1
|
Hou Y, Zuo Y, Song S, Zhang T. Long-term variable photoperiod exposure impairs hippocampal synapse involving of the glutamate system and leads to memory deficits in male Wistar rats. Exp Neurol 2025; 387:115191. [PMID: 39971149 DOI: 10.1016/j.expneurol.2025.115191] [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: 12/25/2024] [Revised: 02/07/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
Excessive artificial light at night can induce the human circadian misalignment, potentially impairing memory consolidation and the rhythms of hippocampal clock genes. To investigate the impact of circadian misalignment on hippocampal function, we measured various field excitatory postsynaptic potentials (fEPSP) and golgi staining in the CA1 and dentate gyrus (DG) regions in Wistar rats. Our findings revealed that circadian misalignment resulted in a leftward shift in the input-output (I-O) curve within the CA1 region, decreased long-term potentiation (LTP), multi-time interval paired-pulse ratio (PPR), as well as dendritic spines and complexity across both CA1 and DG regions. Additionally, magnetic resonance spectroscopy (MRS) showed that circadian misalignment downregulated glutamate-related neurotransmitters (Glu + Gln) in the hippocampus, contributing to impaired synaptic function. Furthermore, disruptions to glutamate receptor subunits due to circadian misalignment led to reduced expression of AMPA receptor and NMDA receptor subunits in the hippocampus. In summary, our results suggest that memory impairments resulting from circadian misalignment are associated with diminished functionality within the glutamatergic system; this includes reductions in both Glx levels and availability of glutamate receptor subunits-key factors contributing to compromised synaptic function within the hippocampus.
Collapse
Affiliation(s)
- Yuanyuan Hou
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yao Zuo
- Department of Neurology, Affiliated Hospital of Guizhou Medical University, 550004, China
| | - Shaofei Song
- Rehabilitation Medicine Center, West China Hospital, Sichuan University, 610041, China
| | - Tong Zhang
- Department of Neurological Rehabilitation, China Rehabilitation Research Center, Beijing Boai Hospital, Beijing, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, China; School of Rehabilitation, Capital Medical University, Beijing 100068, China.
| |
Collapse
|
2
|
Cheng P, Ding K, Chen D, Yang C, Wang J, Yang S, Chen M, Zhu G. mPFC DCC coupling with CaMKII + neuronal excitation participates in behavioral despair in male mice. Transl Psychiatry 2025; 15:52. [PMID: 39952936 PMCID: PMC11829057 DOI: 10.1038/s41398-025-03266-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025] Open
Abstract
A longed lack of control over harmful stimuli can lead to learned helplessness (LH), a significant factor in depression. However, the cellular and molecular mechanisms underlying LH, and eventually behavioral despair, remain largely unknown. The deleted in colorectal cancer (dcc) gene is associated with the risk of depression. However, the therapeutic potential and regulation mechanism of DCC in behavioral despair are still uncertain. In this study, we showed that depressive stimulators, including LH, lipopolysaccharide, and unpredictable chronic mild stress, triggered an elevation in DCC expression in the medial prefrontal cortex (mPFC). Additionally, elevated DCC expression in the mPFC was crucial in inducing behavioral despair, as evidenced by the induction of behavioral despair in normal mice and exacerbation of behavioral despair in LH mice upon DCC overexpression. By contrast, neutralizing DCC activity ameliorated LH-induced behavioral despair. Importantly, we elucidated that pathological DCC expression was attributable to the excessive excitation of CaMKII+ neurons in a manner dependent on the calpain-mediated degradation of SCOP and aberrant phosphorylation of the ERK signaling pathway. In addition, the increase in DCC expression led to a decreased excitability threshold in CaMKII+ neurons in the mPFC, which was supported by the observation that the ligand netrin 1 increased the frequency of action potential firing and of spontaneous excitatory postsynaptic currents in CaMKII+ neurons. In conclusion, our data indicate that LH triggers the excessive excitation of CaMKII+ neurons and activation of calpain-SCOP/ERK signaling to promote DCC expression, and DCC represents a crucial target for the treatment of LH-induced behavioral despair in male mice.
Collapse
Affiliation(s)
- Ping Cheng
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Keke Ding
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Daokang Chen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Chen Yang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Juan Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Shaojie Yang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Ming Chen
- MOE Frontier Center for Brain Science, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, China.
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China.
| |
Collapse
|
3
|
JianHua Z, Li M, Hu Q, Donoghue P, Jiang S, Li J, Li S, Ren X, Zhang Z, Du J, Yu Y, Chazot P, Lu C. CaMKIIα-TARPγ8 signaling mediates hippocampal synaptic impairment in aging. Aging Cell 2025; 24:e14349. [PMID: 39380368 PMCID: PMC11709088 DOI: 10.1111/acel.14349] [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: 03/14/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Aging-related decline in memory and synaptic function are associated with the dysregulation of calcium homeostasis, attributed to the overexpression of voltage-gated calcium channels (VGCC). The membrane insertion of AMPAR governed by the AMPAR auxiliary proteins is essential for synaptic transmission and plasticity (LTP). In this study, we demonstrated the hippocampal expression of the transmembrane AMPAR regulatory proteins γ-8 (TARPγ8) was reduced in aged mice along with the reduced CaMKIIα activity and memory impairment. We further showed that TARPγ8 expression was dependent on CaMKIIα activity. Inhibition of CaMKIIα activity significantly reduced the hippocampal TARPγ8 expression and CA3-CA1 LTP in young mice to a similar level to that of the aged mice. Furthermore, the knockdown of hippocampal TARPγ8 impaired LTP and memory in young mice, which mimicked the aging-related changes. We confirmed the enhanced hippocampal VGCC (Cav-1.3) expression in aged mice and found that inhibition of VGCC activity largely increased both p-CaMKIIα and TARPγ8 expression in aged mice, whereas inhibition of NMDAR or Calpains had no effect. In addition, we found that the exogenous expression of human TARPγ8 in the hippocampus in aged mice restored LTP and memory function. Collectively, these results indicate that the synaptic and cognitive impairment in aging is associated with the downregulation of CaMKIIα-TARPγ8 signaling caused by VGCC activation. Our results suggest that TARPγ8 may be a key molecular biomarker for brain aging and that boosting CaMKIIα-TARPγ8 signaling may be critical for the restoration of synaptic plasticity of aging and aging-related diseases.
Collapse
Affiliation(s)
- Zhao JianHua
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
| | - MingCan Li
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
- Institute of Psychiatry and Neuroscience, Xinxiang Medical UniversityXinxiangHenanChina
| | - Qilin Hu
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
- School of Medical EngineeringXinxiang Medical UniversityXinxiangChina
| | - Peter Donoghue
- Department of BiosciencesWolfson Research Institute for Health and Wellbeing, Durham UniversityDurhamUK
| | - Sanwei Jiang
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
| | - Junmei Li
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
- Institute of Psychiatry and Neuroscience, Xinxiang Medical UniversityXinxiangHenanChina
| | - Songji Li
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
| | - Xinyi Ren
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
| | - Ziyuan Zhang
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
| | - Jingzhi Du
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
| | - Yi Yu
- School of Medical EngineeringXinxiang Medical UniversityXinxiangChina
| | - Paul Chazot
- Department of BiosciencesWolfson Research Institute for Health and Wellbeing, Durham UniversityDurhamUK
| | - Chengbiao Lu
- Henan International Joint Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of NeurologyFirst Affiliated Hospital of Xinxiang Medical UniversityWeihui, XinxiangHenanChina
- Henan International Joint Laboratory of Non‐Invasive Neuromodulation, Department of Physiology and PathophysiologyXinxiang Medical UniversityXinxiangHenanChina
- Institute of Psychiatry and Neuroscience, Xinxiang Medical UniversityXinxiangHenanChina
| |
Collapse
|
4
|
Le AA, Lauterborn JC, Jia Y, Cox CD, Lynch G, Gall CM. Metabotropic NMDAR Signaling Contributes to Sex Differences in Synaptic Plasticity and Episodic Memory. J Neurosci 2024; 44:e0438242024. [PMID: 39424366 PMCID: PMC11638816 DOI: 10.1523/jneurosci.0438-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024] Open
Abstract
NMDA receptor (NMDAR)-mediated calcium influx triggers the induction and initial expression of long-term potentiation (LTP). Here we report that in male rodents, ion flux-independent (metabotropic) NMDAR signaling is critical for a third step in the production of enduring LTP, i.e., cytoskeletal changes that stabilize the activity-induced synaptic modifications. Surprisingly, females rely upon estrogen receptor alpha (ERα) for the metabotropic NMDAR operations used by males. Blocking NMDAR channels with MK-801 eliminated LTP expression in hippocampal field CA1 of both sexes but left intact theta burst stimulation (TBS)-induced actin polymerization within dendritic spines. A selective antagonist (Ro25-6981) of the NMDAR GluN2B subunit had minimal effects on synaptic responses but blocked actin polymerization and LTP consolidation in males only. Conversely, an ERα antagonist thoroughly disrupted TBS-induced actin polymerization and LTP in females while having no evident effect in males. In an episodic memory paradigm, Ro25-6981 prevented acquisition of spatial locations by males but not females, whereas an ERα antagonist blocked acquisition in females but not males. Sex differences in LTP consolidation were accompanied by pronounced differences in episodic memory in tasks involving minimal (for learning) cue sampling. Males did better on acquisition of spatial information whereas females had much higher scores than males on tests for acquisition of the identity of cues (episodic "what") and the order in which the cues were sampled (episodic "when"). We propose that sex differences in synaptic processes used to stabilize LTP result in differential encoding of the basic elements of episodic memory.
Collapse
Affiliation(s)
- Aliza A Le
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Julie C Lauterborn
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Yousheng Jia
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Conor D Cox
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
| | - Gary Lynch
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
- Psychiatry and Human Behavior, University of California, Irvine, California 92697
| | - Christine M Gall
- Departments of Anatomy and Neurobiology, University of California, Irvine, California 92697
- Neurobiology and Behavior, University of California, Irvine, California 92697
| |
Collapse
|
5
|
Jain A, Nakahata Y, Pancani T, Watabe T, Rusina P, South K, Adachi K, Yan L, Simorowski N, Furukawa H, Yasuda R. Dendritic, delayed, stochastic CaMKII activation in behavioural time scale plasticity. Nature 2024; 635:151-159. [PMID: 39385027 PMCID: PMC11540904 DOI: 10.1038/s41586-024-08021-8] [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: 08/08/2023] [Accepted: 09/05/2024] [Indexed: 10/11/2024]
Abstract
Behavioural time scale plasticity (BTSP) is non-Hebbian plasticity induced by integrating presynaptic and postsynaptic components separated by a behaviourally relevant time scale (seconds)1. BTSP in hippocampal CA1 neurons underlies place cell formation. However, the molecular mechanisms that enable synapse-specific plasticity on a behavioural time scale are unknown. Here we show that BTSP can be induced in a single dendritic spine using two-photon glutamate uncaging paired with postsynaptic current injection temporally separated by a behavioural time scale. Using an improved Ca2+/calmodulin-dependent kinase II (CaMKII) sensor, we did not detect CaMKII activation during this BTSP induction. Instead, we observed dendritic, delayed and stochastic CaMKII activation (DDSC) associated with Ca2+ influx and plateau potentials 10-100 s after BTSP induction. DDSC required both presynaptic and postsynaptic activity, which suggests that CaMKII can integrate these two signals. Also, optogenetically blocking CaMKII 15-30 s after the BTSP protocol inhibited synaptic potentiation, which indicated that DDSC is an essential mechanism of BTSP. IP3-dependent intracellular Ca2+ release facilitated both DDSC and BTSP. Thus, our study suggests that non-synapse-specific CaMKII activation provides an instructive signal with an extensive time window over tens of seconds during BTSP.
Collapse
Affiliation(s)
- Anant Jain
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
- Centre for High Impact Neuroscience and Translational Applications (CHINTA), TCG CREST, Kolkata, India
| | - Yoshihisa Nakahata
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Tristano Pancani
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Tetsuya Watabe
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Polina Rusina
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kelly South
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Kengo Adachi
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Long Yan
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA
| | - Noriko Simorowski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL, USA.
| |
Collapse
|
6
|
O'Connell A, Quinlan L, Kwakowsky A. β-amyloid's neurotoxic mechanisms as defined by in vitro microelectrode arrays: a review. Pharmacol Res 2024; 209:107436. [PMID: 39369863 DOI: 10.1016/j.phrs.2024.107436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/12/2024] [Accepted: 09/24/2024] [Indexed: 10/08/2024]
Abstract
Alzheimer's disease is characterized by the aggregation of β-amyloid, a pathological feature believed to drive the neuronal loss and cognitive decline commonly seen in the disease. Given the growing prevalence of this progressive neurodegenerative disease, understanding the exact mechanisms underlying this process has become a top priority. Microelectrode arrays are commonly used for chronic, non-invasive recording of both spontaneous and evoked neuronal activity from diverse in vitro disease models and to evaluate therapeutic or toxic compounds. To date, microelectrode arrays have been used to investigate β-amyloids' toxic effects, β-amyloids role in specific pathological features and to assess pharmacological approaches to treat Alzheimer's disease. The versatility of microelectrode arrays means these studies use a variety of methods and investigate different disease models and brain regions. This review provides an overview of these studies, highlighting their disparities and presenting the status of the current literature. Despite methodological differences, the current literature indicates that β-amyloid has an inhibitory effect on synaptic plasticity and induces network connectivity disruptions. β-amyloid's effect on spontaneous neuronal activity appears more complex. Overall, the literature corroborates the theory that β-amyloid induces neurotoxicity, having a progressive deleterious effect on neuronal signaling and plasticity. These studies also confirm that microelectrode arrays are valuable tools for investigating β-amyloid pathology from a functional perspective, helping to bridge the gap between cellular and network pathology and disease symptoms. The use of microelectrode arrays provides a functional insight into Alzheimer's disease pathology which will aid in the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Aoife O'Connell
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Ireland
| | - Leo Quinlan
- Physiology, School of Medicine, Regenerative Medicine Institute, University of Galway, Ireland
| | - Andrea Kwakowsky
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, University of Galway, Ireland.
| |
Collapse
|
7
|
Chen D, Wang J, Cao J, Zhu G. cAMP-PKA signaling pathway and anxiety: Where do we go next? Cell Signal 2024; 122:111311. [PMID: 39059755 DOI: 10.1016/j.cellsig.2024.111311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
Cyclic adenosine monophosphate (cAMP) is an intracellular second messenger that is derived from the conversion of adenosine triphosphate catalysed by adenylyl cyclase (AC). Protein kinase A (PKA), the main effector of cAMP, is a dimeric protein kinase consisting of two catalytic subunits and two regulatory subunits. When cAMP binds to the regulatory subunits of PKA, it leads to the dissociation and activation of PKA, which allows the catalytic subunit of PKA to phosphorylate target proteins, thereby regulating various physiological functions and metabolic processes in cellular function. Recent researches also implicate the involvement of cAMP-PKA signaling in the pathologenesis of anxiety disorder. However, there are still debates on the prevention and treatment of anxiety disorders from this signaling pathway. To review the function of cAMP-PKA signaling in anxiety disorder, we searched the publications with the keywords including "cAMP", "PKA" and "Anxiety" from Pubmed, Embase, Web of Science and CNKI databases. The results showed that the number of publications on cAMP-PKA pathway in anxiety disorder tended to increase. Bioinformatics results displayed a close association between the cAMP-PKA pathway and the occurrence of anxiety. Mechanistically, cAMP-PKA signaling could influence brain-derived neurotrophic factor and neuropeptide Y and participate in the regulation of anxiety. cAMP-PKA signaling could also oppose the dysfunctions of gamma-aminobutyric acid (GABA), intestinal flora, hypothalamic-pituitary-adrenal axis, neuroinflammation, and signaling proteins (MAPK and AMPK) in anxiety. In addition, chemical agents with the ability to activate cAMP-PKA signaling demonstrated therapy potential against anxiety disorders. This review emphasizes the central roles of cAMP-PKA signaling in anxiety and the targets of the cAMP-PKA pathway would be potential candidates for treatment of anxiety. Nevertheless, more laboratory investigations to improve the therapeutic effect and reduce the adverse effect, and continuous clinical research will warrant the drug development.
Collapse
Affiliation(s)
- Daokang Chen
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jingji Wang
- Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei 230061, China.
| | - Jian Cao
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
8
|
Wang J, Wang X, Yang J, Zhen Y, Ban W, Zhu G. Molecular profiling of a rat model of vascular dementia: Evidences from proteomics, metabolomics and experimental validations. Brain Res 2024; 1846:149254. [PMID: 39341485 DOI: 10.1016/j.brainres.2024.149254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/13/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Decrease of cerebral blood flow is the primary cause of vascular dementia (VD), but its pathophysiological mechanisms are still not known. This study aims to profile the molecular changes of a rat model of VD induced by bilateral common carotid artery ligation. The Morris water maze and new object recognition tasks were used to test the cognitive function of rats. Hematoxylin and Eosin (HE) staining was used to detect pathological changes in the hippocampus. After confirming the model, proteomics was used to detect differentially expressed proteins in the hippocampus, and metabolomics was used to detect differential metabolites in rat serum. Thereafter, bioinformatics were used to integrate and analyze the potential molecular profile. The results showed that compared with the sham control group, the spatial and recognition memory of the rats were significantly reduced, and pathological changes were observed in the hippocampal CA1 region of the model group. Proteomic analysis suggested 206 differentially expressed proteins in the hippocampus of VD rats, with 117 proteins upregulated and 89 downregulated. Protein-protein interaction network analysis suggested that those differentially expressed proteins might play crucial roles in lipid metabolism, cell adhesion, intracellular transport, and signal transduction. Metabolomics analysis identified 103 differential metabolites, and comparison with the human metabolome database revealed 22 common metabolites, which predicted 265 potential targets. Afterwards, by intersecting the predicted results from metabolomics with the differentially expressed proteins from proteomics, we identified five potential targets, namely ACE, GABBR1, Rock1, Abcc1 and Mapk10. Furthermore, western blotting confirmed that compared with control group, hippocampal GABBR1 and Rock1 were enhanced in the model group. Together, this study showed the molecular profile of VD rats through a combination of proteomics, metabolomics, and experimental confirmation methods, offering crucial molecular targets for the diagnosis and treatment of VD.
Collapse
Affiliation(s)
- Jingji Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China; Acupuncture and Moxibustion Clinical Medical Research Center of Anhui Province, The Second Affiliation Hospital of Anhui University of Chinese Medicine, Hefei 230061, China
| | - Xueqing Wang
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jun Yang
- The First Affiliation Hospital of Anhui University of Chinese Medicine, Hefei 230031, China.
| | - Yilan Zhen
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Wenming Ban
- Taihe County Hospital of Traditional Chinese Medicine, Fuyang 236600, China
| | - Guoqi Zhu
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
9
|
Stahl A, Tomchik SM. Modeling neurodegenerative and neurodevelopmental disorders in the Drosophila mushroom body. Learn Mem 2024; 31:a053816. [PMID: 38876485 PMCID: PMC11199955 DOI: 10.1101/lm.053816.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 05/01/2024] [Indexed: 06/16/2024]
Abstract
The common fruit fly Drosophila melanogaster provides a powerful platform to investigate the genetic, molecular, cellular, and neural circuit mechanisms of behavior. Research in this model system has shed light on multiple aspects of brain physiology and behavior, from fundamental neuronal function to complex behaviors. A major anatomical region that modulates complex behaviors is the mushroom body (MB). The MB integrates multimodal sensory information and is involved in behaviors ranging from sensory processing/responses to learning and memory. Many genes that underlie brain disorders are conserved, from flies to humans, and studies in Drosophila have contributed significantly to our understanding of the mechanisms of brain disorders. Genetic mutations that mimic human diseases-such as Fragile X syndrome, neurofibromatosis type 1, Parkinson's disease, and Alzheimer's disease-affect MB structure and function, altering behavior. Studies dissecting the effects of disease-causing mutations in the MB have identified key pathological mechanisms, and the development of a complete connectome promises to add a comprehensive anatomical framework for disease modeling. Here, we review Drosophila models of human neurodevelopmental and neurodegenerative disorders via the effects of their underlying mutations on MB structure, function, and the resulting behavioral alterations.
Collapse
Affiliation(s)
- Aaron Stahl
- Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Seth M Tomchik
- Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa 52242, USA
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, Iowa 52242, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa 52242, USA
- Hawk-IDDRC, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
10
|
Baudry M, Bi X. Revisiting the calpain hypothesis of learning and memory 40 years later. Front Mol Neurosci 2024; 17:1337850. [PMID: 38361744 PMCID: PMC10867166 DOI: 10.3389/fnmol.2024.1337850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/08/2024] [Indexed: 02/17/2024] Open
Abstract
In 1984, Gary Lynch and Michel Baudry published in Science a novel biochemical hypothesis for learning and memory, in which they postulated that the calcium-dependent protease, calpain, played a critical role in regulating synaptic properties and the distribution of glutamate receptors, thereby participating in memory formation in hippocampus. Over the following 40 years, much work has been done to refine this hypothesis and to provide convincing arguments supporting what was viewed at the time as a simplistic view of synaptic biochemistry. We have now demonstrated that the two major calpain isoforms in the brain, calpain-1 and calpain-2, execute opposite functions in both synaptic plasticity/learning and memory and in neuroprotection/neurodegeneration. Thus, calpain-1 activation is required for triggering long-term potentiation (LTP) of synaptic transmission and learning of episodic memory, while calpain-2 activation limits the magnitude of LTP and the extent of learning. On the other hand, calpain-1 is neuroprotective while calpain-2 is neurodegenerative, and its prolonged activation following various types of brain insults leads to neurodegeneration. The signaling pathways responsible for these functions have been identified and involve local protein synthesis, cytoskeletal regulation, and regulation of glutamate receptors. Human families with mutations in calpain-1 have been reported to have impairment in motor and cognitive functions. Selective calpain-2 inhibitors have been synthesized and clinical studies to test their potential use to treat disorders associated with acute neuronal damage, such as traumatic brain injury, are being planned. This review will illustrate the long and difficult journey to validate a bold hypothesis.
Collapse
Affiliation(s)
- Michel Baudry
- Western University of Health Sciences, Pomona, CA, United States
| | | |
Collapse
|
11
|
Wang J, Cheng P, Qu Y, Zhu G. Astrocytes and Memory: Implications for the Treatment of Memory-related Disorders. Curr Neuropharmacol 2024; 22:2217-2239. [PMID: 38288836 PMCID: PMC11337689 DOI: 10.2174/1570159x22666240128102039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/29/2023] [Indexed: 08/23/2024] Open
Abstract
Memory refers to the imprint accumulated in the brain by life experiences and represents the basis for humans to engage in advanced psychological activities such as thinking and imagination. Previously, research activities focused on memory have always targeted neurons. However, in addition to neurons, astrocytes are also involved in the encoding, consolidation, and extinction of memory. In particular, astrocytes are known to affect the recruitment and function of neurons at the level of local synapses and brain networks. Moreover, the involvement of astrocytes in memory and memory-related disorders, especially in Alzheimer's disease (AD) and post-traumatic stress disorder (PTSD), has been investigated extensively. In this review, we describe the unique contributions of astrocytes to synaptic plasticity and neuronal networks and discuss the role of astrocytes in different types of memory processing. In addition, we also explore the roles of astrocytes in the pathogenesis of memory-related disorders, such as AD, brain aging, PTSD and addiction, thus suggesting that targeting astrocytes may represent a potential strategy to treat memory-related neurological diseases. In conclusion, this review emphasizes that thinking from the perspective of astrocytes will provide new ideas for the diagnosis and therapy of memory-related neurological disorders.
Collapse
Affiliation(s)
- Juan Wang
- Key Laboratory of Xin’an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Ping Cheng
- Key Laboratory of Xin’an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yan Qu
- Key Laboratory of Xin’an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Guoqi Zhu
- Key Laboratory of Xin’an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
12
|
Lassers SB, Vakilna YS, Tang WC, Brewer GJ. The flow of axonal information among hippocampal sub-regions 2: patterned stimulation sharpens routing of information transmission. Front Neural Circuits 2023; 17:1272925. [PMID: 38144878 PMCID: PMC10739322 DOI: 10.3389/fncir.2023.1272925] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/20/2023] [Indexed: 12/26/2023] Open
Abstract
The sub-regions of the hippocampal formation are essential for episodic learning and memory formation, yet the spike dynamics of each region contributing to this function are poorly understood, in part because of a lack of access to the inter-regional communicating axons. Here, we reconstructed hippocampal networks confined to four subcompartments in 2D cultures on a multi-electrode array that monitors individual communicating axons. In our novel device, somal, and axonal activity was measured simultaneously with the ability to ascertain the direction and speed of information transmission. Each sub-region and inter-regional axons had unique power-law spiking dynamics, indicating differences in computational functions, with abundant axonal feedback. After stimulation, spiking, and burst rates decreased in all sub-regions, spikes per burst generally decreased, intraburst spike rates increased, and burst duration decreased, which were specific for each sub-region. These changes in spiking dynamics post-stimulation were found to occupy a narrow range, consistent with the maintenance of the network at a critical state. Functional connections between the sub-region neurons and communicating axons in our device revealed homeostatic network routing strategies post-stimulation in which spontaneous feedback activity was selectively decreased and balanced by decreased feed-forward activity. Post-stimulation, the number of functional connections per array decreased, but the reliability of those connections increased. The networks maintained a balance in spiking and bursting dynamics in response to stimulation and sharpened network routing. These plastic characteristics of the network revealed the dynamic architecture of hippocampal computations in response to stimulation by selective routing on a spatiotemporal scale in single axons.
Collapse
Affiliation(s)
- Samuel Brandon Lassers
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
| | - Yash S. Vakilna
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Texas Institute of Restorative Neurotechnologies (TIRN), The University of Texas Health Science Center (UTHealth), Houston, TX, United States
| | - William C. Tang
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Department of Biomedical Engineering, National Taiwan University, Taipei, Taiwan
| | - Gregory J. Brewer
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Memory Impairments and Neurological Disorders (MIND) Institute, Center for Neuroscience of Learning and Memory, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
13
|
Su W, Liu Y, Lam A, Hao X, Baudry M, Bi X. Contextual fear memory impairment in Angelman syndrome model mice is associated with altered transcriptional responses. Sci Rep 2023; 13:18647. [PMID: 37903805 PMCID: PMC10616231 DOI: 10.1038/s41598-023-45769-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/24/2023] [Indexed: 11/01/2023] Open
Abstract
Angelman syndrome (AS) is a rare neurogenetic disorder caused by UBE3A deficiency and characterized by severe developmental delay, cognitive impairment, and motor dysfunction. In the present study, we performed RNA-seq on hippocampal samples from both wildtype (WT) and AS male mice, with or without contextual fear memory recall. There were 281 recall-associated differentially expressed genes (DEGs) in WT mice and 268 DEGs in AS mice, with 129 shared by the two genotypes. Gene ontology analysis showed that extracellular matrix and stimulation-induced response genes were prominently enriched in recall-associated DEGs in WT mice, while nuclear acid metabolism and tissue development genes were highly enriched in those from AS mice. Further analyses showed that the 129 shared DEGs belonged to nuclear acid metabolism and tissue development genes. Unique recall DEGs in WT mice were enriched in biological processes critical for synaptic plasticity and learning and memory, including the extracellular matrix network clustered around fibronectin 1 and collagens. In contrast, AS-specific DEGs were not enriched in any known pathways. These results suggest that memory recall in AS mice, while altering the transcriptome, fails to recruit memory-associated transcriptional programs, which could be responsible for the memory impairment in AS mice.
Collapse
Affiliation(s)
- Wenyue Su
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Yan Liu
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Aileen Lam
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA
| | - Xiaoning Hao
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA
| | - Michel Baudry
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA.
| |
Collapse
|
14
|
Mesa MH, Garcia GC, Hoerndli FJ, McCabe KJ, Rangamani P. Spine apparatus modulates Ca 2+ in spines through spatial localization of sources and sinks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.558941. [PMID: 37790389 PMCID: PMC10542496 DOI: 10.1101/2023.09.22.558941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Dendritic spines are small protrusions on dendrites in neurons and serve as sites of postsynaptic activity. Some of these spines contain smooth endoplasmic reticulum (SER), and sometimes an even further specialized SER known as the spine apparatus (SA). In this work, we developed a stochastic spatial model to investigate the role of the SER and the SA in modulating Ca 2+ dynamics. Using this model, we investigated how ryanodine receptor (RyR) localization, spine membrane geometry, and SER geometry can impact Ca 2+ transients in the spine and in the dendrite. Our simulations found that RyR opening is dependent on where it is localized in the SER and on the SER geometry. In order to maximize Ca 2+ in the dendrites (for activating clusters of spines and spine-spine communication), a laminar SA was favorable with RyRs localized in the neck region, closer to the dendrite. We also found that the presence of the SER without the laminar structure, coupled with RyR localization at the head, leads to higher Ca 2+ presence in the spine. These predictions serve as design principles for understanding how spines with an ER can regulate Ca 2+ dynamics differently from spines without ER through a combination of geometry and receptor localization. Highlights 1RyR opening in dendritic spine ER is location dependent and spine geometry dependent. Ca 2+ buffers and SERCA can buffer against runaway potentiation of spines even when CICR is activated. RyRs located towards the ER neck allow for more Ca 2+ to reach the dendrites. RyRs located towards the spine head are favorable for increased Ca 2+ in spines.
Collapse
|
15
|
Brown J, Camporesi E, Lantero-Rodriguez J, Olsson M, Wang A, Medem B, Zetterberg H, Blennow K, Karikari TK, Wall M, Hill E. Tau in cerebrospinal fluid induces neuronal hyperexcitability and alters hippocampal theta oscillations. Acta Neuropathol Commun 2023; 11:67. [PMID: 37095572 PMCID: PMC10127378 DOI: 10.1186/s40478-023-01562-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/03/2023] [Indexed: 04/26/2023] Open
Abstract
Alzheimer's disease (AD) and other tauopathies are characterized by the aggregation of tau into soluble and insoluble forms (including tangles and neuropil threads). In humans, a fraction of both phosphorylated and non-phosphorylated N-terminal to mid-domain tau species, are secreted into cerebrospinal fluid (CSF). Some of these CSF tau species can be measured as diagnostic and prognostic biomarkers, starting from early stages of disease. While in animal models of AD pathology, soluble tau aggregates have been shown to disrupt neuronal function, it is unclear whether the tau species present in CSF will modulate neural activity. Here, we have developed and applied a novel approach to examine the electrophysiological effects of CSF from patients with a tau-positive biomarker profile. The method involves incubation of acutely-isolated wild-type mouse hippocampal brain slices with small volumes of diluted human CSF, followed by a suite of electrophysiological recording methods to evaluate their effects on neuronal function, from single cells through to the network level. Comparison of the toxicity profiles of the same CSF samples, with and without immuno-depletion for tau, has enabled a pioneering demonstration that CSF-tau potently modulates neuronal function. We demonstrate that CSF-tau mediates an increase in neuronal excitability in single cells. We then observed, at the network level, increased input-output responses and enhanced paired-pulse facilitation as well as an increase in long-term potentiation. Finally, we show that CSF-tau modifies the generation and maintenance of hippocampal theta oscillations, which have important roles in learning and memory and are known to be altered in AD patients. Together, we describe a novel method for screening human CSF-tau to understand functional effects on neuron and network activity, which could have far-reaching benefits in understanding tau pathology, thus allowing for the development of better targeted treatments for tauopathies in the future.
Collapse
Affiliation(s)
- Jessica Brown
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Elena Camporesi
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
| | - Juan Lantero-Rodriguez
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
| | - Maria Olsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
| | - Alice Wang
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Blanca Medem
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
- UK Dementia Research Institute at UCL, London, WC1E 6BT, UK
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, WC1E 6BT, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin, Madison, WI, 53792, USA
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53792, USA
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
| | - Thomas K Karikari
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg, 43180, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, 43180, Mölndal, Sweden
- Department of Psychiatry, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Mark Wall
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Emily Hill
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
16
|
Mosleh M, Javan M, Fathollahi Y. The properties of long-term potentiation at SC-CA1/ TA-CA1 hippocampal synaptic pathways depends upon their input pathway activation patterns. IBRO Neurosci Rep 2023; 14:358-365. [PMID: 37020855 PMCID: PMC10067737 DOI: 10.1016/j.ibneur.2023.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/25/2023] [Indexed: 03/30/2023] Open
Abstract
Long-term potentiation (LTP) has been considered as a cellular mechanism of memory. Since the Schaffer collateral (SC) and temporoammonic (TA) inputs to CA1 are distinct synaptic pathways that could mediate different cognitive functions, this study was therefore aimed to separately study and compare the properties of LTP of these two synaptic pathways. In the current study we used slice electrophysiological methods to compare various properties of these two synaptic pathways in response to single, paired pulse stimulation, and to three standard protocols for inducing LTP: the high frequency electrical stimulation (HFS), theta-burst (TBS), and primed burst (PBs) stimulation. We found that the SC-CA1 synapses could produce bigger maximum synaptic responses than TA-CA1 synapses. In addition, we showed that paired-pulse ratios of the SC-CA1 synapses were higher than TA-CA1 synapses at certain inter-pulses intervals. Finally, we showed a higher LTP% was induced by PBs or TBS at the SC-CA1 synapse than the TA-CA1 synapse. Briefly, our findings suggest the differential basal synaptic transmission, paired-pulse evoked synaptic responses, and LTP exhibition of the hippocampal SC-CA1/ TA-CA1 synaptic pathways, which may rely on spontaneous and evoked activity pattern at the local circuit level.
Collapse
|
17
|
Ji M, Zhang Z, Gao F, Yang S, Wang J, Wang X, Zhu G. Curculigoside rescues hippocampal synaptic deficits elicited by PTSD through activating cAMP-PKA signaling. Phytother Res 2023; 37:759-773. [PMID: 36200803 DOI: 10.1002/ptr.7658] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 07/26/2022] [Accepted: 08/12/2022] [Indexed: 02/17/2023]
Abstract
Chronic traumatic stress results in various psychiatric disorders, especially posttraumatic stress disorder (PTSD). Previous study demonstrated that curculigoside (CUR) a component of Rhizoma Curculiginis prevented fear extinction and stress-induced depression-like behaviors. However, its effects on PTSD and the mechanisms are still not completely clear. In this study, we observed typical PTSD-like phenotypes, synaptic deficit, and reduction of BDNF/TrkB signaling pathway in mice receiving modified single prolonged stress and electrical stimulation (SPS&S). By contrast, systemic administration of CUR blocked PTSD-like phenotypes and synaptic deficits, including reduction of BDNF/TrkB signaling pathway, GluA1 and Arc expression. Importantly, CUR reversed the impairment of PKA signaling pathway elicited by PTSD. We further confirmed that the effects of CUR on synaptic function were through PKA signaling pathway, as H-89, an inhibitor of PKA blocked the effect of CUR on behavioral changes and BDNF/TrkB signaling pathway. Thereafter, we verified that CUR on synaptic function were through PKA pathway using direct intracerebral injection of CUR and H-89. Direct intracerebral injection of CUR activated PKA/CREB/BDNF/TrkB, which was blocked by H-89. Additionally, the docking results showed high binding energies of CUR with A2AR, AC, PRKACA, and PRKAR1A, which might indicate that CUR functions through regulating PKA signaling pathway. In conclusion, CUR prevented the behavioral changes and hippocampal synaptic deficits elicited by PTSD through activating cAMP-PKA signaling.
Collapse
Affiliation(s)
- Manman Ji
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Zhengrong Zhang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Feng Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Shaojie Yang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Juan Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Xuncui Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
18
|
Gao F, Wang J, Yang S, Ji M, Zhu G. Fear extinction induced by activation of PKA ameliorates anxiety-like behavior in PTSD mice. Neuropharmacology 2023; 222:109306. [PMID: 36341808 DOI: 10.1016/j.neuropharm.2022.109306] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/18/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022]
Abstract
Prolonged exposure (PE) therapy aiming to promote fear extinction is a useful treatment for post-traumatic stress disorder (PTSD). However, the mechanisms underlying fear extinction and effective methods used to promote fear extinction in PTSD are still lacking. In this study, we displayed dysfunctions of cyclic adenosine 3,5-monophosphate (cAMP)-protein kinase A (PKA), protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and calcium signaling in peripheral serum of PTSD patients using bioinformatics analysis. Later, we confirmed the dysfunctions of cAMP-PKA, AKT/mTOR and calcium signaling in the hippocampus of PTSD mice. Moreover, the reduction of calpain1 in the hippocampus enhanced fear memory acquisition. Single activation of PKA by systemic application of rolipram (ROL) or meglumine cyclic adenylate (M-cAMP) before re-exposure promoted fear extinction and improved anxiety-like behavior in PTSD mice. Moreover, systemic application of ROL before re-exposure improved hippocampal brain-derived neurotrophic factor (BDNF)/tyrosine kinase receptor B (TrkB) signaling and calpain1/AKT/mTOR signaling. Interestingly, the effects of activation of PKA could be partially blocked by TrkB antagonist, ANA-12 and mTOR inhibitor, RAPA. Finally, intranasal administration of ROL could also adjust the abnormality of fear memory and improve anxiety-like behaviors in PTSD mice. Collectively, activation of PKA could promote fear extinction, which correlated with the reduction of anxiety-like behavior. The mechanisms were related to the BDNF/TrkB and calpain1/AKT/mTOR signaling pathways. PKA activation might be a useful complementary therapy for PE in the symptom elimination of PTSD.
Collapse
Affiliation(s)
- Feng Gao
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Juan Wang
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Shaojie Yang
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Manman Ji
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, The Ministry of Education and Key Laboratory of Molecular Biology (Brain Diseases), Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
19
|
Ishola AO, Adetunji AE, Abanum IC, Adeyemi AA, Faleye CK, Martins JB, Ogbe NC, Ogundipe TC, Okewulonu KE, Okon UE, Ovbude DI, Akele RY, Omotade NT, Ajao MS. Datumetine Preferentially Upregulates N-methyl-D-aspartate Receptor Signalling Pathways in Different Brain Regions of Mice. Basic Clin Neurosci 2023; 14:103-116. [PMID: 37346877 PMCID: PMC10279986 DOI: 10.32598/bcn.2021.3397.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/15/2021] [Accepted: 06/30/2021] [Indexed: 06/23/2023] Open
Abstract
Introduction We previously reported that datumetine possesses binding affinity with N-methyl-D-aspartate receptor (NMDAR) and that 14-day exposure to datumetine altered NMDAR signaling by mimicking glutamate toxicity. Here, we investigated the potential neuroprotective effect of a single shot of a low dose of datumetine administration in BALB/c mice. Methods 30 male adult BALB/c mice were used for the study. The mice were randomly divided into three groups of ten mice each with an intraperitoneal injection of 0.1 mL of 10% DMSO for the Vehicle group, Datumetine group were administered 0.1 mg/kg body weight (bw) of datumetine and MK-801+Datumetine group were administered 0.5 mg/kg bw of MK-801 (to block NMDAR) followed by 0.1 mg/kg bw of datumetine after 30 minutes. 24 hours after administration, mice were euthanized in an isoflurane chamber followed by perfusion with 1X PBS. Brains were excised and stored at -20°C till further processing. Mice designated for IHC were further perfused with 4% PFA and brain excised and stored in 4% PFA till further processing. NMDAR signalling molecules expression was evaluated in frozen brain samples and the fixed brain samples were stained for neuron, vGlut and NMDAR subtypes. Results Relative to vehicle (Veh), datumetine downregulate calcium calmodulin kinase II alpha (CamKIIα) expression in the hippocampus and prefrontal cortex (PFC) but not in the cerebellum, cyclic AMP response element binding protein (CREB) was also upregulated only in the PFC but phosphorylated CREB (pCREB) was also upregulated in three brain regions observed, while brain-derived neurotrophic factor (BDNF) was only upregulated in hippocampus and PFC of Datumetine relative to vehicle (Veh). On the other hand, dizocilpine (MK-801) reversed some of the effects of datumetine in the observed brain regions. No major histological alterations were observed in the different brain regions immunohistochemically. Conclusion We conclude that a low dose of datumetine moderately enhances NMDAR activity. This showed the neuroprotective potentials of low datumetine exposure.
Collapse
Affiliation(s)
- Azeez Olakunle Ishola
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Adedeji Enitan Adetunji
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Isaac Chukwunwike Abanum
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Adesola Akorede Adeyemi
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Caleb Kenechukwu Faleye
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Jane Babale Martins
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Nnenna Chimdalu Ogbe
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | | | - Karen Ezichi Okewulonu
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Umo Emmanuel Okon
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Deborah Irenoise Ovbude
- Department of Anatomy, College of Medicine and Health Sciences, Afe Babalola University Ado-Ekiti, Nigeria
| | - Richard Yomi Akele
- Department of Medical Laboratory Science, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | | | | |
Collapse
|
20
|
Kagan BJ, Kitchen AC, Tran NT, Habibollahi F, Khajehnejad M, Parker BJ, Bhat A, Rollo B, Razi A, Friston KJ. In vitro neurons learn and exhibit sentience when embodied in a simulated game-world. Neuron 2022; 110:3952-3969.e8. [PMID: 36228614 DOI: 10.1016/j.neuron.2022.09.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/21/2022] [Accepted: 08/31/2022] [Indexed: 11/06/2022]
Abstract
Integrating neurons into digital systems may enable performance infeasible with silicon alone. Here, we develop DishBrain, a system that harnesses the inherent adaptive computation of neurons in a structured environment. In vitro neural networks from human or rodent origins are integrated with in silico computing via a high-density multielectrode array. Through electrophysiological stimulation and recording, cultures are embedded in a simulated game-world, mimicking the arcade game "Pong." Applying implications from the theory of active inference via the free energy principle, we find apparent learning within five minutes of real-time gameplay not observed in control conditions. Further experiments demonstrate the importance of closed-loop structured feedback in eliciting learning over time. Cultures display the ability to self-organize activity in a goal-directed manner in response to sparse sensory information about the consequences of their actions, which we term synthetic biological intelligence. Future applications may provide further insights into the cellular correlates of intelligence.
Collapse
Affiliation(s)
| | | | - Nhi T Tran
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Forough Habibollahi
- Department of Biomedical Engineering, The University of Melbourne, Parkville, Australia
| | - Moein Khajehnejad
- Department of Data Science and AI, Monash University, Melbourne, Australia
| | - Bradyn J Parker
- Department of Materials Science and Engineering, Monash University, Melbourne, VIC, Australia
| | - Anjali Bhat
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| | - Ben Rollo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Australia
| | - Adeel Razi
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK; Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia; Monash Biomedical Imaging, Monash University, Clayton, VIC, Australia; CIFAR Azrieli Global Scholars Program, CIFAR, Toronto, Canada
| | - Karl J Friston
- Wellcome Trust Centre for Neuroimaging, Institute of Neurology, University College London, London, UK
| |
Collapse
|
21
|
Emerging roles of PHLPP phosphatases in the nervous system. Mol Cell Neurosci 2022; 123:103789. [PMID: 36343848 DOI: 10.1016/j.mcn.2022.103789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/15/2022] [Accepted: 10/22/2022] [Indexed: 11/06/2022] Open
Abstract
It has been more than a decade since the discovery of a novel class of phosphatase, the Pleckstrin Homology (PH) domain Leucine-rich repeat Protein Phosphatases (PHLPP). Over time, they have been recognized as crucial regulators of various cellular processes, such as memory formation, cellular survival and proliferation, maintenance of circadian rhythm, and others, with any deregulation in their expression or cellular localization causing havoc in any cellular system. With the ever-growing number of downstream substrates across multiple tissue systems, a web is emerging wherein the central point is PHLPP. A slight nick in the normal signaling cascade of the two isoforms of PHLPP, namely PHLPP1 and PHLPP2, has been recently found to invoke a variety of neurological disorders including Alzheimer's disease, epileptic seizures, Parkinson's disease, and others, in the neuronal system. Improper regulation of the two isoforms has also been associated with various disease pathologies such as diabetes, cardiovascular disorders, cancer, musculoskeletal disorders, etc. In this review, we have summarized all the current knowledge about PHLPP1 (PHLPP1α and PHLPP1β) and PHLPP2 and their emerging roles in regulating various neuronal signaling pathways to pave the way for a better understanding of the complexities. This would in turn aid in providing context for the development of possible future therapeutic strategies.
Collapse
|
22
|
Sun J, Liu Y, Hao X, Baudry M, Bi X. Lack of UBE3A-Mediated Regulation of Synaptic SK2 Channels Contributes to Learning and Memory Impairment in the Female Mouse Model of Angelman Syndrome. Neural Plast 2022; 2022:3923384. [PMID: 36237484 PMCID: PMC9553421 DOI: 10.1155/2022/3923384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/16/2022] [Indexed: 11/29/2022] Open
Abstract
Angelman syndrome (AS) is a rare neurodevelopmental disorder characterized by severe developmental delay, motor impairment, language and cognition deficits, and often with increased seizure activity. AS is caused by deficiency of UBE3A, which is both an E3 ligase and a cofactor for transcriptional regulation. We previously showed that the small conductance potassium channel protein SK2 is a UBE3A substrate, and that increased synaptic SK2 levels contribute to impairments in synaptic plasticity and fear-conditioning memory, as inhibition of SK2 channels significantly improved both synaptic plasticity and fear memory in male AS mice. In the present study, we investigated UBE3a-mediated regulation of synaptic plasticity and fear-conditioning in female AS mice. Results from both western blot and immunofluorescence analyses showed that synaptic SK2 levels were significantly increased in hippocampus of female AS mice, as compared to wild-type (WT) littermates. Like in male AS mice, long-term potentiation (LTP) was significantly reduced while long-term depression (LTD) was enhanced at hippocampal CA3-CA1 synapses of female AS mice, as compared to female WT mice. Both alterations were significantly reduced by treatment with the SK2 inhibitor, apamin. The shunting effect of SK2 channels on NMDA receptor was significantly larger in female AS mice as compared to female WT mice. Female AS mice also showed impairment in both contextual and tone memory recall, and this impairment was significantly reduced by apamin treatment. Our results indicate that like male AS mice, female AS mice showed significant impairment in both synaptic plasticity and fear-conditioning memory due to increased levels of synaptic SK2 channels. Any therapeutic strategy to reduce SK2-mediated inhibition of NMDAR should be beneficial to both male and female patients.
Collapse
Affiliation(s)
- Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Yan Liu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Xiaoning Hao
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| | - Michel Baudry
- College of Dental Medicine, Western University of Health Sciences, Pomona, California 91766, USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766, USA
| |
Collapse
|
23
|
Bell MK, Holst MV, Lee CT, Rangamani P. Dendritic spine morphology regulates calcium-dependent synaptic weight change. J Gen Physiol 2022; 154:e202112980. [PMID: 35819365 PMCID: PMC9280073 DOI: 10.1085/jgp.202112980] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 05/28/2022] [Accepted: 06/07/2022] [Indexed: 01/14/2023] Open
Abstract
Dendritic spines act as biochemical computational units and must adapt their responses according to their activation history. Calcium influx acts as the first signaling step during postsynaptic activation and is a determinant of synaptic weight change. Dendritic spines also come in a variety of sizes and shapes. To probe the relationship between calcium dynamics and spine morphology, we used a stochastic reaction-diffusion model of calcium dynamics in idealized and realistic geometries. We show that despite the stochastic nature of the various calcium channels, receptors, and pumps, spine size and shape can modulate calcium dynamics and subsequently synaptic weight updates in a deterministic manner. Through a series of exhaustive simulations and analyses, we found that the calcium dynamics and synaptic weight change depend on the volume-to-surface area of the spine. The relationships between calcium dynamics and spine morphology identified in idealized geometries also hold in realistic geometries, suggesting that there are geometrically determined deterministic relationships that may modulate synaptic weight change.
Collapse
Affiliation(s)
- Miriam K. Bell
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA
| | - Maven V. Holst
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA
| | - Christopher T. Lee
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA
| |
Collapse
|
24
|
Gao F, Yang S, Wang J, Zhu G. cAMP-PKA cascade: An outdated topic for depression? Biomed Pharmacother 2022; 150:113030. [PMID: 35486973 DOI: 10.1016/j.biopha.2022.113030] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 11/02/2022] Open
Abstract
Depression is a common neuropsychiatric disorder characterized by persistent depressed mood and causes serious socioeconomic burden worldwide. Hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis, deficiency of monoamine transmitters, neuroinflammation and abnormalities of the gut flora are strongly associated with the onset of depression. The cyclic AMP (cAMP)/protein kinase A (PKA) cascade, a major cross-species cellular signaling pathway, is supposed as important player and regulator of depression onset by controlling synaptic plasticity, cytokinesis, transcriptional regulation and HPA axis. In the central nervous system, the cAMP-PKA cascade can dynamically shape neural circuits by enhancing synaptic plasticity, and affect K+ channels by phosphorylating Kir4.1, thereby regulating neuronal excitation. The reduction of cAMP-PKA cascade affects neuronal excitation as well as synaptic plasticity, ultimately leading to pathological outcome of depression, while activation of cAMP-PKA cascade would provide a rapid antidepressant effect. In this review, we proposed to reconsider the function of cAMP-PKA cascade, especially in the rapid antidepressant effect. Local activation or indirect activation of PKA through adjusting anchor proteins would provide new idea for acute treatment of depression.
Collapse
Affiliation(s)
- Feng Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Shaojie Yang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Juan Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
25
|
Wang J, Gao F, Cui S, Yang S, Gao F, Wang X, Zhu G. Utility of 7,8-dihydroxyflavone in preventing astrocytic and synaptic deficits in the hippocampus elicited by PTSD. Pharmacol Res 2022; 176:106079. [PMID: 35026406 DOI: 10.1016/j.phrs.2022.106079] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 02/07/2023]
Abstract
Astrocytic functions and brain-derived neurotrophic factor (BDNF)-tyrosine kinase receptor B (TrkB) signaling pathways are impaired in stress-related neuropsychiatric diseases. Previous studies have reported neuroprotective effects of 7,8-dihydroxyflavone (7,8-DHF), a TrkB activator. Here, we investigated the molecular mechanisms underlying pathogenesis of post-traumatic stress disorder (PTSD) using a modified single-prolonged stress (SPS&S) model and the potential beneficial effects of 7,8-DHF. SPS&S reduced the hippocampal expression of glial fibrillary acidic protein (GFAP), a marker of astrocytes, and induced morphological changes in astrocytes. From the perspective of synaptic function, the SPS&S model displayed reduced expression of BDNF, p-TrkB, postsynaptic density protein 95 (PSD95), AMPA receptor subunit GluR1 (GluA1), NMDA receptor subunit N2A/N2B ratio, calpain-1, phosphorylated protein kinase B (Akt) and phosphorylated mammalian target of rapamycin (mTOR) and conversely, higher phosphatase and tension homolog (PTEN) expression in the hippocampus. Acute or continuous intraperitoneal administration of 7,8-DHF (5 mg/kg) after SPS&S procedures prevented SPS&S-induced fear memory generalization and anxiety-like behaviors as well as abnormalities of hippocampal oscillations. Most importantly, 7,8-DHF attenuated SPS&S-induced abnormal BDNF-TrkB signaling and calpain-1-dependent cascade of synaptic deficits. Furthermore, treatment with a TrkB inhibitor completely blocked while an mTOR inhibitor partially blocked the effects of 7,8-DHF on behavioral changes of SPS&S model mice. Our collective findings suggest that 7,8-DHF effectively alleviates PTSD-like symptoms, including fear generalization and anxiety-like behavior, potentially by preventing astrocytic and synaptic deficits in the hippocampus through targeting of TrkB.
Collapse
Affiliation(s)
- Juan Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Feng Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Shuai Cui
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Shaojie Yang
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Fang Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Xuncui Wang
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education, Anhui University of Chinese Medicine, China; Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.
| |
Collapse
|
26
|
Yang S, Yin Z, Zhu G. A review of the functions of G protein-coupled estrogen receptor 1 in vascular and neurological aging. Eur J Pharmacol 2021; 908:174363. [PMID: 34297966 DOI: 10.1016/j.ejphar.2021.174363] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/11/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Aging-related diseases, especially vascular and neurological disorders cause huge economic burden. How to delay vascular and neurological aging is one of the insurmountable questions. G protein-coupled estrogen receptor 1 (GPER) has been extensively investigated in recent years due to its multiple biological responses. In this review, the function of GPER in aging-related diseases represented by vascular diseases, and neurological disorders were discussed. Apart from that, activation of GPER was also found to renovate the aging brain characterized by memory decline, but in a manner different from another two nuclear estrogen receptors estrogen receptor (ER)α and ERβ. This salutary effect would be better clarified from the aspects of synaptic inputs and transmission. Furthermore, we carefully described molecular mechanisms underpinning GPER-mediated effects. This review would update our understanding of GPER in the aging process. Targeting GPER may represent a promising strategy in the aging-related disorders.
Collapse
Affiliation(s)
- Shaojie Yang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Zhe Yin
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, China.
| |
Collapse
|
27
|
Baudry M, Su W, Seinfeld J, Sun J, Bi X. Role of Calpain-1 in Neurogenesis. Front Mol Biosci 2021; 8:685938. [PMID: 34212005 PMCID: PMC8239220 DOI: 10.3389/fmolb.2021.685938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/01/2021] [Indexed: 12/25/2022] Open
Abstract
While calpains have been implicated in neurogenesis for a long time, there is still little information regarding the specific contributions of various isoforms in this process. We took advantage of the availability of mutant mice with complete deletion of calpain-1 to analyze its contribution to neurogenesis. We first used the incorporation of BrdU in newly-generated cells in the subgranular zone of the dentate gyrus to determine the role of calpain-1 deletion in neuronal proliferation. Our results showed that the lack of calpain-1 decreased the rate of cell proliferation in adult hippocampus. As previously shown, it also decreased the long-term survival of newly-generated neurons. We also used data from previously reported RNA and miRNA sequencing analyses to identify differentially expressed genes in brain of calpain-1 knock-out mice related to cell division, cell migration, cell proliferation and cell survival. A number of differentially expressed genes were identified, which could play a significant role in the changes in neurogenesis in calpain-1 knock out mice. The results provide new information regarding the role of calpain-1 in neurogenesis and have implications for better understanding the pathologies associated with calpain-1 mutations in humans.
Collapse
Affiliation(s)
- Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Wenyue Su
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Jeffrey Seinfeld
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
28
|
Zhuang Q, Fan M, Shen J, Chen Z, Xue D, Lu H, Xu R, He X, Hou J. Overexpression of Capns1 Predicts Poor Prognosis and Correlates with Tumor Progression in Renal Cell Carcinoma. Urol Int 2021; 105:697-704. [PMID: 33887737 DOI: 10.1159/000511638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 09/11/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Calpain small subunit 1 (Capns1) has shown its correlation with the metastasis and invasion of hepatocellular carcinoma and intrahepatic cholangiocarcinoma. However, the expression and function of Capns1 in human renal cell carcinoma (RCC) have not been clarified. This study aimed to examine the expression of Capns1 in RCC tissues and cell lines and to assess its role performed in RCC. METHODS Capns1 expression was evaluated in 75 pairs of RCC and matched adjacent non-tumor tissues by immunohistochemistry. The prognostic value of Capns1 in RCC was assessed by Kaplan-Meier and Cox regression analyses. The action of Capns1 in the proliferation, adhesion, migration, and invasion of RCC cells and the effects on matrix metalloproteinase (MMP) 2 and 9 expression were evaluated after Capns1 silence. RESULTS Capns1 expression was significantly higher in RCC tissues compared with the adjacent non-tumor tissues. Multivariate analysis showed that Capns1 overexpression was an independent poor prognostic marker in RCC. The silencing of Capns1 prohibited cell adhesion and impaired the migration and invasion ability of 786-O cells in vitro. Furthermore, Capns1 silence reduced MMP2 and MMP9 expression. CONCLUSION Capns1 overexpression predicts poor prognosis and correlates with tumor progression in RCC. Capns1 expression might serve a prognostic marker and therapeutic target for RCC.
Collapse
Affiliation(s)
- Qianfeng Zhuang
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Min Fan
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jie Shen
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhen Chen
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dong Xue
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hao Lu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Renfang Xu
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaozhou He
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jianquan Hou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
29
|
Hegemann RU, Abraham WC. Postsynaptic cell firing triggers bidirectional metaplasticity depending on the LTP induction protocol. J Neurophysiol 2021; 125:1624-1635. [PMID: 33760659 DOI: 10.1152/jn.00514.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cell firing has been reported to variably upregulate or downregulate subsequently induced long-term potentiation (LTP). The aim of this study was to elucidate the parameters critical to driving each direction of the metaplasticity effect. The main focus was on the commonly used θ-burst stimulation (TBS) and high-frequency stimulation (HFS) protocols that are known to trigger distinct intracellular signaling cascades. To study action potential (AP)-induced metaplasticity, we used intracellular recordings from CA1 pyramidal cells of rat hippocampal slices. Somatic current injections were used to induce θ-burst firing (TBF) or high-frequency firing (HFF) for priming purposes, whereas LTP was induced 15 min later via TBS of Schaffer collaterals in stratum radiatum. TBS-LTP was inhibited by both priming protocols. Conversely, HFS-LTP was facilitated by HFF priming but not affected by TBF priming. Interestingly, both priming protocols reduced AP firing during TBS-LTP induction, and this effect correlated with the reduction of TBS-LTP. However, LTP was not rescued by restoring AP firing with somatic current injections during the TBS. Analysis of intrinsic properties revealed few changes, apart from a priming-induced increase in the medium afterhyperpolarization (HFF priming) and a decrease in the EPSP amplitude/slope ratio (TBF priming), which could in principle contribute to the inhibition of TBS-LTP by reducing depolarization and associated Ca2+ influx following synaptic activity or AP backpropagation. Overall, these data indicate that the more physiological TBS protocol for inducing LTP is particularly susceptible to homeostatic feedback inhibition by prior bouts of postsynaptic cell firing.NEW & NOTEWORTHY The induction of LTP in the hippocampus was bidirectionally regulated by prior postsynaptic cell firing, with θ-burst stimulation-induced LTP being consistently impaired by prior spiking, whereas high-frequency stimulation-induced LTP was either not changed or facilitated. Reductions in cell firing during LTP induction did not explain the LTP impairment. Overall, different patterns of postsynaptic firing induce distinct intracellular changes that can increase or decrease LTP depending on the induction protocol.
Collapse
Affiliation(s)
- Regina U Hegemann
- Department of Psychology, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| | - Wickliffe C Abraham
- Department of Psychology, Brain Health Research Centre and Brain Research New Zealand, University of Otago, Dunedin, New Zealand
| |
Collapse
|
30
|
Lu C, Yang W, Zhou J, Zhang Z, Gong Y, Hu F, Yu W, Dong X. Inhibition of Pre-B Cell Colony Enhancing Factor Reduces Lung Injury in Rats Receiving Cardiopulmonary Bypass. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:51-60. [PMID: 33442236 PMCID: PMC7800440 DOI: 10.2147/dddt.s281554] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/17/2020] [Indexed: 11/30/2022]
Abstract
Objective Pre-B cell colony enhancing factor (PBEF) is an important proinflammatory cytokine involved in acute lung injury. However, whether PBEF participates in lung injury caused by cardiopulmonary bypass (CPB) is still unknown. This study aimed to investigate the effects of silencing PBEF on lung injury and the sodium and water transport system in rats receiving CPB. Methods Morphological changes in lung tissues were evaluated using hematoxylin and eosin (H&E) staining. PBEF was detected using immunohistochemistry. The sodium and water transport system-related proteins and cellular signaling pathways were detected by Western blotting. Results Rats receiving CPB (model group) had more severe alveolar wall damage and higher expression of PBEF in free form than the control rats. Western blotting showed that the expression of PBEF, surfactant protein D (SP), aquaporin (AQP) 1, AQP5, and epithelial sodium channel (ENaC) was significantly higher in the lung tissue of CPB rats than control rats. By contrast, adenovirus-encoding sh-PBEF significantly reduced the expression of PBEF, SP, AQP1, AQP5, and ENaC in the lung tissues of rats treated with CPB. The phosphorylation levels of extracellular signal-regulated protein kinases 1 and 2 (ERK1/2), protein kinase B (AKT), and p38 mitogen-activated protein kinase (MAPK) were significantly increased in the lung tissue of rats that received CPB, and were downregulated by adenovirus-encoding sh-PBEF. Conclusion Adenovirus-encoding sh-PBEF could reduce lung injury and repair the sodium–water transport system in rats receiving CPB, likely through reducing MAPK, ERK1/2, and Akt signaling pathways.
Collapse
Affiliation(s)
- Chao Lu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Wei Yang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Jianliang Zhou
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Zulei Zhang
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Yi Gong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Fajia Hu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Wenpeng Yu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| | - Xiao Dong
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, People's Republic of China
| |
Collapse
|
31
|
Wang Y, Liu Y, Bi X, Baudry M. Calpain-1 and Calpain-2 in the Brain: New Evidence for a Critical Role of Calpain-2 in Neuronal Death. Cells 2020; 9:E2698. [PMID: 33339205 PMCID: PMC7765587 DOI: 10.3390/cells9122698] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 01/24/2023] Open
Abstract
Calpains are a family of soluble calcium-dependent proteases that are involved in multiple regulatory pathways. Our laboratory has focused on the understanding of the functions of two ubiquitous calpain isoforms, calpain-1 and calpain-2, in the brain. Results obtained over the last 30 years led to the remarkable conclusion that these two calpain isoforms exhibit opposite functions in the brain. Calpain-1 activation is required for certain forms of synaptic plasticity and corresponding types of learning and memory, while calpain-2 activation limits the extent of plasticity and learning. Calpain-1 is neuroprotective both during postnatal development and in adulthood, while calpain-2 is neurodegenerative. Several key protein targets participating in these opposite functions have been identified and linked to known pathways involved in synaptic plasticity and neuroprotection/neurodegeneration. We have proposed the hypothesis that the existence of different PDZ (PSD-95, DLG and ZO-1) binding domains in the C-terminal of calpain-1 and calpain-2 is responsible for their association with different signaling pathways and thereby their different functions. Results with calpain-2 knock-out mice or with mice treated with a selective calpain-2 inhibitor indicate that calpain-2 is a potential therapeutic target in various forms of neurodegeneration, including traumatic brain injury and repeated concussions.
Collapse
Affiliation(s)
- Yubin Wang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (Y.W.); (Y.L.)
| | - Yan Liu
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (Y.W.); (Y.L.)
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA;
| | - Michel Baudry
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, USA; (Y.W.); (Y.L.)
| |
Collapse
|
32
|
Sun J, Liu Y, Baudry M, Bi X. SK2 channel regulation of neuronal excitability, synaptic transmission, and brain rhythmic activity in health and diseases. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118834. [PMID: 32860835 PMCID: PMC7541745 DOI: 10.1016/j.bbamcr.2020.118834] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/13/2020] [Accepted: 08/19/2020] [Indexed: 11/20/2022]
Abstract
Small conductance calcium-activated potassium channels (SKs) are solely activated by intracellular Ca2+ and their activation leads to potassium efflux, thereby repolarizing/hyperpolarizing membrane potential. Thus, these channels play a critical role in synaptic transmission, and consequently in information transmission along the neuronal circuits expressing them. SKs are widely but not homogeneously distributed in the central nervous system (CNS). Activation of SKs requires submicromolar cytoplasmic Ca2+ concentrations, which are reached following either Ca2+ release from intracellular Ca2+ stores or influx through Ca2+ permeable membrane channels. Both Ca2+ sensitivity and synaptic levels of SKs are regulated by protein kinases and phosphatases, and degradation pathways. SKs in turn control the activity of multiple Ca2+ channels. They are therefore critically involved in coordinating diverse Ca2+ signaling pathways and controlling Ca2+ signal amplitude and duration. This review highlights recent advances in our understanding of the regulation of SK2 channels and of their roles in normal brain functions, including synaptic plasticity, learning and memory, and rhythmic activities. It will also discuss how alterations in their expression and regulation might contribute to various brain disorders such as Angelman Syndrome, Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Jiandong Sun
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Yan Liu
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Michel Baudry
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, United States of America; Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America.
| |
Collapse
|
33
|
Liu A, Ji H, Ren Q, Meng Y, Zhang H, Collingride G, Xie W, Jia Z. The Requirement of the C-Terminal Domain of GluA1 in Different Forms of Long-Term Potentiation in the Hippocampus Is Age-Dependent. Front Synaptic Neurosci 2020; 12:588785. [PMID: 33192442 PMCID: PMC7661473 DOI: 10.3389/fnsyn.2020.588785] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/25/2020] [Indexed: 11/13/2022] Open
Abstract
Long-term potentiation (LTP) at glutamatergic synapses is an extensively studied form of long-lasting synaptic plasticity widely regarded as the cellular basis for learning and memory. At the CA1 synapse, there are multiple forms of LTP with distinct properties. Although AMPA glutamate receptors (AMPARs) are a key target of LTP expression, whether they are required in all forms of LTP remains unclear. To address this question, we have used our recently developed mouse line, GluA1C2KI, where the c-terminal domain (CTD) of the endogenous GluA1 is replaced by that of GluA2. Unlike traditional GluA1 global or conditional KO mice, GluA1C2KI mice have no changes in basal AMPAR properties or synaptic transmission allowing a better assessment of GluA1 in synaptic plasticity. We previously showed that these mice are impaired in LTP induced by high-frequency stimulation (HFS-LTP), but whether other forms of LTP are also affected in these mice is unknown. In this study, we compared various forms of LTP at CA1 synapses between GluA1C2KI and wild-type littermates by using several induction protocols. We show that HFS-LTP is impaired in both juvenile and adult GluA1C2KI mice. The LTP induced by theta-burst stimulation (TBS-LTP) is also abolished in juvenile GluA1C2KI mice. Interestingly, TBS-LTP can still be induced in adult GluA1C2KI mice, but its mechanisms are altered becoming more sensitive to protein synthesis and the extracellular signal-regulated kinase (ERK) inhibitors compared to wild type (WT) control. The GluA1C2KI mice are also differentially altered in several forms of LTP induced under whole-cell recording paradigms. These results indicate that the CTD of GluA1 is differentially involved in different forms of LTP at CA1 synapse highlighting the complexity and adaptative potential of LTP expression mechanisms in the hippocampus.
Collapse
Affiliation(s)
- An Liu
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Jiangsu Co-Innovation Center of Neuroregeneration, Southeast University, Nanjing, China
| | - Hong Ji
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Jiangsu Co-Innovation Center of Neuroregeneration, Southeast University, Nanjing, China
| | - Qiaoyun Ren
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Jiangsu Co-Innovation Center of Neuroregeneration, Southeast University, Nanjing, China
| | - Yanghong Meng
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Haiwang Zhang
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| | - Graham Collingride
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Wei Xie
- The Key Laboratory of Developmental Genes and Human Disease, Ministry of Education, School of Life Science and Technology, Jiangsu Co-Innovation Center of Neuroregeneration, Southeast University, Nanjing, China
| | - Zhengping Jia
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.,Neurosciences and Mental Health, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
34
|
Nadei OV, Khvorova IA, Agalakova NI. Cognitive Decline of Rats with Chronic Fluorosis Is Associated with Alterations in Hippocampal Calpain Signaling. Biol Trace Elem Res 2020; 197:495-506. [PMID: 31797207 DOI: 10.1007/s12011-019-01993-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 11/25/2019] [Indexed: 12/15/2022]
Abstract
The study was designed to evaluate an influence of excessive fluoride (F-) intake on cognitive capacities of adult rats and on proteins of memory-related calpain signaling in hippocampus. Control animals were given water with natural F- content of 0.4 ppm; rats from other groups consumed the same water supplemented with 5, 20, and 50 ppm F- (as NaF) for 12 months. The efficiency of learning and memory formation was evaluated by novel object recognition (NOR) and Morris water maze tests. The expression of enzymes of calpain-1 and calpain-2 signaling in hippocampus was detected by Western blotting. Excessive F- consumption had moderate impact on short-term memory, but impaired spatial learning and long-term memory of animals. Intoxication of rats with 5-50 ppm F- led to stimulation of calpain-1 in hippocampal cells and its translocation from cytosol to membranes, accompanied by activation of GTPase RhoA. Exposure to 20-50 ppm F- resulted in proteolytic cleavage of phosphatase PHLPP1 and increased expression of phospho-ERK1/2 kinase with insignificant decline of total ERK1/2 activity. In contrast, F- did not change the expression of calpain-2 and its substrates-phosphatase PTEN and kinase mTOR. However, F- intake led to downregulation of cAMP-response element binding protein (CREB) and brain-derived neurotrophic factor (BDNF). Thus, altered expression of calpain-1 and its downstream effectors at a background of stable activity of calpain-2 indicates overstimulation of signaling pathways of early LTP phase and disrupted link between early and late LTP phases, most probably due to altered activity of transcriptional and neurotrophic factors.
Collapse
Affiliation(s)
- Olga V Nadei
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez av., Saint Petersburg, Russia, 194223
| | - Irina A Khvorova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez av., Saint Petersburg, Russia, 194223
| | - Natalia I Agalakova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez av., Saint Petersburg, Russia, 194223.
| |
Collapse
|
35
|
Song Z, Shen F, Zhang Z, Wu S, Zhu G. Calpain inhibition ameliorates depression-like behaviors by reducing inflammation and promoting synaptic protein expression in the hippocampus. Neuropharmacology 2020; 174:108175. [DOI: 10.1016/j.neuropharm.2020.108175] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023]
|
36
|
Donkor IO. An update on the therapeutic potential of calpain inhibitors: a patent review. Expert Opin Ther Pat 2020; 30:659-675. [PMID: 32700591 DOI: 10.1080/13543776.2020.1797678] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Calpain is a cytosolic proteinase that regulates of a wide range of physiological functions. The enzyme has been implicated in various pathological conditions including neurodegenerative disorders, cardiovascular disorders, cancer, and several other diseases. Therefore, calpain inhibitors are of interest as therapeutic agents and have been studied in preclinical models of several diseases in which the enzyme has been implicated. AREAS COVERED Calpain inhibitors that were disclosed over the last 5 years (2015-2019) include calpastatin-based peptidomimetics; thalassospiramide lipopeptides; disulfide analogs of alpha-mercaptoacrylic acids; allosteric modulators; azoloimidazolidenones; and macrocyclic/non-macrocyclic carboxamides. The effectiveness of some of the inhibitors in preclinical animal models is discussed. EXPERT OPINION Significant milestones that were made over this time frame include: a) disclosure of novel blood-brain barrier (BBB) permeable calpastatin analogs as calpain inhibitors; b) disclosure that potent calpain inhibitors can be obtained by targeting the hydrophobic pockets on chain A of PEF(S) of the small subunit of calpain; c) use of PEF(S) (PDB ID: 4WQ2) in virtual screening to identify novel structurally diverse calpain inhibitors; and d) mitigation of the metabolic instability of the alpha-ketoamide warhead of calpain inhibitors.
Collapse
Affiliation(s)
- Isaac O Donkor
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, College of Pharmacy , Memphis, Tennessee, United States
| |
Collapse
|
37
|
Knockout of PINK1 altered the neural connectivity of Drosophila dopamine PPM3 neurons at input and output sites. INVERTEBRATE NEUROSCIENCE 2020; 20:11. [PMID: 32766952 DOI: 10.1007/s10158-020-00244-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 07/09/2020] [Indexed: 12/19/2022]
Abstract
Impairment of the dopamine system is the main cause of Parkinson disease (PD). PTEN-induced kinase 1 (PINK1) is possibly involved in pathogenesis of PD. However, its role in dopaminergic neurons has not been fully established yet. In the present investigation, we have used the PINK1 knockout Drosophila model to explore the role of PINK1 in dopaminergic neurons. Electrophysiological and behavioral tests indicated that PINK1 elimination enhances the neural transmission from the presynaptic part of dopaminergic neurons in the protocerebral posterior medial region 3 (PPM3) to PPM3 neurons (which are homologous to those in the substantia nigra in humans). Firing properties of the action potential in PPM3 neurons were also altered in the PINK1 knockout genotypes. Abnormal motor ability was also observed in these PINK1 knockout animals. Our results indicate that knockout of PINK1 could alter both the input and output properties of PPM3 neurons.
Collapse
|
38
|
Wang X, Xu W, Chen H, Li W, Li W, Zhu G. Astragaloside IV prevents Aβ 1-42 oligomers-induced memory impairment and hippocampal cell apoptosis by promoting PPARγ/BDNF signaling pathway. Brain Res 2020; 1747:147041. [PMID: 32739157 DOI: 10.1016/j.brainres.2020.147041] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/11/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023]
Abstract
Astragaloside IV (AS-IV), a natural product derived from Radix Astragali (Astragalus membranaceus), is beneficial for the treatment of Alzheimer's disease (AD), but the mechanisms underlying this benefit are not completely understood. Peroxisome proliferator-activated receptor gamma (PPARγ) and brain-derived neurotrophic factor (BDNF) are potential therapeutic targets for AD. In this study, we found that amyloid β protein fragment 1-42 oligomers (AβO) suppressed BDNF and PPARγ expression, and inhibited tyrosine receptor kinase B (TrkB) phosphorylation in cultured hippocampal neurons; these changes were ameliorated by treatment with AS-IV. Inhibition of PPARγ by genetic and pharmacological methods also blocked the effect of AS-IV on BDNF expression in AβO-treated cells. Importantly, exogenous BDNF protected against neurotoxicity and apoptosis induced by AβO, whereas inhibition of PPARγ reversed protective effects of AS-IV against these outcomes. In vivo data further revealed that AS-IV improved AβO-induced memory impairment and reduced apoptosis of hippocampal neurons. Moreover, AS-IV suppressed the AβO-induced reduction in BDNF by promoting PPARγ activation in the hippocampus. Taken together, these results indicate that AS-IV prevents AβO-induced memory impairment and hippocampal neuronal apoptosis, probably by promoting the PPARγ/BDNF signaling pathway.
Collapse
Affiliation(s)
- Xuncui Wang
- Department of Pharmacology, College of Basic Medicine, Anhui Medical University, Hefei 230032, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Experimental Center for Scientific Research, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Wen Xu
- Department of Neurology, The First Affiliated Hospital of University of Science and Technology of China, Anhui Provincial Hospital, Hefei 230001, China
| | - Hejuntao Chen
- Key Laboratory of Xin'an Medicine, Ministry of Education, Experimental Center for Scientific Research, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Weizu Li
- Department of Pharmacology, College of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Weiping Li
- Department of Pharmacology, College of Basic Medicine, Anhui Medical University, Hefei 230032, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Experimental Center for Scientific Research, Anhui University of Chinese Medicine, Hefei 230038, China.
| |
Collapse
|
39
|
Ahnaou A, Broadbelt T, Biermans R, Huysmans H, Manyakov NV, Drinkenburg WHIM. The phosphodiesterase-4 and glycine transporter-1 inhibitors enhance in vivo hippocampal theta network connectivity and synaptic plasticity, whereas D-serine does not. Transl Psychiatry 2020; 10:197. [PMID: 32555167 PMCID: PMC7303193 DOI: 10.1038/s41398-020-00875-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022] Open
Abstract
Dysfunctional N-methyl-D-aspartate receptors (NMDARs) and cyclic adenosine monophosphate (cAMP) have been associated with deficits in synaptic plasticity and cognition found in neurodegenerative and neuropsychiatric disorders such as Alzheimer's disease (AD) and schizophrenia. Therapeutic approaches that indirectly enhance NMDAR function through increases in glycine and/or D-serine levels as well as inhibition of phosphodiesterases that reduces degradation of cAMP, are expected to enhance synaptic strength, connectivity and to potentially impact cognition processes. The present in vivo study investigated effects of subcutaneous administration of D-serine, the glycine transporter 1 (GlyT1) inhibitor SSR504734 and the PDE4 inhibitor rolipram, on network oscillations, connectivity and long-term potentiation (LTP) at the hippocampi circuits in Sprague-Dawley rats. In conscious animals, multichannel EEG recordings assessed network oscillations and connectivity at frontal and hippocampal CA1-CA3 circuits. Under urethane anaesthesia, field excitatory postsynaptic potentials (fEPSPs) were measured in the CA1 subfield of the hippocampus after high-frequency stimulation (HFS) of the Schaffer collateral-CA1 (SC) pathway. SSR504734 and rolipram significantly increased slow theta oscillations (4-6.5 Hz) at the CA1-CA3, slow gamma oscillations (30-50 Hz) in the frontal areas and enhanced coherence in the CA1-CA3 network, which were dissociated from motor behaviour. SSR504734 enhanced short-term potentiation (STP) and fEPSP responses were extended into LTP response, whereas the potentiation of EPSP slope was short-lived to STP with rolipram. Unlike glycine, increased levels of D-serine had no effect on network oscillations and limits the LTP induction and expression. The present data support a facilitating role of glycine and cAMP on network oscillations and synaptic efficacy at the CA3-CA1 circuit in rats, whereas raising endogenous D-serine levels had no such beneficial effects.
Collapse
Affiliation(s)
- A. Ahnaou
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - T. Broadbelt
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - R. Biermans
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - H. Huysmans
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - N. V. Manyakov
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - W. H. I. M. Drinkenburg
- grid.419619.20000 0004 0623 0341Department of Neuroscience, Janssen Research & Development, A Division of Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
40
|
PKA and Ube3a regulate SK2 channel trafficking to promote synaptic plasticity in hippocampus: Implications for Angelman Syndrome. Sci Rep 2020; 10:9824. [PMID: 32555345 PMCID: PMC7299966 DOI: 10.1038/s41598-020-66790-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/04/2020] [Indexed: 12/29/2022] Open
Abstract
The ubiquitin ligase, Ube3a, plays important roles in brain development and functions, since its deficiency results in Angelman Syndrome (AS) while its over-expression increases the risk for autism. We previously showed that the lack of Ube3a-mediated ubiquitination of the Ca2+-activated small conductance potassium channel, SK2, contributes to impairment of synaptic plasticity and learning in AS mice. Synaptic SK2 levels are also regulated by protein kinase A (PKA), which phosphorylates SK2 in its C-terminal domain, facilitating its endocytosis. Here, we report that PKA activation restores theta burst stimulation (TBS)-induced long-term potentiation (LTP) in hippocampal slices from AS mice by enhancing SK2 internalization. While TBS-induced SK2 endocytosis is facilitated by PKA activation, SK2 recycling to synaptic membranes after TBS is inhibited by Ube3a. Molecular and cellular studies confirmed that phosphorylation of SK2 in the C-terminal domain increases its ubiquitination and endocytosis. Finally, PKA activation increases SK2 phosphorylation and ubiquitination in Ube3a-overexpressing mice. Our results indicate that, although both Ube3a-mediated ubiquitination and PKA-induced phosphorylation reduce synaptic SK2 levels, phosphorylation is mainly involved in TBS-induced endocytosis, while ubiquitination predominantly inhibits SK2 recycling. Understanding the complex interactions between PKA and Ube3a in the regulation of SK2 synaptic levels might provide new platforms for developing treatments for AS and various forms of autism.
Collapse
|
41
|
Chen QY, Zhang ZL, Liu Q, Chen CJ, Zhang XK, Xu PY, Zhuo M. Presynaptic long-term potentiation requires extracellular signal-regulated kinases in the anterior cingulate cortex. Mol Pain 2020; 16:1744806920917245. [PMID: 32264746 PMCID: PMC7144679 DOI: 10.1177/1744806920917245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Extracellular signal-regulated kinases are widely expressed protein kinases in neurons, which serve as important intracellular signaling molecules for central plasticity such as long-term potentiation. Recent studies demonstrate that there are two major forms of long-term potentiation in cortical areas related to pain: postsynaptic long-term potentiation and presynaptic long-term potentiation. In particular, presynaptic long-term potentiation in the anterior cingulate cortex has been shown to contribute to chronic pain-related anxiety. In this review, we briefly summarized the components and roles of extracellular signal-regulated kinases in neuronal signaling, especially in the presynaptic long-term potentiation of anterior cingulate cortex, and discuss the possible molecular mechanisms and functional implications in pain-related emotional disorders.
Collapse
Affiliation(s)
- Qi-Yu Chen
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Zhi-Ling Zhang
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qin Liu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chao-Jun Chen
- Department of Neurology, Guangzhou Chinese Medical Integrated Hospital (Huadu), Guangdong, China
| | - Xiao-Kang Zhang
- The First Affiliated Hospital of Gan-Nan Medical University, Ganzhopu, China
| | - Ping-Yi Xu
- Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min Zhuo
- Center for Neuron and Disease, Frontier Institutes of Science and Technology, Xi'an Jiaotong University, Xi'an, China.,Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Neurology, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
42
|
The Relationship Between Glutamate Dynamics and Activity-Dependent Synaptic Plasticity. J Neurosci 2020; 40:2793-2807. [PMID: 32102922 DOI: 10.1523/jneurosci.1655-19.2020] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 02/05/2020] [Accepted: 02/13/2020] [Indexed: 12/22/2022] Open
Abstract
The spatiotemporal dynamics of excitatory neurotransmission must be tightly regulated to achieve efficient synaptic communication. By limiting spillover, glutamate transporters are believed to prevent excessive activation of extrasynaptically located receptors that can impair synaptic plasticity. While glutamate transporter expression is reduced in numerous neurodegenerative diseases, the contributions of transporter dysfunction to disease pathophysiology remain ambiguous as the fundamental relationship between glutamate dynamics and plasticity, and the mechanisms linking these two phenomena, remain poorly understood. Here, we combined electrophysiology and real-time high-speed imaging of extracellular glutamate transients during LTP induction and characterized the sensitivity of the relationship between glutamate dynamics during theta burst stimulation (TBS) and the resulting magnitude of LTP consolidation, both in control conditions and following selective and nonselective glutamate transporter blockade. Glutamate clearance times were negatively correlated with LTP magnitude following nonselective glutamate transporter inhibition but not following selective blockade of a majority of GLT-1, the brain's most abundant glutamate transporter. Although glutamate transporter inhibition reduced the postsynaptic population response to TBS, calcium responses to TBS were greatly exaggerated. The source of excess calcium was dependent on NMDARs, L-type VGCCs, GluA2-lacking AMPARs, and internal calcium stores. Surprisingly, inhibition of L-type VGCCs, but not GluA2-lacking AMPARs or ryanodine receptors, was required to restore robust LTP. In all, these data provide a detailed understanding of the relationship between glutamate dynamics and plasticity and uncover important mechanisms by which poor glutamate uptake can negatively impact LTP consolidation.SIGNIFICANCE STATEMENT Specific patterns of neural activity can promote long-term changes in the strength of synaptic connections through a phenomenon known as synaptic plasticity. Synaptic plasticity is well accepted to represent the cellular mechanisms underlying learning and memory, and many forms of plasticity are initiated by the excitatory neurotransmitter glutamate. While essential for rapid cellular communication in the brain, excessive levels of extracellular glutamate can negatively impact brain function. In this study, we demonstrate that pharmacological manipulations that increase the availability of extracellular glutamate during neural activity can have profoundly negative consequences on synaptic plasticity. We identify mechanisms through which excess glutamate can negatively influence synaptic plasticity, and we discuss the relevance of these findings to neurodegenerative diseases and in the aging brain.
Collapse
|
43
|
Capone F, Pellegrino G, Motolese F, Rossi M, Musumeci G, Di Lazzaro V. Extremely Low Frequency Magnetic Fields Do Not Affect LTP-Like Plasticity in Healthy Humans. Front Hum Neurosci 2020; 14:14. [PMID: 32116603 PMCID: PMC7014826 DOI: 10.3389/fnhum.2020.00014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/14/2020] [Indexed: 11/17/2022] Open
Abstract
Introduction Several studies explored the biological effects of extremely low-frequency magnetic fields (ELF-MFs) in vitro, reporting the induction of functional changes in neuronal activity. In particular, ELF-MFs can influence synaptic plasticity both in vitro and in animal models but some studies reported an increase in long-term potentiation (LTP) whereas others suggested its reduction. However, no specific study has investigated such effect on humans. Aims To evaluate whether ELF-MFs affect the propensity of the human cortex to undergo LTP-like plasticity. Methods We designed a randomized, single-blind, sham-controlled, cross-over study on 10 healthy subjects. Cortical plasticity was induced by intermittent theta burst stimulation (iTBS) before and after 45-min ELF-MFs (75 Hz; 1.8 mT) or sham exposure and was estimated by measuring the changes of motor evoked potentials (MEP) amplitude before and after each iTBS. Results No adverse events were reported. No significant effects of ELF-MFs on cortical plasticity were found. Conclusion Whole-brain exposure to ELF-MFs (75 Hz; 1.8 mT) is safe and does not seem to significantly affect LTP-like plasticity in human motor cortex.
Collapse
Affiliation(s)
- Fioravante Capone
- Unit of Neurology, Neurophysiology, and Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy.,NeXT: Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit, Campus Bio-Medico University, Rome, Italy
| | - Giovanni Pellegrino
- Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Francesco Motolese
- Unit of Neurology, Neurophysiology, and Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy.,NeXT: Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit, Campus Bio-Medico University, Rome, Italy
| | - Mariagrazia Rossi
- Unit of Neurology, Neurophysiology, and Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy.,NeXT: Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit, Campus Bio-Medico University, Rome, Italy
| | - Gabriella Musumeci
- Unit of Neurology, Neurophysiology, and Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy.,NeXT: Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit, Campus Bio-Medico University, Rome, Italy
| | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, and Neurobiology, Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
44
|
Tyssowski KM, Gray JM. The neuronal stimulation-transcription coupling map. Curr Opin Neurobiol 2019; 59:87-94. [PMID: 31163285 PMCID: PMC6885097 DOI: 10.1016/j.conb.2019.05.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/06/2019] [Indexed: 12/17/2022]
Abstract
Neurons transcribe different genes in response to different extracellular stimuli, and these genes regulate neuronal plasticity. Thus, understanding how different stimuli regulate different stimulus-dependent gene modules would deepen our understanding of plasticity. To systematically dissect the coupling between stimulation and transcription, we propose creating a 'stimulation-transcription coupling map' that describes the transcription response to each possible extracellular stimulus. While we are currently far from having a complete map, recent genomic experiments have begun to facilitate its creation. Here, we describe the current state of the stimulation-transcription coupling map as well as the transcriptional regulation that enables this coupling.
Collapse
Affiliation(s)
- Kelsey M Tyssowski
- Harvard Medical School, Department of Genetics, 77 Ave Louis Pasteur, Boston, MA 02115, United States
| | - Jesse M Gray
- Harvard Medical School, Department of Genetics, 77 Ave Louis Pasteur, Boston, MA 02115, United States.
| |
Collapse
|
45
|
Yang SJ, Song ZJ, Wang XC, Zhang ZR, Wu SB, Zhu GQ. Curculigoside facilitates fear extinction and prevents depression-like behaviors in a mouse learned helplessness model through increasing hippocampal BDNF. Acta Pharmacol Sin 2019; 40:1269-1278. [PMID: 31028292 PMCID: PMC6786307 DOI: 10.1038/s41401-019-0238-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
Curculigoside (CUR) is the main active component of traditional Chinese medicine Curculigoorchioides Gaertn (Xianmao in Chinese), which exhibits a variety of pharmacological activities. In this study we investigated the effects of CUR on fear extinction and related depression-like behaviors in mice. In fear conditioning task, we found that administration of CUR (1.6, 8, 40 mg·kg-1·d-1, ip, for 7 days) did not affect memory consolidation, but CUR at higher doses (8, 40 mg·kg-1·d-1) significantly facilitated fear extinction, especially on D3 and D4. Moreover, CUR administration significantly ameliorated the fear conditioning-induced depression-like behaviors, likely through promoting fear extinction. We showed that CUR increased the expression of brain-derived neurotrophic factor (BDNF) and phosphorylation of tropomyosin receptor kinase B (TrkB) in the hippocampus, and activated protein kinase B (Akt)-mammalian target of the rapamycin (mTOR) signaling pathway. Administration of the selective TrkB agonist 7,8-dihydroxyflavone (7,8-DHF, 5 mg·kg-1·d-1, ip) also facilitated fear extinction, ameliorated depression-like behaviors. We established a mouse learned helplessness (LH) model to evaluate the antidepressant activity of CUR. The spatial memory was assessed in Morris water maze. We showed that LH-induced depression-like behaviors, including prolonged immobility times in forced swim and tail suspension tests as well as spatial memory impairments; LH also downregulated BDNF expression and the Akt-mTOR signaling pathway in the hippocampus. Administration of CUR (1.6, 8, 40 mg·kg-1·d-1, ip, for 14 days) or 7,8-DHF (5 mg·kg-1·d-1, ip, for 3 days) prevented LH-induced depression-like behaviors and promoted BDNF expression and the Akt-mTOR signaling pathway. In conclusion, CUR can accelerate the fear memory extinction and ameliorate depression-like behaviors in mice via promoting BDNF expression and activating the Akt-mTOR signaling pathway in the hippocampus.
Collapse
Affiliation(s)
- San-Juan Yang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Zhu-Jin Song
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Xun-Cui Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Zheng-Rong Zhang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Sheng-Bing Wu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, China
- Anhui Academy of Chinese Medicine, Hefei, 230038, China
| | - Guo-Qi Zhu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, China.
- Anhui Academy of Chinese Medicine, Hefei, 230038, China.
| |
Collapse
|
46
|
Tumor Necrosis Factor-α-Mediated Metaplastic Inhibition of LTP Is Constitutively Engaged in an Alzheimer's Disease Model. J Neurosci 2019; 39:9083-9097. [PMID: 31570539 DOI: 10.1523/jneurosci.1492-19.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 01/08/2023] Open
Abstract
LTP, a fundamental mechanism of learning and memory, is a highly regulated process. One form of regulation is metaplasticity (i.e., the activity-dependent and long-lasting changes in neuronal state that orchestrate the direction, magnitude, and persistence of future synaptic plasticity). We have previously described a heterodendritic metaplasticity effect, whereby strong high-frequency priming stimulation in stratum oriens inhibits subsequent LTP in the stratum radiatum of hippocampal area CA1, potentially by engagement of the enmeshed astrocytic network. This effect may occur due to neuron-glia interactions in response to priming stimulation that leads to the release of gliotransmitters. Here we found in male rats that TNFα and associated signal transduction enzymes, but not interleukin-1β (IL-1β), were responsible for mediating the metaplasticity effect. Replacing priming stimulation with TNFα incubation reproduced these effects. As TNFα levels are elevated in Alzheimer's disease, we examined whether heterodendritic metaplasticity is dysregulated in a transgenic mouse model of the disease, either before or after amyloid plaque formation. We showed that TNFα and IL-1β levels were significantly increased in aged but not young transgenic mice. Although control LTP was impaired in the young transgenic mice, it was not TNFα-dependent. In the older transgenic mice, however, LTP was impaired in a way that occluded further reduction by heterosynaptic metaplasticity, whereas LTP was entirely rescued by incubation with a TNFα antibody, but not an IL-1β antibody. Thus, TNFα mediates a heterodendritic metaplasticity in healthy rodents that becomes constitutively and selectively engaged in a mouse model of Alzheimer's disease.SIGNIFICANCE STATEMENT The proinflammatory cytokine TNFα is known to be capable of inhibiting LTP and is upregulated several-fold in brain tissue, serum, and CSF of Alzheimer's disease (AD) patients. However, the mechanistic roles played by TNFα in plasticity and AD remain poorly understood. Here we show that TNFα and its downstream signaling molecules p38 MAPK, ERK, and JNK contribute fundamentally to a long-range metaplastic inhibition of LTP in rats. Moreover, the impaired LTP in aged APP/PS1 mice is rescued by incubation with a TNFα antibody. Thus, there is an endogenous engagement of the metaplasticity mechanism in this mouse model of AD, supporting the idea that blocking TNFα might be of therapeutic benefit in the disease.
Collapse
|
47
|
Zheng Y, Ma XX, Dong L, Ma W, Cheng JH. Effects of uninterrupted sinusoidal LF-EMF stimulation on LTP induced by different combinations of TBS/HFS at the Schaffer collateral-CA1 of synapses. Brain Res 2019; 1725:146487. [PMID: 31580873 DOI: 10.1016/j.brainres.2019.146487] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 08/31/2019] [Accepted: 09/29/2019] [Indexed: 10/25/2022]
Abstract
Long-term potentiation (LTP) is an important aspect of synaptic plasticity and is one of the main mechanisms involved in memory. Low-frequency electromagnetic fields (LF-EMFs) such as transcranial magnetic stimulation are emerging neuromodulation tools for the regulation of LTP. However, whether LF-EMFs have different effects on different types of LTP has not yet been verified. Herein, we studied the regulatory effects of 15 Hz/2 mT sinusoidal magnetic field as pre-magnetic stimulation on several types of LTP, which were induced by theta-burst(TBS) or high-frequency stimulation (HFS) or some combination of them, and applied N-methyl-D-aspartate receptor(NMDAR) antagonists to observe the relationship between the regulation of LTP by LF-EMFs and NMDAR in the Schaffer collateral pathway of rat brain slices in vitro. The results presented in this paper are the performance of TBS and HFS was not exactly the same and the recovery speed of TBS-LTP was faster than HFS-LTP after receiving the regulation of LF-EMFs; moreover, the LTP level was affected by the order of combination and the effect of pre-magnetic stimulation could maintain the entire process of the combined induction experiment, while NMDAR antagonists could not completely offset the influence of LF-EMFs. The memory patterns are diverse, and this study has shown LF-EMFs can regulate LTP such as TBS-LTP and HFS-LTP and can continuously affect multiple LTP induction processes. However, different memory processes may have different performance in the face of LF-EMFs regulation. In terms of the mechanism of LF-EMFs-induced LTP regulation, NMDARs may be involved in the process of LF-EMF regulation of LTP, but are not the only factor.
Collapse
Affiliation(s)
- Yu Zheng
- School of Electronics and Information Engineering, Tianjin Polytechnic University, Tianjin 300387, China.
| | - Xiao-Xu Ma
- School of Electronics and Information Engineering, Tianjin Polytechnic University, Tianjin 300387, China
| | - Lei Dong
- State Key Laboratory of Precision Measurement Technology and Instruments, Tianjin University, Tianjin 300072, China
| | - Wei Ma
- School of Electronics and Information Engineering, Tianjin Polytechnic University, Tianjin 300387, China
| | - Jian-Hao Cheng
- School of Electronics and Information Engineering, Tianjin Polytechnic University, Tianjin 300387, China
| |
Collapse
|
48
|
Kwon HH, Neupane C, Shin J, Gwon DH, Yin Y, Shin N, Shin HJ, Hong J, Park JB, Yi Y, Kim DW, Kang JW. Calpain-2 as a Treatment Target in Prenatal Stress-induced Epileptic Spasms in Infant Rats. Exp Neurobiol 2019; 28:529-536. [PMID: 31495081 PMCID: PMC6751866 DOI: 10.5607/en.2019.28.4.529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/21/2019] [Accepted: 07/08/2019] [Indexed: 12/23/2022] Open
Abstract
Stress can induce a serious epileptic encephalopathy that occurs during early infancy. Recent studies have revealed that prenatal stress exposure is a risk factor for the development of infantile spasms. Our previous work demonstrates that prenatal stress with betamethasone-induced alterations to the expression of the K+/Cl- co-transporter (KCC2) in gamma-aminobutyric acid (GABA) interneurons lowers the seizure threshold in exposed animals. Here, we further investigated the mechanisms involved in this KCC2 dysfunction and explored possible treatment options. We stressed Sprague-Dawley rats prenatally and further treated dams with betamethasone on gestational day 15, which increases seizure susceptibility and NMDA (N-Methyl-D-aspartate)-triggered spasms on postnatal day 15. In this animal model, first, we evaluated baseline calpain activity. Second, we examined the cleavage and dephosphorylation of KCC2. Finally, we checked the effect of a calpain inhibitor on seizure occurrence. The phosphorylated-N-methyl-Daspartate Receptor 2B (NR2B):non-phosphorylated NR2B ratio was found to be higher in the cortex of the prenatally stressed betamethasone model. We further found that the betamethasone model exhibited increased phosphorylation of calpain-2 and decreased phosphorylation of KCC2 and Glutamic acid decarboxylase 67 (GAD67). After using a calpain inhibitor in prenatal-stress rats, the seizure frequency decreased, while latency increased. GABAergic depolarization was further normalized in prenatal-stress rats treated with the calpain inhibitor. Our study suggests that calpain-dependent cleavage and dephosphorylation of KCC2 decreased the seizure threshold of rats under prenatal stress. Calpain-2 functions might, thus, be targeted in the future for the development of treatments for epileptic spasms.
Collapse
Affiliation(s)
- Hyeok Hee Kwon
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 35015, Korea.,Department of Pediatrics, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Chiranjivi Neupane
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Department of Physiology, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Juhee Shin
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Do Hyeong Gwon
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Yuhua Yin
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Nara Shin
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 35015, Korea.,Brain Research Institute, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Jinpyo Hong
- Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 35015, Korea.,Brain Research Institute, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Jin Bong Park
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Brain Research Institute, School of Medicine, Chungnam National University, Daejeon 35015, Korea.,Department of Physiology, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - YoonYoung Yi
- Department of Pediatrics, Chungnam National University Hospital, Daejeon 35015, Korea
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Department of Anatomy, School of Medicine, Chungnam National University, Daejeon 35015, Korea.,Brain Research Institute, School of Medicine, Chungnam National University, Daejeon 35015, Korea
| | - Joon Won Kang
- Department of Medical Science, Chungnam National University, Daejeon 35015, Korea.,Brain Research Institute, School of Medicine, Chungnam National University, Daejeon 35015, Korea.,Department of Pediatrics, Chungnam National University Hospital, Daejeon 35015, Korea
| |
Collapse
|
49
|
Park H, Kaang BK. Balanced actions of protein synthesis and degradation in memory formation. ACTA ACUST UNITED AC 2019; 26:299-306. [PMID: 31416903 PMCID: PMC6699412 DOI: 10.1101/lm.048785.118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/02/2019] [Indexed: 11/24/2022]
Abstract
Storage of long-term memory requires not only protein synthesis but also protein degradation. In this article, we overview recent publications related to this issue, stressing that the balanced actions of protein synthesis and degradation are critical for long-term memory formation. We particularly focused on the brain-derived neurotrophic factor signaling that leads to protein synthesis; proteasome- and autophagy-dependent protein degradation that removes molecular constraints; the role of Fragile X mental retardation protein in translational suppression; and epigenetic modifications that control gene expression at the genomic level. Numerous studies suggest that an imbalance between protein synthesis and degradation leads to intellectual impairment and cognitive disorders.
Collapse
Affiliation(s)
- Hyungju Park
- Department of Structure and Function of Neural Network, Korea Brain Research Institute (KBRI), Daegu 41062, South Korea.,Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, South Korea
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| |
Collapse
|
50
|
Quirion JG, Parsons MP. The Onset and Progression of Hippocampal Synaptic Plasticity Deficits in the Q175FDN Mouse Model of Huntington Disease. Front Cell Neurosci 2019; 13:326. [PMID: 31379510 PMCID: PMC6650530 DOI: 10.3389/fncel.2019.00326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/03/2019] [Indexed: 12/22/2022] Open
Abstract
Huntington disease (HD) is an inherited neurodegenerative disease characterized by a clinical triad of motor, psychiatric and cognitive symptoms. HD is caused by a CAG repeat expansion in the gene encoding the huntingtin protein. Homozygosity for the HD-causing mutation is extremely rare; thus, the majority of HD patients express the mutant huntingtin protein in addition to reduced levels of the non-pathogenic huntingtin protein. Deficits in synaptic plasticity, including hippocampal long-term potentiation (LTP), have been identified in various mouse models of HD and are thought to contribute to the debilitating cognitive symptoms associated with the disease. However, the bulk of these studies used N-terminal fragment or homozygous knock-in mouse models of HD at symptomatic ages, and our understanding of the onset and progression of synaptic plasticity deficits in the HD brain is lacking. To better understand the time-course of synaptic plasticity deficits in HD, as well as the impact of heterozygous and homozygous huntingtin mutations, we quantified basal synaptic connectivity, presynaptic release probability, presynaptically mediated post-tetanic potentiation (PTP) and postsynaptically mediated LTP at presymptomatic, early symptomatic and late symptomatic ages in heterozygous and homozygous Q175FDN knock-in HD mice. Our results demonstrate clear age-dependent effects of the HD-causing mutation on both short and long-term plasticity that generally emerge earlier in homozygous mice. Interestingly, deficits in presynaptic short-term plasticity were more closely linked to disease progression than deficits in postsynaptic LTP, and heterozygous mice were more susceptible to an LTP deficit when induced by high frequency stimulation compared to theta burst stimulation. To the best of our knowledge, the present study represents the most thorough characterization to date of the onset and progression of hippocampal synaptic plasticity deficits in a mouse model of HD, and should prove valuable to future studies exploring cellular mechanisms underlying the debilitating cognitive decline in HD.
Collapse
Affiliation(s)
- Jade G Quirion
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Matthew P Parsons
- Division of Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|