Jiang G, Yin A, Liu Q, Zhang L. Sevoflurane Preconditioning Alleviates Posttraumatic Stress Disorder-Induced Apoptosis in the Hippocampus via the EZH2-Regulated Akt/mTOR Axis and Improves Synaptic Plasticity. J Mol Neurosci 2023; 73:225-236. [PMID: 36930428 DOI: 10.1007/s12031-023-02114-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a persistent and severe psychological and mental disorder resulting from experiences of serious trauma or stress and is suffered by many individuals. Previous studies have shown that pretreatment with sevoflurane is efficient in reducing the incidence of PTSD. However, we require a more comprehensive understanding of the specific mechanisms by which sevoflurane works. Enhancer of zeste homolog 2 (EZH2) has been reported to be regulated by sevoflurane, and to improve patient cognition. In this study, we aimed to explore the mechanisms of sevoflurane and the role of EZH2 in PTSD cases. We explored the effects of sevoflurane and EPZ-6438 (inhibitor of EZH2) on rat behavior, followed by an investigation of EZH2 mRNA and protein expression. The effects of sevoflurane and EZH2 on neuronal survival were assessed by western blotting and TUNEL staining, while western blotting was used to examine the expression of PSD95 and the AKT/mTOR proteins. Sevoflurane preconditioning restored EZH2 expression and significantly inhibited apoptosis by regulating phosphorylation of the AKT/mTOR pathway. Synaptic plasticity was also significantly improved. These results suggest that pretreatment with sevoflurane could play an important role in PTSD prevention by regulating EZH2 expression.
Collapse
Affiliation(s)
- Tingting Gu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chang Xu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaozhou Meng
- Department of Anesthesiology, Jinling Hospital, Medical College of Nanjing Medical University, Nanjing, China
| | - Dapeng Gao
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guanghao Jiang
- Department of Anesthesiology, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, China
| | - Anqi Yin
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qingzhen Liu
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China.
| | - Lidong Zhang
- Department of Anesthesiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, China. .,Department of Anesthesiology, Jinling Hospital, Medical College of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Thakkar B, Acevedo EO. BDNF as a biomarker for neuropathic pain: Consideration of mechanisms of action and associated measurement challenges. Brain Behav 2023; 13:e2903. [PMID: 36722793 PMCID: PMC10013954 DOI: 10.1002/brb3.2903] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 01/08/2023] [Accepted: 01/16/2023] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION The primary objective of this paper is to (1) provide a summary of human studies that have used brain derived neurotrophic factor (BDNF) as a biomarker, (2) review animal studies that help to elucidate the mechanistic involvement of BDNF in the development and maintenance of neuropathic pain (NP), and (3) provide a critique of the existing measurement techniques to highlight the limitations of the methods utilized to quantify BDNF in different biofluids in the blood (i.e., serum and plasma) with the intention of presenting a case for the most reliable and valid technique. Lastly, this review also explores potential moderators that can influence the measurement of BDNF and provides recommendations to standardize its quantification to reduce the inconsistencies across studies. METHODS In this manuscript we examined the literature on BDNF, focusing on its role as a biomarker, its mechanism of action in NP, and critically analyzed its measurement in serum and plasma to identify factors that contribute to the discrepancy in results between plasma and serum BDNF values. RESULTS A large heterogenous literature was reviewed that detailed BDNF's utility as a potential biomarker in healthy volunteers, patients with chronic pain, and patients with neuropsychiatric disorders but demonstrated inconsistent findings. The literature provides insight into the mechanism of action of BDNF at different levels of the central nervous system using animal studies. We identified multiple factors that influence the measurement of BDNF in serum and plasma and based on current evidence, we recommend assessing serum BDNF levels to quantify peripheral BDNF as they are more stable and sensitive to changes than plasma BDNF. CONCLUSION Although mechanistic studies clearly explain the role of BDNF, results from human studies are inconsistent. More studies are needed to evaluate the methodological challenges in using serum BDNF as a biomarker in NP.
Collapse
Affiliation(s)
- Bhushan Thakkar
- Department of Physical Therapy, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Edmund O Acevedo
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
9
|
Teal LB, Ingram SM, Bubser M, McClure E, Jones CK. The Evolving Role of Animal Models in the Discovery and Development of Novel Treatments for Psychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2023; 30:37-99. [PMID: 36928846 DOI: 10.1007/978-3-031-21054-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Historically, animal models have been routinely used in the characterization of novel chemical entities (NCEs) for various psychiatric disorders. Animal models have been essential in the in vivo validation of novel drug targets, establishment of lead compound pharmacokinetic to pharmacodynamic relationships, optimization of lead compounds through preclinical candidate selection, and development of translational measures of target occupancy and functional target engagement. Yet, with decades of multiple NCE failures in Phase II and III efficacy trials for different psychiatric disorders, the utility and value of animal models in the drug discovery process have come under intense scrutiny along with the widespread withdrawal of the pharmaceutical industry from psychiatric drug discovery. More recently, the development and utilization of animal models for the discovery of psychiatric NCEs has undergone a dynamic evolution with the application of the Research Domain Criteria (RDoC) framework for better design of preclinical to clinical translational studies combined with innovative genetic, neural circuitry-based, and automated testing technologies. In this chapter, the authors will discuss this evolving role of animal models for improving the different stages of the discovery and development in the identification of next generation treatments for psychiatric disorders.
Collapse
Affiliation(s)
- Laura B Teal
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Shalonda M Ingram
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Michael Bubser
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA
| | - Elliott McClure
- College of Pharmacy and Health Sciences, Lipscomb University, Nashville, TN, USA
| | - Carrie K Jones
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA.
- Warren Center for Neuroscience Drug Discovery, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
10
|
Huang J, Xu F, Yang L, Tuolihong L, Wang X, Du Z, Zhang Y, Yin X, Li Y, Lu K, Wang W. Involvement of the GABAergic system in PTSD and its therapeutic significance. Front Mol Neurosci 2023; 16:1052288. [PMID: 36818657 PMCID: PMC9928765 DOI: 10.3389/fnmol.2023.1052288] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
The neurobiological mechanism of post-traumatic stress disorder (PTSD) is poorly understood. The inhibition of GABA neurons, especially in the amygdala, is crucial for the precise regulation of the consolidation, expression, and extinction of fear conditioning. The GABAergic system is involved in the pathophysiological process of PTSD, with several studies demonstrating that the function of the GABAergic system decreases in PTSD patients. This paper reviews the preclinical and clinical studies, neuroimaging techniques, and pharmacological studies of the GABAergic system in PTSD and summarizes the role of the GABAergic system in PTSD. Understanding the role of the GABAergic system in PTSD and searching for new drug targets will be helpful in the treatment of PTSD.
Collapse
Affiliation(s)
| | - Fei Xu
- Department of Psychiatry of School of Public Health, Southern Medical University, Guangzhou, China
| | - Liping Yang
- Department of Applied Psychology of School of Public Health, Southern Medical University, Guangzhou, China
| | - Lina Tuolihong
- Department of Basic Medical of Basic Medical College, Southern Medical University, Guangzhou, China
| | - Xiaoyu Wang
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Zibo Du
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Yiqi Zhang
- Eight-Year Master's and Doctoral Program in Clinical Medicine of the First Clinical Medical College, Southern Medical University, Guangzhou, China
| | - Xuanlin Yin
- Department of Basic Medical of Basic Medical College, Southern Medical University, Guangzhou, China
| | - Yingjun Li
- Department of Medical Laboratory Science, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Kangrong Lu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Southern Medical University, Guangzhou, China
| | - Wanshan Wang
- Department of Laboratory Animal Center, Southern Medical University, Guangzhou, China
| |
Collapse
|
11
|
Lu D, Sapkota Y, Valdimarsdóttir UA, Koenen KC, Li N, Leisenring WM, Gibson T, Wilson CL, Robison LL, Hudson MM, Armstrong GT, Krull KR, Yasui Y, Bhatia S, Recklitis CJ. Genome-wide association study of posttraumatic stress disorder among childhood cancer survivors: results from the Childhood Cancer Survivor Study and the St. Jude Lifetime Cohort. Transl Psychiatry 2022; 12:342. [PMID: 35999196 PMCID: PMC9399128 DOI: 10.1038/s41398-022-02110-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 11/19/2022] Open
Abstract
Genetic influence shapes who develops posttraumatic stress disorder (PTSD) after traumatic events. However, the genetic variants identified for PTSD may in fact be associated with traumatic exposures (e.g., interpersonal violence), which appear heritable as well. Childhood cancer survivors (CCS) are at risk for PTSD, but genetic influences affecting cancer are unlikely to overlap with those affecting PTSD. This offers a unique opportunity to identify variants specific to PTSD risk. In a genome-wide association study (GWAS), 3984 5-year survivors of childhood cancer of European-ancestry from the Childhood Cancer Survivor Study (CCSS) were evaluated for discovery and 1467 survivors from the St. Jude Lifetime (SJLIFE) cohort for replication. Childhood cancer-related PTSD symptoms were assessed using the Posttraumatic Stress Diagnostic Scale in CCSS. GWAS was performed in CCSS using logistic regression and lead markers were replicated/meta-analyzed using SJLIFE. Cross-associations of identified loci were examined between CCS and the general population. PTSD criteria were met for 671 participants in CCSS and 161 in SJLIFE. Locus 10q26.3 was significantly associated with PTSD (rs34713356, functionally mapped to ECHS1, P = 1.36 × 10-8, OR 1.57), and was replicated in SJLIFE (P = 0.047, OR 1.37). Variants in locus 6q24.3-q25.1 reached marginal significance (rs9390543, SASH1, P = 3.56 × 10-6, OR 0.75) in CCSS and significance when meta-analyzing with SJLIFE (P = 2.02 × 10-8, OR 0.75). Both loci were exclusively associated with PTSD in CCS rather than PTSD/stress-related disorders in general population (P-for-heterogeneity < 5 × 10-6). Our CCS findings support the role of genetic variation in PTSD development and may provide implications for understanding PTSD heterogeneity.
Collapse
Affiliation(s)
- Donghao Lu
- Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, 17177, Stockholm, Sweden. .,Perini Family Survivors' Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA. .,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, 02115, US.
| | - Yadav Sapkota
- grid.240871.80000 0001 0224 711XDepartment of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Unnur A. Valdimarsdóttir
- grid.4714.60000 0004 1937 0626Unit of Integrative Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, 17177 Stockholm, Sweden ,grid.38142.3c000000041936754XDepartment of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115 US ,grid.14013.370000 0004 0640 0021Center of Public Health Sciences, Faculty of Medicine, University of Iceland, 101 Reykjavík, Iceland
| | - Karestan C. Koenen
- grid.38142.3c000000041936754XDepartment of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115 US ,grid.32224.350000 0004 0386 9924Department of Psychiatry, Psychiatric and Neurodevelopmental Genetics Research Unit, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Nan Li
- grid.240871.80000 0001 0224 711XDepartment of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Wendy M. Leisenring
- grid.270240.30000 0001 2180 1622Public Health Sciences and Clinical Research Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA 98109 USA
| | - Todd Gibson
- grid.94365.3d0000 0001 2297 5165Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, 20892 MD US
| | - Carmen L. Wilson
- grid.240871.80000 0001 0224 711XDepartment of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Leslie L. Robison
- grid.240871.80000 0001 0224 711XDepartment of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Melissa M. Hudson
- grid.240871.80000 0001 0224 711XDepartment of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA ,grid.240871.80000 0001 0224 711XDepartment of Oncology, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Gregory T. Armstrong
- grid.240871.80000 0001 0224 711XDepartment of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Kevin R. Krull
- grid.240871.80000 0001 0224 711XDepartment of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA ,grid.240871.80000 0001 0224 711XDepartment of Psychology, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Yutaka Yasui
- grid.240871.80000 0001 0224 711XDepartment of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105 USA
| | - Smita Bhatia
- grid.265892.20000000106344187Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233 USA
| | - Christopher J. Recklitis
- grid.38142.3c000000041936754XPerini Family Survivors’ Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215 USA
| |
Collapse
|
12
|
Chen YL, Tong L, Chen Y, Fu CH, Peng JB, Ji LL. MiR-153 downregulation alleviates PTSD-like behaviors and reduces cell apoptosis by upregulating the Sigma-1 receptor in the hippocampus of rats exposed to single-prolonged stress. Exp Neurol 2022; 352:114034. [PMID: 35259352 DOI: 10.1016/j.expneurol.2022.114034] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/24/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022]
Abstract
Posttraumatic stress disorder (PTSD) is a psychiatric disorder that may lead to a series of changes in the central nervous system, including impaired synaptic plasticity, neuronal dendritic spine loss, enhanced apoptosis and increased inflammation. However, the specific mechanism of PTSD has not been studied clearly. In the present study, we found that the level of miR-153-3p in the hippocampus of rats exposed tosingle-prolonged stresss (SPS) was upregulated, but its downstream target σ-1R showed a significant decrease. The downregulation of miR-153 could alleviate the PTSD-like behaviors in the rats exposed to SPS, and this effect might be related to the upregulation of σ-1R and PSD95. Furthermore, anti-miR-153 could also increase the dendritic spine density and reduce cell apoptosis in the hippocampus of SPS rats. In addition, we showed that the mTOR signaling pathway might be involved in the regulation of σ-1R in the hippocampus of rats exposed to SPS. The results of this study indicated that miR-153 might alleviate PTSD-like behaviors by regulating cell morphology and reducing cell apoptosis in the hippocampus of rats exposed to SPS by targeting σ-1R, which might be related to the mTOR signaling pathway.
Collapse
Affiliation(s)
- Yu-Lu Chen
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yao Chen
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jun-Bo Peng
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| | - Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
13
|
Anxiety and hippocampal neuronal activity: Relationship and potential mechanisms. COGNITIVE, AFFECTIVE & BEHAVIORAL NEUROSCIENCE 2022; 22:431-449. [PMID: 34873665 DOI: 10.3758/s13415-021-00973-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 12/15/2022]
Abstract
The hippocampus has been implicated in modulating anxiety. It interacts with a variety of brain regions, both cortical and subcortical areas regulating emotion and stress responses, including prefrontal cortex, amygdala, hypothalamus, and the nucleus accumbens, to adjust anxiety levels in response to a variety of stressful conditions. Growing evidence indicates that anxiety is associated with increased neuronal excitability in the hippocampus, and alterations in local regulation of hippocampal excitability have been suggested to underlie behavioral disruptions characteristic of certain anxiety disorders. Furthermore, studies have shown that some anxiolytics can treat anxiety by altering the excitability and plasticity of hippocampal neurons. Hence, identifying cellular and molecular mechanisms and neural circuits that regulate hippocampal excitability in anxiety may be beneficial for developing targeted interventions for treatment of anxiety disorders particularly for the treatment-resistant cases. We first briefly review a role of the hippocampus in fear. We then review the evidence indicating a relationship between the hippocampal activity and fear/anxiety and discuss some possible mechanisms underlying stress-induced hippocampal excitability and anxiety-related behavior.
Collapse
|
14
|
Palamarchuk IS, Vaillancourt T. Integrative Brain Dynamics in Childhood Bullying Victimization: Cognitive and Emotional Convergence Associated With Stress Psychopathology. Front Integr Neurosci 2022; 16:782154. [PMID: 35573445 PMCID: PMC9097078 DOI: 10.3389/fnint.2022.782154] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 03/15/2022] [Indexed: 12/14/2022] Open
Abstract
Bullying victimization is a form of psychological stress that is associated with poor outcomes in the areas of mental health and learning. Although the emotional maladjustment and memory impairment following interpersonal stress are well documented, the mechanisms of complex cerebral dysfunctions have neither been outlined nor studied in depth in the context of childhood bullying victimization. As a contribution to the cross-disciplinary field of developmental psychology and neuroscience, we review the neuropathophysiology of early life stress, as well as general psychological stress to synthesize the data and clarify the versatile dynamics within neuronal networks linked to bullying victimization. The stress-induced neuropsychological cascade and associated cerebral networks with a focus on cognitive and emotional convergence are described. The main findings are that stress-evoked neuroendocrine reactivity relates to neuromodulation and limbic dysregulation that hinder emotion processing and executive functioning such as semantic cognition, cognitive flexibility, and learning. Developmental aspects and interacting neural mechanisms linked to distressed cognitive and emotional processing are pinpointed and potential theory-of-mind nuances in targets of bullying are presented. The results show that childhood stress psychopathology is associated with a complex interplay where the major role belongs to, but is not limited to, the amygdala, fusiform gyrus, insula, striatum, and prefrontal cortex. This interplay contributes to the sensitivity toward facial expressions, poor cognitive reasoning, and distress that affect behavioral modulation and emotion regulation. We integrate the data on major brain dynamics in stress neuroactivity that can be associated with childhood psychopathology to help inform future studies that are focused on the treatment and prevention of psychiatric disorders and learning problems in bullied children and adolescents.
Collapse
|
15
|
Wang J, Chen F, Zhu S, Li X, Shi W, Dai Z, Hao L, Wang X. Adverse effects of prenatal dexamethasone exposure on fetal development. J Reprod Immunol 2022; 151:103619. [DOI: 10.1016/j.jri.2022.103619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 02/20/2022] [Accepted: 03/24/2022] [Indexed: 12/15/2022]
|
16
|
Weis CN, Webb EK, deRoon-Cassini TA, Larson CL. Emotion Dysregulation Following Trauma: Shared Neurocircuitry of Traumatic Brain Injury and Trauma-Related Psychiatric Disorders. Biol Psychiatry 2022; 91:470-477. [PMID: 34561028 PMCID: PMC8801541 DOI: 10.1016/j.biopsych.2021.07.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/13/2021] [Accepted: 07/25/2021] [Indexed: 11/02/2022]
Abstract
The psychological trauma associated with events resulting in traumatic brain injury (TBI) is an important and frequently overlooked factor that may impede brain recovery and worsen mental health following TBI. Indeed, individuals with comorbid posttraumatic stress disorder (PTSD) and TBI have significantly poorer clinical outcomes than individuals with a sole diagnosis. Emotion dysregulation is a common factor leading to poor cognitive and affective outcomes following TBI. Here, we synthesize how acute postinjury molecular processes stemming from either physical or emotional trauma may adversely impact circuitry subserving emotion regulation and ultimately yield long-term system-level functional and structural changes that are common to TBI and PTSD. In the immediate aftermath of traumatic injury, glucocorticoids stimulate excess glutamatergic activity, particularly in prefrontal cortex-subcortical circuitry implicated in emotion regulation. In human neuroimaging work, assessing this same circuitry well after the acute injury, TBI and PTSD show similar impacts on prefrontal and subcortical connectivity and activation. These neural profiles indicate that emotion regulation may be a useful target for treatment and early intervention to prevent the adverse sequelae of TBI. Ultimately, the success of future TBI and PTSD early interventions depends on the fields' ability to address both the physical and emotional impact of physical injury.
Collapse
|
17
|
Al Yacoub ON, Awwad HO, Zhang Y, Standifer KM. Therapeutic potential of nociceptin/orphanin FQ peptide (NOP) receptor modulators for treatment of traumatic brain injury, traumatic stress, and their co-morbidities. Pharmacol Ther 2022; 231:107982. [PMID: 34480968 DOI: 10.1016/j.pharmthera.2021.107982] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
The nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor is a member of the opioid receptor superfamily with N/OFQ as its endogenous agonist. Wide expression of the NOP receptor and N/OFQ, both centrally and peripherally, and their ability to modulate several biological functions has led to development of NOP receptor modulators by pharmaceutical companies as therapeutics, based upon their efficacy in preclinical models of pain, anxiety, depression, Parkinson's disease, and substance abuse. Both posttraumatic stress disorder (PTSD) and traumatic brain injury (TBI) are debilitating conditions that significantly affect the quality of life of millions of people around the world. PTSD is often a consequence of TBI, and, especially for those deployed to, working and/or living in a war zone or are first responders, they are comorbid. PTSD and TBI share common symptoms, and negatively influence outcomes as comorbidities of the other. Unfortunately, a lack of effective therapies or therapeutic agents limits the long term quality of life for either TBI or PTSD patients. Ours, and other groups, demonstrated that PTSD and TBI preclinical models elicit changes in the N/OFQ-NOP receptor system, and that administration of NOP receptor ligands alleviated some of the neurobiological and behavioral changes induced by brain injury and/or traumatic stress exposure. Here we review the past and most recent progress on understanding the role of the N/OFQ-NOP receptor system in PTSD and TBI neurological and behavioral sequelae. There is still more to understand about this neuropeptide system in both PTSD and TBI, but current findings warrant further examination of the potential utility of NOP modulators as therapeutics for these disorders and their co-morbidities. We advocate the development of standards for common data elements (CDE) reporting for preclinical PTSD studies, similar to current preclinical TBI CDEs. That would provide for more standardized data collection and reporting to improve reproducibility, interpretation and data sharing across studies.
Collapse
Affiliation(s)
- Omar N Al Yacoub
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, OUHSC, Oklahoma City, OK 73117, United States of America
| | - Hibah O Awwad
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, OUHSC, Oklahoma City, OK 73117, United States of America
| | - Yong Zhang
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, OUHSC, Oklahoma City, OK 73117, United States of America
| | - Kelly M Standifer
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, OUHSC, Oklahoma City, OK 73117, United States of America.
| |
Collapse
|
18
|
Trauma-like exposure alters neuronal apoptosis, Bin1, Fkbp5 and NR2B expression in an amyloid-beta (1-42) rat model of Alzheimer's disease. Neurobiol Learn Mem 2022; 190:107611. [DOI: 10.1016/j.nlm.2022.107611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/01/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022]
|
19
|
Metabolomic signature and mitochondrial dynamics outline the difference between vulnerability and resilience to chronic stress. Transl Psychiatry 2022; 12:87. [PMID: 35228511 PMCID: PMC8885712 DOI: 10.1038/s41398-022-01856-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 12/14/2022] Open
Abstract
Stress is the foremost environmental factor involved in the pathophysiology of major depressive disorder (MDD). However, individual differences among people are critical as some people exhibit vulnerability while other are resilient to repeated exposure to stress. Among the others, a recent theory postulates that alterations of energy metabolism might contribute to the development of psychopathologies. Here we show that the bioenergetic status in the ventral hippocampus (vHip), a brain subregion tightly involved in the regulation of MDD, defined the development of vulnerability or resilience following two weeks of chronic mild stress. Among the different metabolomic signatures observed, the glycolysis and tricarboxylic acid cycle may be specifically involved in defining vulnerability, revealing a previously unappreciated mechanism of sensitivity to stress. These findings point to mitochondrial morphology and recycling as critical in the ability to cope with stress. We show that vulnerable rats favor mitochondrial fusion to counteract the overproduction of reactive oxidative species whereas resilient rats activate fission to guarantee metabolic efficiency. Our results indicate that the modulation of the energetic metabolite profile in vHip under chronic stress exposure may represent a mechanism to explain the difference between vulnerable and resilient rats, unraveling novel and promising targets for specific therapeutic interventions.
Collapse
|
20
|
Repeated cocaine exposure prior to fear conditioning induces persistency of PTSD-like symptoms and enhancement of hippocampal and amygdala cell density in male rats. Brain Struct Funct 2021; 226:2219-2241. [PMID: 34195855 DOI: 10.1007/s00429-021-02320-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/11/2021] [Indexed: 12/29/2022]
Abstract
Pre- and post-trauma drug use can interfere with recovery from post-traumatic stress disorder (PTSD). However, the biological underpinnings of this interference are poorly understood. Here we examined the effect of pre-fear conditioning cocaine self-administration on PTSD-like symptoms in male rats, and defined impairment of fear extinction as difficulty to recover from PTSD. We also examined cell density changes in brain regions suspected of being involved in resistance to PTSD recovery. Before footshock stress testing, rats were trained to self-administer cocaine during 20 consecutive days, after which they were exposed to footshocks, while other rats continued to self-administer cocaine until the end of the experiment. Upon assessment of three PTSD-like symptoms (fear during situational reminders, anxiety-like behavior, and impairment of recognition memory) and fear extinction learning and memory, changes in cell density were measured in the medial prefrontal cortex, hippocampus, and amygdala. Results show that pre-footshock cocaine exposure did not affect fear during situational reminders. Fear conditioning did not lead to an increase in cocaine consumption. However, in footshock stressed rats, cocaine induced a reduction of anxiety-like behavior, an aggravation of recognition memory decline, and an impairment of extinction memory. These behavioral alterations were associated with increased cell density in the hippocampal CA1, CA2, and CA3 regions and basolateral amygdala, but not in the medial prefrontal cortex. Our findings suggest that enhancement of cell density in the hippocampus and amygdala may be changes associated with drug use, interfering with PTSD recovery.
Collapse
|
21
|
Dysregulation of miR-15a-5p, miR-497a-5p and miR-511-5p Is Associated with Modulation of BDNF and FKBP5 in Brain Areas of PTSD-Related Susceptible and Resilient Mice. Int J Mol Sci 2021; 22:ijms22105157. [PMID: 34068160 PMCID: PMC8153003 DOI: 10.3390/ijms22105157] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/21/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a neuropsychiatric disorder occurring in susceptible individuals following a traumatic event. Understanding the mechanisms subserving trauma susceptibility/resilience is essential to develop new effective treatments. Increasing evidence suggests that non-coding RNAs, such as microRNAs (miRNAs), may play a prominent role in mediating trauma susceptibility/resilience. In this study, we evaluated the transcriptional expression of two key PTSD-related genes (FKBP5 and BDNF) and the relative targeting miRNAs (miR-15a-5p, miR-497a-5p, miR-511-5p, let-7d-5p) in brain areas of PTSD-related susceptible and resilient mice identified through our recently developed mouse model of PTSD (arousal-based individual screening (AIS) model). We observed lower transcript levels of miR-15a-5p, miR-497a-5p, and miR-511a-5p in the hippocampus and hypothalamus of susceptible mice compared to resilient mice, suggesting that the expression of these miRNAs could discriminate the two different phenotypes of stress-exposed mice. These miRNA variations could contribute, individually or synergically, to the inversely correlated transcript levels of FKBP5 and BDNF. Conversely, in the medial prefrontal cortex, downregulation of miR-15a-5p, miR-511-5p, and let-7d-5p was observed both in susceptible and resilient mice, and not accompanied by changes in their mRNA targets. Furthermore, miRNA expression in the different brain areas correlated to stress-induced behavioral scores (arousal score, avoidance-like score, social memory score and PTSD-like score), suggesting a linear connection between miRNA-based epigenetic modulation and stress-induced phenotypes. Pathway analysis of a miRNA network showed a statistically significant enrichment of molecular processes related to PTSD and stress. In conclusion, our results indicate that PTSD susceptibility/resilience might be shaped by brain-area-dependent modulation of miRNAs targeting FKBP5, BDNF, and other stress-related genes.
Collapse
|
22
|
El-Ansary A, Zayed N, Al-Ayadhi L, Qasem H, Anwar M, Meguid NA, Bhat RS, Doşa MD, Chirumbolo S, Bjørklund G. GABA synaptopathy promotes the elevation of caspases 3 and 9 as pro-apoptotic markers in Egyptian patients with autism spectrum disorder. Acta Neurol Belg 2021; 121:489-501. [PMID: 31673995 DOI: 10.1007/s13760-019-01226-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/10/2019] [Indexed: 12/14/2022]
Abstract
Autism spectrum disorder (ASD) is classified as a neurodevelopmental disorder characterized by reduced social communication as well as repetitive behaviors. Many studies have proved that defective synapses in ASD influence how neurons in the brain connect and communicate with each other. Synaptopathies arise from alterations that affecting the integrity and/or functionality of synapses and can contribute to synaptic pathologies. This study investigated the GABA levels in plasma being an inhibitory neurotransmitter, caspase 3 and 9 as pro-apoptotic proteins in 20 ASD children and 20 neurotypical controls using the ELISA technique. Analysis of receiver-operating characteristic (ROC) of the data that was obtained to evaluate the diagnostic value of the aforementioned evaluated biomarkers. Pearson's correlations and multiple regressions between the measured variables were also done. While GABA level was reduced in ASD patients, levels of caspases 3 and 9 were significantly higher when compared to neurotypical control participants. ROC and predictiveness curves showed that caspases 3, caspases 9, and GABA might be utilized as predictive markers in autism diagnosis. The present study indicates that the presence of GABAergic dysfunction promotes apoptosis in Egyptian ASD children. The obtained GABA synaptopathies and their connection with apoptosis can both relate to neuronal excitation, and imbalance of the inhibition system, which can be used as reliable predictive biomarkers for ASD.
Collapse
|
23
|
Gao J, Qin Z, Qu X, Wu S, Xie X, Liang C, Liu J. Endogenous neuroprotective mechanism of ATP2B1 in transcriptional regulation of ischemic preconditioning. Am J Transl Res 2021; 13:1170-1183. [PMID: 33841647 PMCID: PMC8014370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/18/2021] [Indexed: 06/12/2023]
Abstract
UNLABELLED Ischemic stroke is the main cause of disability and mortality in the world. Clinical studies have shown that patients who undergo mild transient ischemic attack (TIA) before more severe ischemic stroke have lower clinical severity of stroke and better functional prognosis. This phenomenon is called ischemic preconditioning (IPC). IPC is a powerful intrinsic protection of the brain against ischemic injury, but the underlying mechanism of IPC-mediated endogenous protection of the brain is not clear. METHODS Using transcriptome method, we sequenced the serum of 3 stroke patients with progenitor TIA and 3 stroke patients without prodromal TIA. We explored the expression profiles of miRNAs and mRNAs in response to IPC, and predicted the regulatory pathway of IPC related genes and their expression in cerebral neurons. The methylation consistent expression of IPC-related gene ATP2B1 in blood and brain and alternative polyadenylate (APA) analysis were used to identify the pathway and molecular mechanism of endogenous neuroprotection of IPC. RESULTS We found that the brain protective effect of IPC was related to platelet homeostasis and Ca2+ concentration. IPC-related gene ATP2B1 was highly expressed in γ-aminobutyric acid (GABA)-containing neurons in the brain. From the mechanism, we speculated that ATP2B1 was representative of the same methylation in blood and brain and was affected by alternative polyadenylation. CONCLUSION We speculate that IPC can induce alternative polyadenylation of ATP2B1 and trigger the mechanism of brain endogenous neuroprotection by regulating the decrease of Ca2+ concentration in platelet homeostasis pathway and the activation of GABAB receptor.
Collapse
Affiliation(s)
- Jinggui Gao
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning China
| | - Zhenxiu Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning China
| | - Xiang Qu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning China
| | - Shuang Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning China
| | - Xiaoyun Xie
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning China
| | - Chengwei Liang
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning China
| | - Jingli Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University in Nanning China
| |
Collapse
|
24
|
Loss of α7 nicotinic acetylcholine receptors in GABAergic neurons causes sex-dependent decreases in radial glia-like cell quantity and impairments in cognitive and social behavior. Brain Struct Funct 2021; 226:365-379. [PMID: 33398432 DOI: 10.1007/s00429-020-02179-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 11/10/2020] [Indexed: 12/25/2022]
Abstract
The dentate gyrus (DG) is a unique brain structure in that neurons can be generated postnatally and integrated within existing circuitry throughout life. The maturation process of these newly generated neurons (granule cells) is modulated by nicotinic acetylcholine receptors (nAChRs) through a variety of mechanisms such as neural stem pool proliferation, cell survival, signal modulation, and dendritic integration. Disrupted nAChR signaling has been implicated in neuropsychiatric and neurodegenerative disorders, potentially via alterations in DG neurogenesis. GABAergic interneurons are known to express nAChRs, predominantly the α7 subtype, and have been shown to shape development, integration, and circuit reorganization of DG granule cells. Therefore, we examined histological and behavioral effects of knocking out α7 nAChRs in GABAergic neurons. Deletion of α7 nAChRs resulted in a reduction of radial glia-like cells within the subgranular zone of the DG and a concomitant trend towards decreased immature neurons, specifically in male mice, as well as sex-dependent changes in several behaviors, including social recognition and spatial learning. Overall, these findings suggest α7 nAChRs expressed in GABAergic neurons play an important role in regulating the adult neural stem cell pool and behavior in a sex-dependent manner. This provides important insight into the mechanisms by which cholinergic dysfunction contributes to the cognitive and behavioral changes associated with neurodevelopmental and neurodegenerative disorders.
Collapse
|
25
|
Batelaan NM, Seldenrijk A, van den Heuvel OA, van Balkom AJLM, Kaiser A, Reneman L, Tan HL. Anxiety, Mental Stress, and Sudden Cardiac Arrest: Epidemiology, Possible Mechanisms and Future Research. Front Psychiatry 2021; 12:813518. [PMID: 35185641 PMCID: PMC8850954 DOI: 10.3389/fpsyt.2021.813518] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 12/29/2021] [Indexed: 12/26/2022] Open
Abstract
Sudden cardiac arrest (SCA) is a leading cause of mortality and morbidity in affluent societies, which underscores the need to identify persons at risk. The etiology of SCA is however complex, with predisposing and precipitating factors interacting. Although anxiety and mental stress have been linked to SCA for decades, their precise role and impact remain unclear and the biological underpinnings are insufficiently understood. In this paper, we systematically reviewed various types of observational studies (total n = 20) examining the association between anxiety or mental stress and SCA. Multiple methodological considerations challenged the summarizing and interpretation of the findings. For anxiety, the overall picture suggests that it predisposes for SCA in physically healthy populations (unadjusted OR = 2.44; 95% CI: 1.06-5.59; n = 3). However, in populations at risk for SCA (n = 4), associations were heterogeneous but not significant. Anxiety may partly predispose to SCA by contributing to other risk factors such as cardiovascular disease and diabetes mellitus via mechanisms such as unhealthy lifestyle and metabolic abnormalities. Mental stress appears to precipitate SCA, presumably by more directly impacting on the cardiac ion channels that control the heart's electrical properties. This may lead to ventricular fibrillation, the arrhythmia that underlies SCA. To advance this field of research, experimental studies that unravel the underlying biological mechanisms are deemed important, and most easily designed for mental stress as a precipitating factor because of the short timeframe. These proof-of-concept studies should examine the whole pathway from the brain to the autonomic nervous system, and eventually to cardiac ion channels. Ultimately, such studies may facilitate the identification of persons at risk and the development of novel preventive strategies.
Collapse
Affiliation(s)
- Neeltje M Batelaan
- Department of Psychiatry, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, Netherlands
| | - Adrie Seldenrijk
- Department of Psychiatry, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, Netherlands
| | - Odile A van den Heuvel
- Department of Psychiatry, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam Neuroscience Research Institute, Amsterdam, Netherlands.,Department of Anatomy and Neuroscience, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Anton J L M van Balkom
- Department of Psychiatry, Amsterdam University Medical Center (UMC), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Amsterdam Public Health Research Institute, Amsterdam, Netherlands
| | - Antonia Kaiser
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Department of Psychiatry, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Liesbeth Reneman
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands
| | - Hanno L Tan
- Department of Clinical and Experimental Cardiology, Amsterdam University Medical Center (UMC), University of Amsterdam, Amsterdam, Netherlands.,Netherlands Heart Institute, Utrecht, Netherlands
| |
Collapse
|
26
|
Allene C, Kalalou K, Durand F, Thomas F, Januel D. Acute and Post-Traumatic Stress Disorders: A biased nervous system. Rev Neurol (Paris) 2020; 177:23-38. [PMID: 32800536 DOI: 10.1016/j.neurol.2020.05.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 11/24/2022]
Abstract
Acute stress disorder and post-traumatic stress disorder are generally triggered by an exceptionally intense threat. The consequences of this traumatogenic situation are explored here in chronological order, from exposure to the threat to development of symptoms. Such a situation may disrupt the equilibrium between two fundamental brain circuits, referred to as the "defensive" and "cognitive". The defensive circuit triggers the stress response as well as the formation of implicit memory. The cognitive circuit triggers the voluntary response and the formation of explicit autobiographical memory. During a traumatogenic situation, the defensive circuit could be over-activated while cognitive circuit is under-activated. In the most severe cases, overactivation of the defensive circuit may cause its brutal deactivation, resulting in dissociation. Here, we address the underlying neurobiological mechanisms at every scale: from neurons to behaviors, providing a detailed explanatory model of trauma.
Collapse
Affiliation(s)
- C Allene
- Unité de recherche clinique, établissement public de santé Ville-Evrard, 202, avenue Jean-Jaurès, 93332 Neuilly-sur-Marne, France; Centre de psychothérapie, établissement public de santé Ville-Evrard, 5, rue du Docteur-Delafontaine, 93200 Saint-Denis, France.
| | - K Kalalou
- Unité de recherche clinique, établissement public de santé Ville-Evrard, 202, avenue Jean-Jaurès, 93332 Neuilly-sur-Marne, France; Centre de psychothérapie, établissement public de santé Ville-Evrard, 5, rue du Docteur-Delafontaine, 93200 Saint-Denis, France.
| | - F Durand
- Unité de recherche clinique, établissement public de santé Ville-Evrard, 202, avenue Jean-Jaurès, 93332 Neuilly-sur-Marne, France; Centre de psychothérapie, établissement public de santé Ville-Evrard, 5, rue du Docteur-Delafontaine, 93200 Saint-Denis, France.
| | - F Thomas
- Unité de recherche clinique, établissement public de santé Ville-Evrard, 202, avenue Jean-Jaurès, 93332 Neuilly-sur-Marne, France.
| | - D Januel
- Unité de recherche clinique, établissement public de santé Ville-Evrard, 202, avenue Jean-Jaurès, 93332 Neuilly-sur-Marne, France.
| |
Collapse
|
27
|
Unno K, Furushima D, Nomura Y, Yamada H, Iguchi K, Taguchi K, Suzuki T, Ozeki M, Nakamura Y. Antidepressant Effect of Shaded White Leaf Tea Containing High Levels of Caffeine and Amino Acids. Molecules 2020; 25:molecules25153550. [PMID: 32756488 PMCID: PMC7435365 DOI: 10.3390/molecules25153550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/21/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022] Open
Abstract
The young leaves of green tea become lighter in color than usual when protected from sunlight by a shading net for about two weeks while growing. These leaves are called "shaded white leaf tea" or SWLT. In the eluate of SWLT, the amount of amino acids (361 mg/L) was significantly higher than that in regular tea (53.5 mg/L). Since theanine and arginine, the first and second most abundant amino acids in SWLT, have significant antistress effects, we examined the antistress effect of SWLT on humans. SWLT or placebo green tea (3 g) was eluted with room-temperature water (500 mL). Participants consumed the tea for one week prior to pharmacy practice and continued for 10 days in the practice period. The state-trait anxiety inventory, an anxiety questionnaire, tended to be scored lower in the SWLT group than the placebo, but other stress markers showed no differences. The effect of the difference in SWLT components examined with mice showed that aspartic acid and asparagine, which are abundant in SWLT, counteracted the antistress effects of theanine and arginine. Large amounts of caffeine also interfered with SWLT's antistress effect. Thus, SWLT, which is high in caffeine and amino acids, suppressed depressant behavior in mice.
Collapse
Affiliation(s)
- Keiko Unno
- Tea Science Center, Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.T.); (Y.N.)
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
- Correspondence: ; Tel.: +81-54-264-5822
| | - Daisuke Furushima
- Department of Drug Evaluation & Informatics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (D.F.); (Y.N.); (H.Y.)
| | - Yuzuki Nomura
- Department of Drug Evaluation & Informatics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (D.F.); (Y.N.); (H.Y.)
| | - Hiroshi Yamada
- Department of Drug Evaluation & Informatics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (D.F.); (Y.N.); (H.Y.)
| | - Kazuaki Iguchi
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan;
| | - Kyoko Taguchi
- Tea Science Center, Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.T.); (Y.N.)
| | - Toshikazu Suzuki
- Tea Research Center, Shizuoka Prefectural Research Institute of Agriculture and Forestry, Kikugawa, Shizuoka 439-0002, Japan;
| | - Makoto Ozeki
- Nutrition Division, R&D Group, Taiyo Kagaku Co., Ltd., Yokkaichi, Mie 510-0844, Japan;
| | - Yoriyuki Nakamura
- Tea Science Center, Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka 422-8526, Japan; (K.T.); (Y.N.)
| |
Collapse
|
28
|
Vinnakota C, Govindpani K, Tate WP, Peppercorn K, Anekal PV, Waldvogel HJ, Faull RLM, Kwakowsky A. An 5 GABAA Receptor Inverse Agonist, 5IA, Attenuates Amyloid Beta-Induced Neuronal Death in Mouse Hippocampal Cultures. Int J Mol Sci 2020; 21:ijms21093284. [PMID: 32384683 PMCID: PMC7247548 DOI: 10.3390/ijms21093284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/30/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder for which no cognition-restoring therapies exist. Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter in the brain. Increasing evidence suggests a remodeling of the GABAergic system in AD, which might represent an important therapeutic target. An inverse agonist of α5 subunit-containing GABAA receptors (α5GABAARs), 3-(5-Methylisoxazol-3-yl)-6-[(1-methyl-1,2,3-triazol-4-yl)methyloxy]-1,2,4-triazolo[3–a]phthalazine (α5IA) has cognition-enhancing properties. This study aimed to characterize the effects of α5IA on amyloid beta (Aβ1–42)-induced molecular and cellular changes. Mouse primary hippocampal cultures were exposed to either Aβ1-42 alone, or α5IA alone, α5IA with Aβ1–42 or vehicle alone, and changes in cell viability and mRNA expression of several GABAergic signaling components were assessed. Treatment with 100 nM of α5IA reduced Aβ1–42-induced cell loss by 23.8% (p < 0.0001) after 6 h and by 17.3% after 5 days of treatment (p < 0.0001). Furthermore, we observed an Aβ1-42-induced increase in ambient GABA levels, as well as upregulated mRNA expression of the GABAAR α2,α5,β2/3 subunits and the GABABR R1 and R2 subunits. Such changes in GABARs expression could potentially disrupt inhibitory neurotransmission and normal network activity. Treatment with α5IA restored Aβ1-42-induced changes in the expression of α5GABAARs. In summary, this compound might hold neuroprotective potential and represent a new therapeutic avenue for AD.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Karan Govindpani
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Warren Perry Tate
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (W.P.T.); (K.P.)
| | - Katie Peppercorn
- Department of Biochemistry, University of Otago, Dunedin 9054, New Zealand; (W.P.T.); (K.P.)
| | - Praju Vikas Anekal
- Biomedical Imaging Research Unit, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand;
| | - Henry John Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Richard Lewis Maxwell Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health, Sciences, University of Auckland, Auckland 1023, New Zealand; (C.V.); (K.G.); (H.J.W.); (R.L.M.F.)
- Correspondence: ; Tel.: +64-9923-9346
| |
Collapse
|
29
|
LPA 1 receptor and chronic stress: Effects on behaviour and the genes involved in the hippocampal excitatory/inhibitory balance. Neuropharmacology 2020; 164:107896. [PMID: 31811875 DOI: 10.1016/j.neuropharm.2019.107896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 11/22/2022]
Abstract
The LPA1 receptor, one of the six characterized G protein-coupled receptors (LPA1-6) through which lysophosphatidic acid acts, is likely involved in promoting normal emotional behaviours. Current data suggest that the LPA-LPA1-receptor pathway may be involved in mediating the negative consequences of stress on hippocampal function. However, to date, there is no available information regarding the mechanisms whereby the LPA1 receptor mediates this adaptation. To gain further insight into how the LPA-LPA1 pathway may prevent the negative consequences of chronic stress, we assessed the effects of the continuous delivery of LPA on depressive-like behaviours induced by a chronic restraint stress protocol. Because a proper excitatory/inhibitory balance seems to be key for controlling the stress response system, the gene expression of molecular markers of excitatory and inhibitory neurotransmission was also determined. In addition, the hippocampal expression of mineralocorticoid receptor genes and glucocorticoid receptor genes and proteins as well as plasma corticosterone levels were determined. Contrary to our expectations, the continuous delivery of LPA in chronically stressed animals potentiated rather than inhibited some (e.g., anhedonia, reduced latency to the first immobility period), though not all, behavioural effects of stress. Furthermore, this treatment led to an alteration in the genes coding for proteins involved in the excitatory/inhibitory balance in the ventral hippocampus and to changes in corticosterone levels. In conclusion, the results of this study reinforce the assumption that LPA is involved in emotional regulation, mainly through the LPA1 receptor, and regulates the effects of stress on hippocampal gene expression and hippocampus-dependent behaviour.
Collapse
|
30
|
Behuet S, Cremer JN, Cremer M, Palomero-Gallagher N, Zilles K, Amunts K. Developmental Changes of Glutamate and GABA Receptor Densities in Wistar Rats. Front Neuroanat 2019; 13:100. [PMID: 31920569 PMCID: PMC6933313 DOI: 10.3389/fnana.2019.00100] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 12/02/2019] [Indexed: 12/02/2022] Open
Abstract
Neurotransmitters and their receptors are key molecules of signal transduction and subject to various changes during pre- and postnatal development. Previous studies addressed ontogeny at the level of neurotransmitters and expression of neurotransmitter receptor subunits. However, developmental changes in receptor densities to this day are not well understood. Here, we analyzed developmental changes in excitatory glutamate and inhibitory γ-aminobutyric acid (GABA) receptors in adjacent sections of the rat brain by means of quantitative in vitro receptor autoradiography. Receptor densities of the ionotropic glutamatergic receptors α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), kainate and N-methyl-D-aspartate (NMDA) as well as of the ionotropic GABAA and metabotropic GABAB receptors were investigated using specific high-affinity ligands. For each receptor binding site, significant density differences were demonstrated in the investigated regions of interest [olfactory bulb, striatum, hippocampus, and cerebellum] and developmental stages [postnatal day (P) 0, 10, 20, 30 and 90]. In particular, we showed that the glutamatergic and GABAergic receptor densities were already present between P0 and P10 in all regions of interest, which may indicate the early relevance of these receptors for brain development. A transient increase of glutamatergic receptor densities in the hippocampus was found, indicating their possible involvement in synaptic plasticity. We demonstrated a decline of NMDA receptor densities in the striatum and hippocampus from P30 to P90, which could be due to synapse elimination, a process that redefines neuronal networks in postnatal brains. Furthermore, the highest increase in GABAA receptor densities from P10 to P20 coincides with the developmental shift from excitatory to inhibitory GABA transmission. Moreover, the increase from P10 to P20 in GABAA receptor densities in the cerebellum corresponds to a point in time when functional GABAergic synapses are formed. Taken together, the present data reveal differential changes in glutamate and GABA receptor densities during postnatal rat brain development, which may contribute to their specific functions during ontogenesis, thus providing a deeper understanding of brain ontogenesis and receptor function.
Collapse
Affiliation(s)
- Sabrina Behuet
- Institute of Neuroscience and Medicine (INM-1), Jülich Research Centre, Jülich, Germany
| | | | - Markus Cremer
- Institute of Neuroscience and Medicine (INM-1), Jülich Research Centre, Jülich, Germany
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Jülich Research Centre, Jülich, Germany.,Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Karl Zilles
- Institute of Neuroscience and Medicine (INM-1), Jülich Research Centre, Jülich, Germany
| | - Katrin Amunts
- Institute of Neuroscience and Medicine (INM-1), Jülich Research Centre, Jülich, Germany.,Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| |
Collapse
|
31
|
Luo ML, Pan L, Wang L, Wang HY, Li S, Long ZY, Zeng L, Liu Y. Transplantation of NSCs Promotes the Recovery of Cognitive Functions by Regulating Neurotransmitters in Rats with Traumatic Brain Injury. Neurochem Res 2019; 44:2765-2775. [PMID: 31701381 DOI: 10.1007/s11064-019-02897-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 10/11/2019] [Accepted: 10/19/2019] [Indexed: 10/25/2022]
Abstract
Transplantation of neural stem cells (NSCs) may be a potential strategy for traumatic brain injury treatment (TBI) due to their intrinsic advantages, such as cell replacement, secretion of neurotrophins and formation of functional synapses with host. However the underlying effects of transplanted NSCs on host micro-environment still need to be further elucidated. In this manuscript the effects of NSCs on release of neurotransmitter, survival of hippocampal neurons, reactivity of astrocytes and recovery of cognitive function after TBI were observed. The NSCs were isolated from cortex of neonatal Sprague-Dawley rat and then transplanted into injured brain regions caused by free-weight drop. The proliferation of astrocytes around injured sites were examined by GFAP immunofluorescent staining on 3, 7, 14 days after injury. The survival of neurons at CA1 regions of hippocampus toward contused regions was observed by HE staining on 3 and 14 days post-injury. The content of glutamic acid (Glu) and GABA in hippocampal tissues was examined on 1, 3, 7, 14, 28 days after injury by ELISA. On third day post-injury, hippocampal-dependent spatial memory was measured for 5 days without intermittent. NSCs in culture have the ability to proliferate and differentiate into different phenotypes of neural cells. After transplantation of NSCs, the proliferation of astrocytes around injured site was significantly inhibited compared to the injured group. At the same time the survival of neurons in hippocampal CA1 region were much more than those in injured group on 14 days post-injury. Meanwhile, the cognitive functions in NSC transplanted group was remarkably improved compared with injured group (p < 0.05). Furthermore, NSCs transplantation dramatically inhibited the release of Glu and maintained the content of GABA in injured hippocampal tissues on 1, 3, 7, 14, 28 days post-injury, which was of difference in statistics (p < 0.05). NSCs transplantation can effectively alleviate the formation of glial scar, enhance the survival of hippocampal neurons and improve cognitive function defects in rats with TBI. The underlying mechanism may be related to their effects on inhibiting the release of Glu and maintaining the content of GABA, so as to down-regulate excitotoxicity of neurotransmitter and improve the micro-environment in injured sites.
Collapse
Affiliation(s)
- Mei-Ling Luo
- Research Institute of Surgery, Daping Hospital, the Army Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400042, China
| | - Lu Pan
- Research Institute of Surgery, Daping Hospital, the Army Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400042, China
| | - Li Wang
- Research Institute of Surgery, Daping Hospital, the Army Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400042, China
| | - Hai-Yan Wang
- Research Institute of Surgery, Daping Hospital, the Army Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400042, China
| | - Sen Li
- Research Institute of Surgery, Daping Hospital, the Army Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400042, China
| | - Zai-Yun Long
- Research Institute of Surgery, Daping Hospital, the Army Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400042, China
| | - Lin Zeng
- Research Institute of Surgery, Daping Hospital, the Army Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400042, China
| | - Yuan Liu
- Research Institute of Surgery, Daping Hospital, the Army Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, 400042, China.
| |
Collapse
|
32
|
Huang S, Dong W, Jiao Z, Liu J, Li K, Wang H, Xu D. Prenatal Dexamethasone Exposure Induced Alterations in Neurobehavior and Hippocampal Glutamatergic System Balance in Female Rat Offspring. Toxicol Sci 2019; 171:369-384. [PMID: 31518422 DOI: 10.1093/toxsci/kfz163] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Epidemiological investigations have suggested that periodic use of dexamethasone during pregnancy is a risk factor for abnormal behavior in offspring, but the potential mechanism remains unclear. In this study, we investigated the changes in the glutamatergic system and neurobehavior in female offspring with prenatal dexamethasone exposure (PDE) to explore intrauterine programing mechanisms. Compared with the control group, rat offspring with PDE exhibited spatial memory deficits and anxiety-like behavior. The expression of hippocampal glucocorticoid receptors (GR) and histone deacetylase 2 (HDAC2) increased, whereas histone H3 lysine 14 acetylation (H3K14ac) of brain-derived neurotrophic factor (BDNF) exon IV (BDNF IV) and expression of BDNF decreased. The glutamatergic system also changed. We further observed that changes in the fetal hippocampus were consistent with those in adult offspring. In vitro, the administration of 0.5 μM dexamethasone to the H19-7 fetal hippocampal neuron cells directly led to a cascade of changes in the GR/HDAC2/BDNF pathway, whereas the GR antagonist RU486 and the HDAC2 inhibitor romidepsin (Rom) reversed changes caused by dexamethasone to the H3K14ac level of BDNF IV and to the expression of BDNF. The increase in HDAC2 can be reversed by RU486, and the changes in the glutamatergic system can be partially reversed after supplementation with BDNF. It is suggested that PDE increases the expression of HDAC2 by activating GR, reducing the H3K14ac level of BDNF IV, inducing alterations in neurobehavior and hippocampal glutamatergic system balance. The findings suggest that BDNF supplementation and glutamatergic system improvement are potential therapeutic targets for the fetal origins of abnormal neurobehavior.
Collapse
Affiliation(s)
- Songqiang Huang
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Wanting Dong
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Zhexiao Jiao
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Jie Liu
- *Department of Pharmacology, School of Basic Medical Sciences
| | - Ke Li
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, Hubei Province, China
| | - Hui Wang
- *Department of Pharmacology, School of Basic Medical Sciences
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University
| | - Dan Xu
- *Department of Pharmacology, School of Basic Medical Sciences
- Demonstration Center for Experimental Basic Medicine Education, Wuhan University
| |
Collapse
|
33
|
Lee JH, Chae EJ, Park SJ, Choi JW. Label-free detection of γ-aminobutyric acid based on silicon nanowire biosensor. NANO CONVERGENCE 2019; 6:13. [PMID: 31041617 PMCID: PMC6491525 DOI: 10.1186/s40580-019-0184-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/29/2019] [Indexed: 05/24/2023]
Abstract
γ-Aminobutyric acid (GABA) is an important inhibitory neurotransmitter in the central nervous system (CNS), which acts as a major biomarker for neurological disorders such as Parkinson's disease and Meningitis. To this end, the precise measurement of GABA molecule arisen as an important subject for the effective diagnosis and treatment of neurological disorders. However, yet highly sensitive biosensor systems which can analyze a wide range of GABA molecule in a fast response manner have not been reported. In this study, for the first time, a silicon nanowire field-effect transistor (FET) device based immunosensor was developed to detect GABA molecule. Zig-zag shaped silicon nanowires has been fabricated by electron beam lithography and the electrical property p-type FET device was validated through semiconductor analyzer. The optimal immobilizing condition of antibody against GABA molecule was determined by the fluorescent signal measurement. Various concentrations of GABA ranging from 970 fM to 9.7 μM were sensitively measured by conductance change on silicon nanowire-based through the immunoreactions. Further, owing to the ease of miniaturization and label-free system, we believe that the suggested device system has a potential to be utilized for an implantable biosensor to detect neurotransmitter in the brain and can create new opportunities in the field of diagnosis and treatment of neurological disorders.
Collapse
Affiliation(s)
- Jin-Ho Lee
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107 Korea
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854 USA
| | - Eun-Ji Chae
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107 Korea
| | - Soo-jeong Park
- Research Center for Disease Biophysics of Sogang-Harvard, Sogang University, Seoul, 04107 Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107 Korea
- Research Center for Disease Biophysics of Sogang-Harvard, Sogang University, Seoul, 04107 Korea
| |
Collapse
|
34
|
Zhang C, Fan SJ, Sun AB, Liu ZZ, Liu L. Prenatal nicotine exposure induces depression‑like behavior in adolescent female rats via modulating neurosteroid in the hippocampus. Mol Med Rep 2019; 19:4185-4194. [PMID: 30942466 PMCID: PMC6471439 DOI: 10.3892/mmr.2019.10105] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 02/28/2019] [Indexed: 01/01/2023] Open
Abstract
Prenatal nicotine exposure (PNE) is closely related to depression in offspring. However, the underlying mechanism is still unclear. We hypothesized that neurosteroid in the hippocampus may mediate PNE-induced depression-like behaviors. Nicotine was subcutaneously administered (1.0 mg/kg) to pregnant rats twice daily from gestational day (GD) 9 to 20. In adolescent offspring, PNE significantly increased immobility time and decreased the sucrose preference in female rats. The numbers of hippocampal neurons declined in the CA3 and DG regions. Steroidogenic acute regulatory protein (StAR) expression was suppressed in female rats. In fetal offspring, the neuronal numbers of CA3 regions in PNE female fetal hippocampal were significantly decreased, accompanied by the enhanced content of corticosterone and StAR expression. These data indicated that PNE induced depression-like behavior in adolescent female rats via the regulation of neurosteroid levels in the hippocampus.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Pharmacy, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei 441000, P.R. China
| | - Si-Jing Fan
- Department of Pharmacology, Medical School of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - An-Bang Sun
- Laboratory of Neuronal and Brain Disease Modulation, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Zhen-Zhen Liu
- Department of Pharmacology, Medical School of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Lian Liu
- Department of Pharmacology, Medical School of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|
35
|
Wang JY, Zhao YH, Zhang JH, Lei HW. Anti-N-Methyl-D-Aspartic Acid Receptor 2 (Anti-NR2) Antibody in Neuropsychiatric Lupus Serum Damages the Blood-Brain Barrier and Enters the Brain. Med Sci Monit 2019; 25:532-539. [PMID: 30657743 PMCID: PMC6346811 DOI: 10.12659/msm.912389] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Brain microvessel endothelial cells constitute an important component in the blood-brain barrier. Cell-culture-based models of the blood-brain barrier (BBB) have been extensively applied in pharmacology, pathology and physiology. This study investigated effects of anti-N-methyl-D-aspartic acid receptor 2 (anti-NR2), N-methyl-D-aspartic acid (NMDA) receptor antibodies, NMDA receptor antagonists, and NMDA receptor agonists on brain microvessel endothelial cell models, and verified the effect of anti-NR2 antibody on the BBB as a receptor agonist. Material/Methods The primary brain microvessel endothelial cells were isolated and cultured, and an in vitro BBB model was established based on microvessel endothelial cells. Anti-NR2 antibody, glutamic acid, ifenprodil, and memantine were added in the BBB model to analyze changes in transepithelial electrical resistance (TEER) and to examine the permeability of the brain microvessel endothelial cell model. Results The results showed that TEER values were significantly decreased by the addition of anti-NR2 antibody and glutamate, but were significantly increased by the addition of ifenprodil and memantine. TEER values showed no changes when treated by anti-NR2 antibody and ifenprodil, as well as anti-NR2 antibody and memantine. When dexamethasone was added, the TEER values increased by 23.8%, 39.4%, and 29.6% by treating with anti-NR2 antibody, positive cerebrospinal fluid, and positive serum, respectively. Conclusions Our findings show that anti-NR2 antibody in neuropsychiatric lupus serum can damage the BBB and enter the brain.
Collapse
Affiliation(s)
- Jing-Yuan Wang
- Department of Rheumatology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China (mainland)
| | - Yin-Huan Zhao
- Department of Rheumatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Ji-Hui Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| | - Hong-Wei Lei
- Department of Rheumatology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China (mainland)
| |
Collapse
|
36
|
Yoga: Balancing the excitation-inhibition equilibrium in psychiatric disorders. PROGRESS IN BRAIN RESEARCH 2019; 244:387-413. [PMID: 30732846 DOI: 10.1016/bs.pbr.2018.10.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Social behavioral disturbances are central to most psychiatric disorders. A disequilibrium within the cortical excitatory and inhibitory neurotransmitter systems underlies these deficits. Gamma-aminobutyric acid (GABA) and glutamate are the most abundant excitatory and inhibitory neurotransmitters in the brain that contribute to this equilibrium. Several contemporary therapies used in treating psychiatric disorders, regulate this GABA-glutamate balance. Yoga has been studied as an adjuvant treatment across a broad range of psychiatric disorders and is shown to have short-term therapeutic gains. Emerging evidence from recent clinical in vivo experiments suggests that yoga improves GABA-mediated cortical-inhibitory tone and enhances peripheral oxytocin levels. This is likely to have a more controlled downstream response of the hypothalamo-pituitary-adrenal system by means of reduced cortisol release and hence a blunted sympathetic response to stress. Animal and early fetal developmental studies suggest an inter-dependent role of oxytocin and GABA in regulating social behaviors. In keeping with these observations, we propose an integrated neurobiological model to study the mechanisms of therapeutic benefits with yoga. Apart from providing a neuroscientific basis for applying a traditional system of practice in the clinical setting, this model can be used as a framework for studying yoga mechanisms in future clinical trials.
Collapse
|
37
|
Chronic ethanol forced administration from adolescence to adulthood reduces cell density in the rat spinal cord. Tissue Cell 2018; 55:77-82. [PMID: 30503063 DOI: 10.1016/j.tice.2018.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/15/2018] [Accepted: 10/20/2018] [Indexed: 11/21/2022]
Abstract
Ethanol (EtOH) consumption is a risk factor for central nervous system damage, especially during adolescence. This study aimed to investigate the possible effects of chronic EtOH forced administration on gray and white matter of the spinal cord, from adolescence to adulthood. For this, male Wistar rats were administered EtOH by gavage (6.5 g/kg/day; 22.5% w/v) from the 35th to the 90th day of life, while control animals received only distilled water. After exposure, animals were euthanized and their spinal cords processed to obtain cervical and thoracic segments for histological analyses. Quantitative analyses of total cell density and motor neurons of white and gray matter from the ventral horns were evaluated. Forced EtOH administration model showed a decrease in the motoneuron density in the spinal cord in both segments evaluated. Analyses of total cell density showed that the cervical segment was more susceptible to damages promoted by EtOH, with a significant decrease in cell density. Our results showed that chronic EtOH exposure during adolescence could promote injuries to the spinal cord, with neurodegeneration of motoneurons and other cell types present in neural parenchyma.
Collapse
|
38
|
Neurobehavioral and biochemical modulation following administration of MgO and ZnO nanoparticles in the presence and absence of acute stress. Life Sci 2018; 203:72-82. [DOI: 10.1016/j.lfs.2018.04.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 04/12/2018] [Accepted: 04/15/2018] [Indexed: 12/11/2022]
|
39
|
Renicker MD, Cysewski N, Palmer S, Nakonechnyy D, Keef A, Thomas M, Magori K, Daberkow DP. Ameliorating Impact of Prophylactic Intranasal Oxytocin on Signs of Fear in a Rat Model of Traumatic Stress. Front Behav Neurosci 2018; 12:105. [PMID: 29892216 PMCID: PMC5985313 DOI: 10.3389/fnbeh.2018.00105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/07/2018] [Indexed: 12/17/2022] Open
Abstract
Oxytocin treatment reduces signs of long-term emotional stress after exposure to trauma; however, little is known about the potential protective effects of oxytocin treatment on behavioral and physiological changes associated with extreme stress exposure. The objective of this study was to investigate oxytocin treatment as a prophylactic measure against rat signs of fear. Two separate experiments were conducted in which the time of intranasal oxytocin administration differed. Intranasal oxytocin (1.0 μg/kg) was administered 5 min after daily exposure to foot shock in Experiment #1 and 1 h before foot shock in Experiment #2. In Experiment #1, possible massage-evoked oxytocin release (5 min after foot shock) was also investigated. In both experiments, a contextual fear conditioning procedure was employed in which stress was induced via inescapable foot shock (3 days, 40 shocks/day, 8 mA/shock) in a fear conditioning chamber. Male Sprague-Dawley rats (n = 24) were divided into four groups (n = 6, per group) for each experiment. Experiment #1 groups: Control Exp#1 (intranasal saline and no foot shock); Stress Exp#1 (intranasal saline 5 min after foot shock); Massage+Stress Exp#1 (massage-like stroking and intranasal saline 5 min after foot shock); Oxytocin+Stress Exp#1 (intranasal oxytocin 5 min after foot shock). Experiment #2 groups: Control Exp#2 (intranasal saline and no foot shock); Stress Exp#2 (intranasal saline 1 h before foot shock); Oxytocin Exp#2 (intranasal oxytocin and no foot shock); Oxytocin+Stress Exp#2 (intranasal oxytocin 1 h before foot shock). One week after fear conditioning (and other treatments), rats were independently evaluated for behavioral signs of fear. Two weeks after conditioning, physiological signs of fear were also assessed (Experiment #1). Relative to controls, rats treated with intranasal oxytocin 5 min after daily foot shock sessions exhibited significantly less immobility upon re-exposure to the shock chamber and attenuated physiological responses related to fear (e.g., elevated heart rate and blood pressure). Furthermore, intranasal oxytocin treatment given 1 h before daily foot shock sessions significantly decreased immobility and defecation upon re-exposure to the shock chamber, relative to controls. The results of this study suggest that prophylactic intranasal oxytocin, administered contemporaneously with aversive stimuli, mitigates behavioral and physiological responses associated with traumatic stress.
Collapse
Affiliation(s)
- Micah D Renicker
- Department of Biology, Eastern Washington University, Cheney, WA, United States
| | - Nicholas Cysewski
- Department of Biology, Eastern Washington University, Cheney, WA, United States
| | - Samuel Palmer
- Department of Biology, Eastern Washington University, Cheney, WA, United States
| | - Dmytro Nakonechnyy
- Department of Biology, Eastern Washington University, Cheney, WA, United States
| | - Andrew Keef
- Department of Biology, Eastern Washington University, Cheney, WA, United States
| | - Morgan Thomas
- Department of Biology, Eastern Washington University, Cheney, WA, United States
| | - Krisztian Magori
- Department of Biology, Eastern Washington University, Cheney, WA, United States
| | - David P Daberkow
- Department of Biology, Eastern Washington University, Cheney, WA, United States
| |
Collapse
|
40
|
Deslauriers J, Toth M, Der-Avakian A, Risbrough VB. Current Status of Animal Models of Posttraumatic Stress Disorder: Behavioral and Biological Phenotypes, and Future Challenges in Improving Translation. Biol Psychiatry 2018; 83:895-907. [PMID: 29338843 PMCID: PMC6085893 DOI: 10.1016/j.biopsych.2017.11.019] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 10/05/2017] [Accepted: 11/03/2017] [Indexed: 12/23/2022]
Abstract
Increasing predictability of animal models of posttraumatic stress disorder (PTSD) has required active collaboration between clinical and preclinical scientists. Modeling PTSD is challenging, as it is a heterogeneous disorder with ≥20 symptoms. Clinical research increasingly utilizes objective biological measures (e.g., imaging, peripheral biomarkers) or nonverbal behaviors and/or physiological responses to complement verbally reported symptoms. This shift toward more-objectively measurable phenotypes enables refinement of current animal models of PTSD, and it supports the incorporation of homologous measures across species. We reviewed >600 articles to examine the ability of current rodent models to probe biological phenotypes of PTSD (e.g., sleep disturbances, hippocampal and fear-circuit dysfunction, inflammation, glucocorticoid receptor hypersensitivity) in addition to behavioral phenotypes. Most models reliably produced enduring generalized anxiety-like or depression-like behaviors, as well as hyperactive fear circuits, glucocorticoid receptor hypersensitivity, and response to long-term selective serotonin reuptake inhibitors. Although a few paradigms probed fear conditioning/extinction or utilized peripheral immune, sleep, and noninvasive imaging measures, we argue that these should be incorporated more to enhance translation. Data on female subjects, on subjects at different ages across the life span, or on temporal trajectories of phenotypes after stress that can inform model validity and treatment study design are needed. Overall, preclinical (and clinical) PTSD researchers are increasingly incorporating homologous biological measures to assess markers of risk, response, and treatment outcome. This shift is exciting, as we and many others hope it not only will support translation of drug efficacy from animal models to clinical trials but also will potentially improve predictability of stage II for stage III clinical trials.
Collapse
Affiliation(s)
- Jessica Deslauriers
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, California
| | - Mate Toth
- Department of Behavioural Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andre Der-Avakian
- Department of Psychiatry, University of California San Diego, La Jolla, California
| | - Victoria B Risbrough
- Department of Psychiatry, University of California San Diego, La Jolla, California; Center of Excellence for Stress and Mental Health, Veterans Affairs Hospital, La Jolla, California.
| |
Collapse
|
41
|
Bridges NR, McKinley RA, Boeke D, Sherwood MS, Parker JG, McIntire LK, Nelson JM, Fletchall C, Alexander N, McConnell A, Goodyear C, Nelson JT. Single Session Low Frequency Left Dorsolateral Prefrontal Transcranial Magnetic Stimulation Changes Neurometabolite Relationships in Healthy Humans. Front Hum Neurosci 2018; 12:77. [PMID: 29632477 PMCID: PMC5879132 DOI: 10.3389/fnhum.2018.00077] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/12/2018] [Indexed: 11/13/2022] Open
Abstract
Background: Dorsolateral prefrontal cortex (DLPFC) low frequency repetitive transcranial magnetic stimulation (LF-rTMS) has shown promise as a treatment and investigative tool in the medical and research communities. Researchers have made significant progress elucidating DLPFC LF-rTMS effects—primarily in individuals with psychiatric disorders. However, more efforts investigating underlying molecular changes and establishing links to functional and behavioral outcomes in healthy humans are needed. Objective: We aimed to quantify neuromolecular changes and relate these to functional changes following a single session of DLPFC LF-rTMS in healthy participants. Methods: Eleven participants received sham-controlled neuronavigated 1 Hz rTMS to the region most activated by a 7-letter Sternberg working memory task (SWMT) within the left DLPFC. We quantified SWMT performance, functional magnetic resonance activation and proton Magnetic resonance spectroscopy (MRS) neurometabolite measure changes before and after stimulation. Results: A single LF-rTMS session was not sufficient to change DLPFC neurometabolite levels and these changes did not correlate with DLPFC activation changes. Real rTMS, however, significantly altered neurometabolite correlations (compared to sham rTMS), both with baseline levels and between the metabolites themselves. Additionally, real rTMS was associated with diminished reaction time (RT) performance improvements and increased activation within the motor, somatosensory and lateral occipital cortices. Conclusion: These results show that a single session of LF-rTMS is sufficient to influence metabolite relationships and causes widespread activation in healthy humans. Investigating correlational relationships may provide insight into mechanisms underlying LF-rTMS.
Collapse
Affiliation(s)
| | - Richard A McKinley
- Warfighter Interfaces Division, Applied Neuroscience Branch, Wright-Patterson AFB (WPAFB), Dayton, OH, United States
| | - Danielle Boeke
- Warfighter Interfaces Division, Applied Neuroscience Branch, Wright-Patterson AFB (WPAFB), Dayton, OH, United States
| | - Matthew S Sherwood
- Wright State Research Institute, Wright State University, Dayton, OH, United States
| | - Jason G Parker
- Kettering Health Network Innovation Center, Kettering, OH, United States
| | | | | | - Catherine Fletchall
- Grandview Medical Center, Kettering Health Network, Dayton, OH, United States
| | - Natasha Alexander
- Grandview Medical Center, Kettering Health Network, Dayton, OH, United States
| | - Amanda McConnell
- Grandview Medical Center, Kettering Health Network, Dayton, OH, United States
| | | | - Jeremy T Nelson
- Research Imaging Institute, School of Medicine, University of Texas Health Science Center, San Antonio, San Antonio, TX, United States
| |
Collapse
|
42
|
Stachowicz K. The role of DSCAM in the regulation of synaptic plasticity: possible involvement in neuropsychiatric disorders. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
43
|
Yang C, Gao J, Du J, Yang X, Jiang J. Altered Neuroendocrine Immune Responses, a Two-Sword Weapon against Traumatic Inflammation. Int J Biol Sci 2017; 13:1409-1419. [PMID: 29209145 PMCID: PMC5715524 DOI: 10.7150/ijbs.21916] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/23/2017] [Indexed: 12/22/2022] Open
Abstract
During the occurrence and development of injury (trauma, hemorrhagic shock, ischemia and hypoxia), the neuroendocrine and immune system act as a prominent navigation leader and possess an inter-system crosstalk between the reciprocal information dissemination. The fundamental reason that neuroendocrinology and immunology could mix each other and permeate toward the field of traumatology is owing to their same biological languages or chemical information molecules (hormones, neurotransmitters, neuropeptides, cytokines and their corresponding receptors) shared by the neuroendocrine and immune systems. The immune system is not only modulated by the neuroendocrine system, but also can modulate the biological functions of the neuroendocrine system. The interactive linkage of these three systems precipitates the complicated space-time patterns for the courses of traumatic inflammation. Recently, compelling evidence indicates that the network linkage pattern that initiating agents of neuroendocrine responses, regulatory elements of immune cells and effecter targets for immune regulatory molecules arouse the resistance mechanism disorders, which supplies the beneficial enlightenment for the diagnosis and therapy of traumatic complications from the view of translational medicine. Here we review the alternative protective and detrimental roles as well as possible mechanisms of the neuroendocrine immune responses in traumatic inflammation.
Collapse
Affiliation(s)
- Ce Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Jie Gao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Juan Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xuetao Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Jianxin Jiang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| |
Collapse
|
44
|
Lin S, Zhou G, Shao W, Fu Z. Impact of dexmedetomidine on amino acid contents and the cerebral ultrastructure of rats with cerebral ischemia-reperfusion injury. Acta Cir Bras 2017; 32:459-466. [PMID: 28700007 DOI: 10.1590/s0102-865020170060000006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/18/2017] [Indexed: 11/22/2022] Open
|
45
|
In vivo evaluation of the hippocampal glutamate, GABA and the BDNF levels associated with spatial memory performance in a rodent model of neuropathic pain. Physiol Behav 2017; 175:97-103. [DOI: 10.1016/j.physbeh.2017.03.025] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/18/2017] [Accepted: 03/18/2017] [Indexed: 11/22/2022]
|
46
|
Hajheidari S, Sameni HR, Bandegi AR, Miladi-gorji H. Effects of prolonged abstinence from METH on the hippocampal BDNF levels, neuronal numbers and apoptosis in methamphetamine-sensitized rats. Neurosci Lett 2017; 645:80-85. [DOI: 10.1016/j.neulet.2017.02.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 02/06/2017] [Accepted: 02/20/2017] [Indexed: 02/02/2023]
|
47
|
Naud A, Schmitt E, Wirth M, Hascoet JM. Determinants of Indices of Cerebral Volume in Former Very Premature Infants at Term Equivalent Age. PLoS One 2017; 12:e0170797. [PMID: 28125676 PMCID: PMC5268368 DOI: 10.1371/journal.pone.0170797] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/11/2017] [Indexed: 01/07/2023] Open
Abstract
Conventional magnetic resonance imaging (MRI) at term equivalent age (TEA) is suggested to be a reliable tool to predict the outcome of very premature infants. The objective of this study was to determine simple reproducible MRI indices, in premature infants and to analyze their neonatal determinants at TEA. A cohort of infants born before 32 weeks gestational age (GA) underwent a MRI at TEA in our center. Two axial images (T2 weighted), were chosen to realize nine measures. We defined 4 linear indices (MAfhlv: thickness of lateral ventricle; CSI: cortex-skull index; VCI: ventricular-cortex index; BOI: bi occipital index) and 1 surface index (VS.A: volume slice area). Perinatal data were recorded. Sixty-nine infants had a GA (median (interquartile range)) of 30.0 weeks GA (27.0; 30.0) and a birth weight of 1240 grams (986; 1477). MRI was done at 41.0 (40.0; 42.0) weeks post menstrual age (PMA). The inter-investigator reproducibility was good. Twenty one MRI (30.5%) were quoted abnormal. We observed an association with retinopathy of prematurity (OR [95CI] = 4.205 [1.231-14.368]; p = 0.017), surgery for patent ductus arteriosus (OR = 4.688 [1.01-21.89]; p = 0.036), early onset infection (OR = 4.688 [1.004-21.889]; p = 0.036) and neonatal treatment by cefotaxime (OR = 3.222 [1.093-9.497]; p = 0.03). There was a difference for VCI between normal and abnormal MRI (0.412 (0.388; 0.429) vs. 0.432 (0.418; 0.449); p = 0,019); BOI was higher when fossa posterior lesions were observed; VS.A seems to be the best surrogate for cerebral volume, 80% of VS.As' variance being explained by a multiple linear regression model including 7 variables (head circumference at birth and at TEA, PMA, dopamine, ibuprofen treatment, blood and platelets transfusions). These indices, easily and rapidly achievable, seem to be useful but need to be validated in a large population to allow generalization for diagnosis and follow-up of former premature infants.
Collapse
Affiliation(s)
- Aurelie Naud
- Department of Neonatology, Maternité Régionale, CHRU NANCY, France
| | | | - Maelle Wirth
- EA 3450 - DevAH, Université de Lorraine, Nancy, France
| | - Jean-Michel Hascoet
- Department of Neonatology, Maternité Régionale, CHRU NANCY, France
- EA 3450 - DevAH, Université de Lorraine, Nancy, France
| |
Collapse
|
48
|
Goddard AW. The Neurobiology of Panic: A Chronic Stress Disorder. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2017; 1:2470547017736038. [PMID: 32440580 PMCID: PMC7219873 DOI: 10.1177/2470547017736038] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 09/05/2017] [Accepted: 09/15/2017] [Indexed: 12/20/2022]
Abstract
Panic disorder is an often chronic and impairing human anxiety syndrome, which frequently results in serious psychiatric and medical comorbidities. Although, to date, there have been many advances in the diagnosis and treatment of panic disorder, its pathophysiology still remains to be elucidated. In this review, recent evidence for a neurobiological basis of panic disorder is reviewed with particular attention to risk factors such as genetic vulnerability, chronic stress, and temperament. In addition, neuroimaging data are reviewed which provides support for the concept of panic disorder as a fear network disorder. The potential impact of the National Institute of Mental Health Research Domain Criteria constructs of acute and chronic threats responses and their implications for the neurobiology of panic disorder are also discussed.
Collapse
Affiliation(s)
- Andrew W. Goddard
- UCSF Fresno Medical Education and
Research Program, University of California, San Francisco, USA
| |
Collapse
|
49
|
Kajimoto M, Ledee DR, Olson AK, Isern NG, Robillard-Frayne I, Des Rosiers C, Portman MA. Selective cerebral perfusion prevents abnormalities in glutamate cycling and neuronal apoptosis in a model of infant deep hypothermic circulatory arrest and reperfusion. J Cereb Blood Flow Metab 2016; 36:1992-2004. [PMID: 27604310 PMCID: PMC5094314 DOI: 10.1177/0271678x16666846] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/27/2016] [Indexed: 12/22/2022]
Abstract
Deep hypothermic circulatory arrest is often required for the repair of complex congenital cardiac defects in infants. However, deep hypothermic circulatory arrest induces neuroapoptosis associated with later development of neurocognitive abnormalities. Selective cerebral perfusion theoretically provides superior neural protection possibly through modifications in cerebral substrate oxidation and closely integrated glutamate cycling. We tested the hypothesis that selective cerebral perfusion modulates glucose utilization, and ameliorates abnormalities in glutamate flux, which occur in association with neuroapoptosis during deep hypothermic circulatory arrest. Eighteen infant male Yorkshire piglets were assigned randomly to two groups of seven (deep hypothermic circulatory arrest or deep hypothermic circulatory arrest with selective cerebral perfusion for 60 minutes at 18℃) and four control pigs without cardiopulmonary bypass support. Carbon-13-labeled glucose as a metabolic tracer was infused, and gas chromatography-mass spectrometry and nuclear magnetic resonance were used for metabolic analysis in the frontal cortex. Following 2.5 h of cerebral reperfusion, we observed similar cerebral adenosine triphosphate levels, absolute levels of lactate and citric acid cycle intermediates, and carbon-13 enrichment among three groups. However, deep hypothermic circulatory arrest induced significant abnormalities in glutamate cycling resulting in reduced glutamate/glutamine and elevated γ-aminobutyric acid/glutamate along with neuroapoptosis, which were all prevented by selective cerebral perfusion. The data suggest that selective cerebral perfusion prevents these modifications in glutamate/glutamine/γ-aminobutyric acid cycling and protects the cerebral cortex from apoptosis.
Collapse
Affiliation(s)
- Masaki Kajimoto
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Dolena R Ledee
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA
| | - Aaron K Olson
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA.,Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Nancy G Isern
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratories, Richland, WA, USA
| | | | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal and Montreal Heart Institute, Montréal, QC, Canada
| | - Michael A Portman
- Center for Developmental Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA .,Division of Cardiology, Department of Pediatrics, University of Washington, Seattle, WA, USA
| |
Collapse
|
50
|
Chen X, Zhou X, Lu D, Yang X, Zhou Z, Chen X, Chen Y, He W, Feng X. Aberrantly expressed long noncoding RNAs are involved in sevoflurane-induced developing hippocampal neuronal apoptosis: a microarray related study. Metab Brain Dis 2016; 31:1031-40. [PMID: 27234990 DOI: 10.1007/s11011-016-9838-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/16/2016] [Indexed: 12/23/2022]
Abstract
The commonly used volatile anesthetic sevoflurane has been shown to induce widespread apoptosis in the developing brain, yet the underlying molecular mechanisms are not fully understood. Accumulating research has demonstrated that long noncoding RNAs (lncRNAs) regulate multiple biological processes, including neural development, differentiation and apoptosis. They are aberrantly expressed in multiple neurodegenerative diseases. In this study, we employed a lncRNA-mRNA microarray analysis to determine whether and how lncRNAs are involved in sevoflurane-induced hippocampal neuronal apoptosis in neonatal mice. Our data showed that a single 6-h sevoflurane exposure of P7 mice resulted in significant morphological changes and apoptosis in the hippocampus. Moreover, the microarray simultaneously revealed 817 lncRNAs and 856 of their potential coding targets that related to apoptosis, of which 31 lncRNAs (19 up and 12 down) and 25 mRNAs were significantly differentially expressed (P < 0.05) after sevoflurane exposure. Importantly, we found that Bcl2l11 (BIM), which potentiates mitochondria-dependent apoptosis and its nearby enhancer-like lncRNA ENSMUST00000136025, were both more highly expressed in sevoflurane-treated samples compared with control samples. Subsequent qRT-PCR results confirmed the changes. Further CNC network indicated that lncRNA ENSMUST00000136025 was positively correlated with Bim. Moreover, sevoflurane induced a significant increase of pro-apoptotic protein BIM and Bax but a reduction of anti-apoptotic proteins Bcl-2 in the hippocampus. Our study first demonstrates that aberrantly expressed lncRNAs play a role in sevoflurane-induced hippocampal apoptosis. We noted that up-regulated ENSMUST00000136025 highly likely induced the over-expression of BIM, which eventually promoted mitochondria-mediated apoptosis. Such findings further broaden the understanding of molecular mechanisms responsible for sevoflurane-induced neurotoxicity.
Collapse
Affiliation(s)
- Xiaohui Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Fujian Provincial Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Xue Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Dihan Lu
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Xiaoyu Yang
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Zhibin Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Xi Chen
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Yanqing Chen
- Department of Anesthesiology, Fujian Provincial Hospital, Fujian Provincial Clinical Medical College, Fujian Medical University, Fuzhou, 350001, China
| | - Wen He
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China
| | - Xia Feng
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, No 58 zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|