1
|
Sun W, Yu H, Li X, Wan Y, Mei Y, Yang Y, An L. Subchronic cyanuric acid treatment impairs spatial flexible behavior in female adolescent rats through depressing GluN2B-dependent neuronal and synaptic function. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 289:117513. [PMID: 39662458 DOI: 10.1016/j.ecoenv.2024.117513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 09/13/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Subchronic exposure to cyanuric acid (CA) and its structural analogue melamine induces long-term effects on brain and behavior in male rodents. To examine if this exposure induced negative effects on cognitive function in females, we examined the behavioral performance and further attempted to investigate synaptic and neuronal function. CA was intraperitoneal treated with 20 or 40 mg/kg/day to adolescent female rats for 4 consecutive weeks. Multiple behavioral tests were employed to assess spatial cognition, learning strategy, locomotion and motivation. Hippocampal synaptic function at Schaffer collaterals-CA1 synapses and excitatory postsynaptic currents (EPSCs) in CA1 pyramidal neurons was evaluated. Meanwhile, the glutamate transport inhibitor DL-threo-β-benzyloxyaspartate (DL-TBOA) was infused into hippocampal CA1 region to certify the underlying mechanism. We found that subchronic CA exposure impairs reversal learning ability with dose-dependent effects but did not affect spatial learning and memory, or learning strategy. The expression and phosphorylation of N-methyl-D-aspartate receptor (NMDAR) GluN2B subunits were simultaneously reduced in the hippocampus and the GluN2B-mediated synaptic function, including long-term depression (LTD) and paired-pulse facilitation (PPF), was suppressed. CA could also diminish postsynaptic density protein-95 (PSD-95) expression but did change the levels of α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionate receptor (AMPAR) GluA1 or NMDAR GluN2A subunit, or hippocampal spine density. Meanwhile, CA depressed frequency and amplitude of GluN2B-mediated EPSCs, indicating the presynaptic and postsynaptic actions of CA on neuronal activity. Furthermore, the DL-TBOA infusions could effectively mitigate the diminished GluN2B-LTD and GluN2B-EPSCs and the impairments in behavioral flexibility. Our findings provide the first evidence that CA can exert neurotoxic effects on females and certify that one of the potential mechanisms for neuronal and synaptic dysfunction is the GluN2B-mediated signaling pathway.
Collapse
Affiliation(s)
- Wei Sun
- Department of Pediatrics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Haiyang Yu
- Department of Pediatrics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Xiaoliang Li
- Department of Chinese Medicine, Jinan Geriatric/Rehabilitation Hospital, Jinan 250013, China
| | - Yiwen Wan
- Department of Rehabilitation Medicine, Shenzhen Bao'an Hospital Affiliated of Southern Medical University, Shenzhen, Guangdong 518100, China
| | - Yazi Mei
- Graduate School of Guangzhou University of Chinese Medicine; Guangzhou 510006, China
| | - Yang Yang
- Department of Pediatrics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China
| | - Lei An
- Department of Pediatrics, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China; Department of Chinese Medicine, Jinan Geriatric/Rehabilitation Hospital, Jinan 250013, China; Graduate School of Guangzhou University of Chinese Medicine; Guangzhou 510006, China; Department of Proctology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, China.
| |
Collapse
|
2
|
Babaei P, Javer S, Abedinzade M. Therapeutic Effects Of Combined and Chronic Treatment of Tat-GluA23y and D-Serine on Cognitive Dysfunction in Postmenopausal Rats. Exp Aging Res 2024; 50:633-651. [PMID: 37660354 DOI: 10.1080/0361073x.2023.2254660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/30/2023] [Indexed: 09/05/2023]
Abstract
BACKGROUND The incidence of Alzheimer's disease (AD) in female gender compared with male has been addressed as a health concern, particularly in menopausal age. We here hypothesized that co-administration of NMDARs agonist (D-serine) and AMPARs endocytosis inhibitor (Tat-GluA23y) might be a potential target for alleviating memory impairment in sporadic Alzheimer model of rats. METHODS Forty-eight female Wistar rats weighing 200-220 randomly divided into six groups. One month later, ovariectomized rats underwent stereotaxic surgery and were cannulated into the brain lateral ventricles. Streptozotocin was injected (3 mg/kg), then animals received the related treatments until the day 51, which experienced acquisition of spatial memory in Morris Water Maze test. Finally, the level of phosphorylated cAMP response element binding protein (CREB) in the hippocampus was measured by Western blotting. RESULTS Co-administration of D-serine and GluA23y significantly enhanced the acquisition and retrieval of impaired spatial memory in ovariectomized rats with AD (p < .001). Compared to Glu-A 23, D-serine caused more improvement in the mentioned parameters above, however, these values for both groups were still significantly different from the control group (P < .05). CONCLUSION Simultaneous treatment with D-serine and GluA23y synergistically improved STZ induced spatial memory impairment in OVX rat, probably partly via increase in phosphorylated CREB protein.
Collapse
Affiliation(s)
- Parvin Babaei
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Cellular &Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of physiology, School of Medicine, Guilan university of medical science, Rasht, Iran
| | - Shirin Javer
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Cellular &Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of physiology, School of Medicine, Guilan university of medical science, Rasht, Iran
| | - Mahmood Abedinzade
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Department of physiology, School of Medicine, Guilan university of medical science, Rasht, Iran
- medical biotechnology research center, School of Paramedicine, Guilan university of medical sciences, Rasht, Iran
| |
Collapse
|
3
|
Wang Y, Zheng AN, Yang H, Wang Q, Dai B, Wang JJ, Wan YT, Liu ZB, Liu SY. Olfactory Three-Needle Electroacupuncture Improved Synaptic Plasticity and Gut Microbiota of SAMP8 Mice by Stimulating Olfactory Nerve. Chin J Integr Med 2024; 30:729-741. [PMID: 37999886 DOI: 10.1007/s11655-023-3614-3] [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] [Accepted: 06/07/2023] [Indexed: 11/25/2023]
Abstract
OBJECTIVE To investigate the effects and mechanisms of olfactory three-needle (OTN) electroacupuncture (EA) stimulation of the olfactory system on cognitive dysfunction, synaptic plasticity, and the gut microbiota in senescence-accelerated mouse prone 8 (SAMP8) mice. METHODS Thirty-six SAMP8 mice were randomly divided into the SAMP8 (P8), SAMP8+OTN (P8-OT), and SAMP8+nerve transection+OTN (P8-N-OT) groups according to a random number table (n=12 per group), and 12 accelerated senescence-resistant (SAMR1) mice were used as the control (R1) group. EA was performed at the Yintang (GV 29) and bilateral Yingxiang (LI 20) acupoints of SAMP8 mice for 4 weeks. The Morris water maze test, transmission electron microscopy, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) staining, Nissl staining, Golgi staining, Western blot, and 16S rRNA sequencing were performed, respectively. RESULTS Compared with the P8 group, OTN improved the cognitive behavior of SAMP8 mice, inhibited neuronal apoptosis, increased neuronal activity, and attenuated hippocampal synaptic dysfunction (P<0.05 or P<0.01). Moreover, the expression levels of synaptic plasticity-related proteins N-methyl-D-aspartate receptor 1 (NMDAR1), NMDAR2B, synaptophysin (SYN), and postsynaptic density protein-95 (PSD95) in hippocampus were increased by OTN treatment (P<0.05 or P<0.01). Furthermore, OTN greatly enhanced the brain-derived neurotrophic factor (BDNF)/cAMP-response element binding (CREB) signaling and phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) signaling compared with the P8 group (P<0.05 or P<0.01). However, the neuroprotective effect of OTN was attenuated by olfactory nerve truncation. Compared with the P8 group, OTN had a very limited effect on the fecal microbial structure and composition of SAMP8 mice, while specifically increased the genera Oscillospira and Sutterella (P<0.05). Interestingly, the P8-N-OT group showed an abnormal fecal microbiota with higher microbial α-diversity, Firmicutes/Bacteroidetes ratio and pathogenic bacteria (P<0.05 or P<0.01). CONCLUSIONS OTN improved cognitive deficits and hippocampal synaptic plasticity by stimulating the olfactory nerve and activating the BDNF/CREB and PI3K/AKT/mTOR signaling pathways. Although the gut microbiota was not the main therapeutic target of OTN for Alzheimer's disease, the olfactory nerve was essential to maintain the homeostasis of gut microbiota.
Collapse
Affiliation(s)
- Yuan Wang
- College of Acu-moxibustion and Massage, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
| | - A-Ni Zheng
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
- The Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712000, China
| | - Huan Yang
- Department of Traditional Chinese Medicine, Baotou Medical College, Baotou, Inner Mongolia Autonomous Region, 014040, China
| | - Qiang Wang
- College of Acu-moxibustion and Massage, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
| | - Biao Dai
- College of Acu-moxibustion and Massage, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
| | - Jia-Ju Wang
- College of Acu-moxibustion and Massage, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
| | - Yi-Tong Wan
- College of Acu-moxibustion and Massage, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
| | - Zhi-Bin Liu
- College of Acu-moxibustion and Massage, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
- Shaanxi Key Laboratory of Acupuncture and Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi Province, 712046, China
| | - Si-Yang Liu
- School of Basic Medical Sciences, Xi'an Medical University, Xi'an, 710021, China.
| |
Collapse
|
4
|
Cao JW, Liu LY, Yu YC. Gap junctions regulate the development of neural circuits in the neocortex. Curr Opin Neurobiol 2023; 81:102735. [PMID: 37263136 DOI: 10.1016/j.conb.2023.102735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/12/2023] [Accepted: 05/07/2023] [Indexed: 06/03/2023]
Abstract
Gap junctions between cells are ubiquitously expressed in the developing brain. They are involved in major steps of neocortical development, including neurogenesis, cell migration, synaptogenesis, and neural circuit formation, and have been implicated in cortical column formation. Dysfunctional gap junctions can contribute to or even cause a variety of brain diseases. Although the role of gap junctions in neocortical development is better known, a comprehensive understanding of their functions is far from complete. Here we explore several critical open questions surrounding gap junctions and their involvement in neural circuit development. Addressing them will greatly impact our understanding of the fundamental mechanisms of neocortical structure and function as well as the etiology of brain disease.
Collapse
Affiliation(s)
- Jun-Wei Cao
- School of Basic Medical Sciences, Xiangnan University, Chenzhou, Hunan 423000, China
| | - Lin-Yun Liu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai 200032, China
| | - Yong-Chun Yu
- Jing'an District Central Hospital of Shanghai, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai 200032, China.
| |
Collapse
|
5
|
Folorunso OO, Brown SE, Baruah J, Harvey TL, Jami SA, Radzishevsky I, Wolosker H, McNally JM, Gray JA, Vasudevan A, Balu DT. D-serine availability modulates prefrontal cortex inhibitory interneuron development and circuit maturation. Sci Rep 2023; 13:9595. [PMID: 37311798 PMCID: PMC10264435 DOI: 10.1038/s41598-023-35615-5] [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: 03/14/2023] [Accepted: 05/21/2023] [Indexed: 06/15/2023] Open
Abstract
The proper development and function of telencephalic GABAergic interneurons is critical for maintaining the excitation and inhibition (E/I) balance in cortical circuits. Glutamate contributes to cortical interneuron (CIN) development via N-methyl-D-aspartate receptors (NMDARs). NMDAR activation requires the binding of a co-agonist, either glycine or D-serine. D-serine (co-agonist at many mature forebrain synapses) is racemized by the neuronal enzyme serine racemase (SR) from L-serine. We utilized constitutive SR knockout (SR-/-) mice to investigate the effect of D-serine availability on the development of CINs and inhibitory synapses in the prelimbic cortex (PrL). We found that most immature Lhx6 + CINs expressed SR and the obligatory NMDAR subunit NR1. At embryonic day 15, SR-/- mice had an accumulation of GABA and increased mitotic proliferation in the ganglionic eminence and fewer Gad1 + (glutamic acid decarboxylase 67 kDa; GAD67) cells in the E18 neocortex. Lhx6 + cells develop into parvalbumin (PV+) and somatostatin (Sst+) CINs. In the PrL of postnatal day (PND) 16 SR-/- mice, there was a significant decrease in GAD67+ and PV+, but not SST + CIN density, which was associated with reduced inhibitory postsynaptic potentials in layer 2/3 pyramidal neurons. These results demonstrate that D-serine availability is essential for prenatal CIN development and postnatal cortical circuit maturation.
Collapse
Affiliation(s)
- Oluwarotimi O Folorunso
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA, 02478, USA.
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02215, USA.
| | - Stephanie E Brown
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA, 02478, USA
| | - Jugajyoti Baruah
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA, 02478, USA
- Angiogenesis and Brain Development Laboratory, Department of Neurosciences, Huntington Medical Research Institutes (HMRI), Pasadena, CA, 91105, USA
| | - Theresa L Harvey
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA, 02478, USA
| | - Shekib A Jami
- Center for Neuroscience, University of California Davis, Davis, CA, 95616, USA
| | - Inna Radzishevsky
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Herman Wolosker
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - James M McNally
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02215, USA
- VA Boston Healthcare System, West Roxbury, MA, 02132, USA
| | - John A Gray
- Center for Neuroscience, University of California Davis, Davis, CA, 95616, USA
| | - Anju Vasudevan
- Angiogenesis and Brain Development Laboratory, Department of Neurosciences, Huntington Medical Research Institutes (HMRI), Pasadena, CA, 91105, USA
| | - Darrick T Balu
- Division of Basic Neuroscience, McLean Hospital, Belmont, MA, 02478, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02215, USA
| |
Collapse
|
6
|
Dysregulated Signaling at Postsynaptic Density: A Systematic Review and Translational Appraisal for the Pathophysiology, Clinics, and Antipsychotics' Treatment of Schizophrenia. Cells 2023; 12:cells12040574. [PMID: 36831241 PMCID: PMC9954794 DOI: 10.3390/cells12040574] [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: 11/30/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Emerging evidence from genomics, post-mortem, and preclinical studies point to a potential dysregulation of molecular signaling at postsynaptic density (PSD) in schizophrenia pathophysiology. The PSD that identifies the archetypal asymmetric synapse is a structure of approximately 300 nm in diameter, localized behind the neuronal membrane in the glutamatergic synapse, and constituted by more than 1000 proteins, including receptors, adaptors, kinases, and scaffold proteins. Furthermore, using FASS (fluorescence-activated synaptosome sorting) techniques, glutamatergic synaptosomes were isolated at around 70 nm, where the receptors anchored to the PSD proteins can diffuse laterally along the PSD and were stabilized by scaffold proteins in nanodomains of 50-80 nm at a distance of 20-40 nm creating "nanocolumns" within the synaptic button. In this context, PSD was envisioned as a multimodal hub integrating multiple signaling-related intracellular functions. Dysfunctions of glutamate signaling have been postulated in schizophrenia, starting from the glutamate receptor's interaction with scaffolding proteins involved in the N-methyl-D-aspartate receptor (NMDAR). Despite the emerging role of PSD proteins in behavioral disorders, there is currently no systematic review that integrates preclinical and clinical findings addressing dysregulated PSD signaling and translational implications for antipsychotic treatment in the aberrant postsynaptic function context. Here we reviewed a critical appraisal of the role of dysregulated PSD proteins signaling in the pathophysiology of schizophrenia, discussing how antipsychotics may affect PSD structures and synaptic plasticity in brain regions relevant to psychosis.
Collapse
|
7
|
Wang J, Cai Y, Sun J, Feng H, Zhu X, Chen Q, Gao F, Ni Q, Mao L, Yang M, Sun B. Administration of intramuscular AAV-BDNF and intranasal AAV-TrkB promotes neurological recovery via enhancing corticospinal synaptic connections in stroke rats. Exp Neurol 2023; 359:114236. [PMID: 36183811 DOI: 10.1016/j.expneurol.2022.114236] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/14/2022] [Accepted: 09/25/2022] [Indexed: 12/30/2022]
Abstract
Stroke causes long-term disability in survivors. BDNF/TrkB plays an important role in synaptic plasticity and synaptic transmission in the central nervous system (CNS), promoting neurological recovery. In this study, we performed non-invasive treatment methods focused on intramuscular injection into stroke-injured forelimb muscles, or intranasal administration using adeno-associated virus (AAV) vectors carrying genes encoding BDNF or TrkB. In a permanent rat middle cerebral artery occlusion (MCAO) model, we assessed the effects of combination therapy with AAV-BDNF and AAV-TrkB on motor functional recovery and synaptic plasticity of the corticospinal connections. Our results showed that BDNF or TrkB gene transduced in the spinal anterior horn neurons and cerebral cortical neurons. Compared to AAV vector treatment alone, behavioral and electrophysiological results showed that the combination therapy significantly improved upper limb motor functional recovery and neurotransmission efficiency after stroke. BDA tracing, immunofluorescence staining, qRT-PCR, and transmission electron microscopy of synaptic ultrastructure results revealed that the combination therapy not only potently increased the expression of Synapsin I, PSD-95, and GAP-43, but also promoted the axonal remodeling and restoration of abnormal synaptic structures. These findings provide a new strategy for enhancing neural plasticity and a potential means to treat stroke clinically.
Collapse
Affiliation(s)
- Jing Wang
- Medical College of Qingdao University, Qingdao 266021, Shandong, China; Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Yichen Cai
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Jingyi Sun
- Department of Spinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Hua Feng
- Department of Otolaryngology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, Shandong, China
| | - Xiaoyu Zhu
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Qian Chen
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Feng Gao
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Qingbin Ni
- Postdoctoral Workstation, Taian Central Hospital, Taian 271000, Shandong, China
| | - Leilei Mao
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| | - Mingfeng Yang
- Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| | - Baoliang Sun
- Medical College of Qingdao University, Qingdao 266021, Shandong, China; Institute for Neurological Research, The Second Affiliated Hospital; School of Basic Medical Sciences of Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China.
| |
Collapse
|
8
|
Tapanes SA, Arizanovska D, Díaz MM, Folorunso OO, Harvey T, Brown SE, Radzishevsky I, Close LN, Jagid JR, Graciolli Cordeiro J, Wolosker H, Balu DT, Liebl DJ. Inhibition of glial D-serine release rescues synaptic damage after brain injury. Glia 2022; 70:1133-1152. [PMID: 35195906 PMCID: PMC9305835 DOI: 10.1002/glia.24161] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/13/2022] [Accepted: 02/04/2022] [Indexed: 11/28/2022]
Abstract
Synaptic damage is one of the most prevalent pathophysiological responses to traumatic CNS injury and underlies much of the associated cognitive dysfunction; however, it is poorly understood. The D-amino acid, D-serine, serves as the primary co-agonist at synaptic NMDA receptors (NDMARs) and is a critical mediator of NMDAR-dependent transmission and synaptic plasticity. In physiological conditions, D-serine is produced and released by neurons from the enzymatic conversion of L-serine by serine racemase (SRR). However, under inflammatory conditions, glial cells become a major source of D-serine. Here, we report that D-serine synthesized by reactive glia plays a critical role in synaptic damage after traumatic brain injury (TBI) and identify the therapeutic potential of inhibiting glial D-serine release though the transporter Slc1a4 (ASCT1). Furthermore, using cell-specific genetic strategies and pharmacology, we demonstrate that TBI-induced synaptic damage and memory impairment requires D-serine synthesis and release from both reactive astrocytes and microglia. Analysis of the murine cortex and acutely resected human TBI brain also show increased SRR and Slc1a4 levels. Together, these findings support a novel role for glial D-serine in acute pathological dysfunction following brain trauma, whereby these reactive cells provide the excess co-agonist levels necessary to initiate NMDAR-mediated synaptic damage.
Collapse
Affiliation(s)
- Stephen A. Tapanes
- The Miami Project to Cure Paralysis, Department of Neurological SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Dena Arizanovska
- The Miami Project to Cure Paralysis, Department of Neurological SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Madelen M. Díaz
- The Miami Project to Cure Paralysis, Department of Neurological SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Oluwarotimi O. Folorunso
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
- Translational Psychiatry LaboratoryMcLean HospitalBelmontMassachusettsUSA
| | - Theresa Harvey
- Translational Psychiatry LaboratoryMcLean HospitalBelmontMassachusettsUSA
| | - Stephanie E. Brown
- Translational Psychiatry LaboratoryMcLean HospitalBelmontMassachusettsUSA
| | - Inna Radzishevsky
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Liesl N. Close
- The Miami Project to Cure Paralysis, Department of Neurological SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jonathan R. Jagid
- The Miami Project to Cure Paralysis, Department of Neurological SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Joacir Graciolli Cordeiro
- The Miami Project to Cure Paralysis, Department of Neurological SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Herman Wolosker
- Department of Biochemistry, Rappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Darrick T. Balu
- Department of PsychiatryHarvard Medical SchoolBostonMassachusettsUSA
- Translational Psychiatry LaboratoryMcLean HospitalBelmontMassachusettsUSA
| | - Daniel J. Liebl
- The Miami Project to Cure Paralysis, Department of Neurological SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
9
|
Ni X, Mori H. Complex Processes Underlying the Dynamic Changes of D-serine Levels in AD Brains. Curr Alzheimer Res 2022; 19:485-493. [PMID: 35346007 DOI: 10.2174/1567205019666220328123048] [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: 09/22/2021] [Revised: 02/02/2022] [Accepted: 02/10/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder characterized by extracellular β-amyloid (Aβ) plaques and cognitive impairments. D-Serine, produced by the enzyme serine racemase (SR) in the brain, functions as an endogenous co-agonist at the glycine-binding site of N-methyl-D-aspartate receptor (NMDAR), has been implicated in the pathophysiological progression of AD. OBJECTIVES Evidence regarding the understanding of the role and dynamic modulation of D-serine during AD progression remains controversial. This literature review aims to offer novel research directions for studying the functions and metabolisms of D-serine in AD brains. METHODS We searched PubMed, using D-serine/SR and AD as keywords. Studies related to NMDAR dysfunction, neuronal excitotoxicity, D-serine dynamic changes and inflammatory response were included. RESULTS This review primarily discusses: (i) Aβ oligomers' role in NMDAR dysregulation, and the subsequent synaptic dysfunction and neuronal damage in AD, (ii) D-serine's role in NMDAR-elicited excitotoxicity, and (iii) the involvement of D-serine and SR in AD-related inflammatory pathological progression. CONCLUSION We also presented supposed metabolism and dynamic changes of D-serine during AD progression and hypothesized that: (i) the possible modulation of D-serine levels or SR expression as an effective method of alleviating neurotoxicity during AD pathophysiological progression, and (ii) the dynamic changes of D-serine levels in AD brains possibly resulting from complex processes.
Collapse
Affiliation(s)
- Xiance Ni
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-0194, Japan
| | - Hisashi Mori
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-0194, Japan.,Research Center for Idling Brain Science (RCIBS), University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
10
|
Park DK, Petshow S, Anisimova M, Barragan EV, Gray JA, Stein IS, Zito K. Reduced d-serine levels drive enhanced non-ionotropic NMDA receptor signaling and destabilization of dendritic spines in a mouse model for studying schizophrenia. Neurobiol Dis 2022; 170:105772. [PMID: 35605760 PMCID: PMC9352378 DOI: 10.1016/j.nbd.2022.105772] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 10/31/2022] Open
Abstract
Schizophrenia is a psychiatric disorder that affects over 20 million people globally. Notably, schizophrenia is associated with decreased density of dendritic spines and decreased levels of d-serine, a co-agonist required for opening of the N-methyl-d-aspartate receptor (NMDAR). We hypothesized that lowered d-serine levels associated with schizophrenia would enhance ion flux-independent signaling by the NMDAR, driving destabilization and loss of dendritic spines. We tested our hypothesis using the serine racemase knockout (SRKO) mouse model, which lacks the enzyme for d-serine production. We show that activity-dependent spine growth is impaired in SRKO mice, but can be acutely rescued by exogenous d-serine. Moreover, we find a significant bias of synaptic plasticity toward spine shrinkage in the SRKO mice as compared to wild-type littermates. Notably, we demonstrate that enhanced ion flux-independent signaling through the NMDAR contributes to this bias toward spine destabilization, which is exacerbated by an increase in synaptic NMDARs in hippocampal synapses of SRKO mice. Our results support a model in which lowered d-serine levels associated with schizophrenia enhance ion flux-independent NMDAR signaling and bias toward spine shrinkage and destabilization.
Collapse
|
11
|
Human Serine Racemase Weakly Binds the Third PDZ Domain of PSD-95. Int J Mol Sci 2022; 23:ijms23094959. [PMID: 35563349 PMCID: PMC9105370 DOI: 10.3390/ijms23094959] [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: 03/15/2022] [Revised: 04/13/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022] Open
Abstract
Human serine racemase (hSR) is a pyridoxal-5'-phosphate (PLP)-dependent dimer that catalyzes the formation of D-serine from L-serine, as well as the dehydration of both L- and D-serine to pyruvate and ammonia. As D-serine is a co-agonist of N-methyl-D-aspartate receptors (NMDARs), hSR is a key enzyme in glutamatergic neurotransmission. hSR activity is finely regulated by Mg2+, ATP, post-translational modifications, and the interaction with protein partners. In particular, the C-terminus of murine SR binds the third PDZ domain (PDZ3) of postsynaptic density protein 95 (PSD-95), a member of the membrane-associated guanylate kinase (MAGUK) family involved in the trafficking and localization of glutamate receptors. The structural details of the interaction and the stability of the complex have not been elucidated yet. We evaluated the binding of recombinant human PSD-95 PDZ3 to hSR by glutaraldehyde cross-linking, pull-down assays, isothermal titration calorimetry, nuclear magnetic resonance, and enzymatic assays. Overall, a weak interaction was observed, confirming the binding for the human orthologs but supporting the hypothesis that a third protein partner (i.e., stargazin) is required for the regulation of hSR activity by PSD-95 and to stabilize their interaction.
Collapse
|
12
|
Takagi S, Puhl MD, Anderson T, Balu DT, Coyle JT. Serine Racemase Expression by Striatal Neurons. Cell Mol Neurobiol 2022; 42:279-289. [PMID: 32445040 PMCID: PMC7680280 DOI: 10.1007/s10571-020-00880-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 05/15/2020] [Indexed: 01/03/2023]
Abstract
D-serine is synthesized by serine racemase (SR) and is a co-agonist at forebrain N-methyl-D-aspartate receptors (NMDARs). D-serine and SR are expressed primarily in neurons, but not in quiescent astrocytes. In this study, we examined the localization of D-serine and SR in the mouse striatum and the effects of genetically silencing SR expression in GABAergic interneurons (iSR-/-). iSR-/- mice had substantially reduced SR expression almost exclusively in striatum, but only exhibited marginal D-serine reduction. SR positive cells in the striatum showed strong co-localization with dopamine- and cyclic AMP-regulated neuronal phosphoprotein (DARPP32) in wild type mice. Transgenic fluorescent reporter mice for either the D1 or D2 dopamine receptors exhibited a 65:35 ratio for co-localization with D1and D2 receptor positive cells, respectively. These results indicate that GABAergic medium spiny neurons receiving dopaminergic inputs in striatum robustly and uniformly express SR. In behavioral tests, iSR-/- mice showed a blunted response to the hedonic and stimulant effects of cocaine, without affecting anxiety-related behaviors. Because the cocaine effects have been shown in the constitutive SR-/- mice, the restriction of the blunted response to cocaine to iSR-/- mice reinforces the conclusion that D-serine in striatal GABAergic neurons plays an important role in mediating dopaminergic stimulant effects. Results in this study suggest that SR in striatal GABAergic neurons is synthesizing D-serine, not as a glutamatergic co-transmitter, but rather as an autocrine whereby the GABAergic neurons control the excitability of their NMDARs by determining the availability of the co-agonist, D-serine.
Collapse
Affiliation(s)
- Shunsuke Takagi
- Department of Psychiatry and Behavioral Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan,Department of Psychiatry, Tokyo Medical and Dental University Hospital, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan,Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan,Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA
| | - Matthew D. Puhl
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA
| | - Thea Anderson
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA
| | - Darrick T. Balu
- Department of Psychiatry, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA,Translational Psychiatry Laboratory, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA
| | - Joseph T. Coyle
- Laboratory for Psychiatric and Molecular Neuroscience, McLean Hospital, 115 Mill Street, Belmont, MA 02478, USA,Department of Psychiatry, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| |
Collapse
|
13
|
Jami SA, Cameron S, Wong JM, Daly ER, McAllister AK, Gray JA. Increased excitation-inhibition balance and loss of GABAergic synapses in the serine racemase knockout model of NMDA receptor hypofunction. J Neurophysiol 2021; 126:11-27. [PMID: 34038186 DOI: 10.1152/jn.00661.2020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
There is substantial evidence that both N-methyl-D-aspartate receptor (NMDAR) hypofunction and dysfunction of GABAergic neurotransmission contribute to schizophrenia, though the relationship between these pathophysiological processes remains largely unknown. Although models using cell-type-specific genetic deletion of NMDARs have been informative, they display overly pronounced phenotypes extending beyond those of schizophrenia. Here, we used the serine racemase knockout (SRKO) mice, a model of reduced NMDAR activity rather than complete receptor elimination, to examine the link between NMDAR hypofunction and decreased GABAergic inhibition. The SRKO mice, in which there is a >90% reduction in the NMDAR coagonist d-serine, exhibit many of the neurochemical and behavioral abnormalities observed in schizophrenia. We found a significant reduction in inhibitory synapses onto CA1 pyramidal neurons in the SRKO mice. This reduction increases the excitation/inhibition balance resulting in enhanced synaptically driven neuronal excitability without changes in intrinsic excitability. Consistently, significant reductions in inhibitory synapse density in CA1 were observed by immunohistochemistry. We further show, using a single-neuron genetic deletion approach, that the loss of GABAergic synapses onto pyramidal neurons observed in the SRKO mice is driven in a cell-autonomous manner following the deletion of SR in individual CA1 pyramidal cells. These results support a model whereby NMDAR hypofunction in pyramidal cells disrupts GABAergic synapses leading to disrupted feedback inhibition and impaired neuronal synchrony.NEW & NOTEWORTHY Recently, disruption of excitation/inhibition (E/I) balance has become an area of considerable interest for psychiatric research. Here, we report a reduction in inhibition in the serine racemase knockout mouse model of schizophrenia that increases E/I balance and enhances synaptically driven neuronal excitability. This reduced inhibition was driven cell-autonomously in pyramidal cells lacking serine racemase, suggesting a novel mechanism for how chronic NMDA receptor hypofunction can disrupt information processing in schizophrenia.
Collapse
Affiliation(s)
- Shekib A Jami
- Center for Neuroscience, University of California, Davis, California
| | - Scott Cameron
- Center for Neuroscience, University of California, Davis, California
| | - Jonathan M Wong
- Center for Neuroscience, University of California, Davis, California
| | - Emily R Daly
- Center for Neuroscience, University of California, Davis, California
| | - A Kimberley McAllister
- Center for Neuroscience, University of California, Davis, California.,Department of Neurobiology, Physiology, and Behavior, University of California, Davis, California.,Department of Neurology, University of California, Davis, California
| | - John A Gray
- Center for Neuroscience, University of California, Davis, California.,Department of Neurology, University of California, Davis, California
| |
Collapse
|
14
|
Folorunso OO, Harvey TL, Brown SE, Cruz C, Shahbo E, Ajjawi I, Balu DT. Forebrain expression of serine racemase during postnatal development. Neurochem Int 2021; 145:104990. [PMID: 33592203 PMCID: PMC8012237 DOI: 10.1016/j.neuint.2021.104990] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 12/23/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are important for synaptogenesis, synaptic maturation and refinement during the early postnatal weeks after birth. Defective synapse formation or refinement underlie cognitive and emotional abnormalities in various neurodevelopmental disorders (NDDs), including schizophrenia (Sz) and autism spectrum disorder (ASD). Serine racemase (SR) is a neuronal enzyme that produces D-serine, a co-agonist required for full NMDAR activation. NMDAR hypofunction as a result of genetic SR elimination and reduced synaptic availability of D-serine reduces neuronal dendritic arborization and spine density. In adult mouse brain, the expression of SR parallels that of NMDARs across forebrain regions including the striatum, amygdala, hippocampus, and medial prefrontal cortex (mPFC). However, there have yet to be studies providing a detailed characterization of the spatial and temporal expression of SR during early periods of synaptogenesis. Here, we examined the postnatal expression of SR in cortical and subcortical brain regions important for learning, memory and emotional regulation, during the first four weeks after birth. Using dual-antigen immunofluorescence, we demonstrate that the number of SR+ neurons steadily increases with postnatal age across the mPFC, amygdala, hippocampus and striatum. We also identified differences in the rate of SR protein induction both across and within brain regions. Analyzing existing human post-mortem brain in situ data, there was a similar developmental mRNA expression profile of SRR and GRIN1 (GluN1 subunit) from infancy through the first decade of life. Our findings further support a developmental role for D-serine mediated NMDAR activation regulating synaptogenesis and neural circuit refinement, which has important implications for the pathophysiology of Sz and other NDDs.
Collapse
Affiliation(s)
- Oluwarotimi O Folorunso
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, United States; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States
| | - Theresa L Harvey
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States
| | - Stephanie E Brown
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States
| | - Cristina Cruz
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States; Harvard University, Cambridge, MA, 02138, United States
| | - Ellie Shahbo
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States; Harvard University, Cambridge, MA, 02138, United States
| | - Ismail Ajjawi
- Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States; Harvard University, Cambridge, MA, 02138, United States
| | - Darrick T Balu
- Department of Psychiatry, Harvard Medical School, Boston, MA, 02115, United States; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, 02478, United States.
| |
Collapse
|
15
|
Lo AC, Rajan N, Gastaldo D, Telley L, Hilal ML, Buzzi A, Simonato M, Achsel T, Bagni C. Absence of RNA-binding protein FXR2P prevents prolonged phase of kainate-induced seizures. EMBO Rep 2021; 22:e51404. [PMID: 33779029 PMCID: PMC8024897 DOI: 10.15252/embr.202051404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 01/26/2021] [Accepted: 01/29/2021] [Indexed: 12/26/2022] Open
Abstract
Status epilepticus (SE) is a condition in which seizures are not self-terminating and thereby pose a serious threat to the patient's life. The molecular mechanisms underlying SE are likely heterogeneous and not well understood. Here, we reveal a role for the RNA-binding protein Fragile X-Related Protein 2 (FXR2P) in SE. Fxr2 KO mice display reduced sensitivity specifically to kainic acid-induced SE. Immunoprecipitation of FXR2P coupled to next-generation sequencing of associated mRNAs shows that FXR2P targets are enriched in genes that encode glutamatergic post-synaptic components. Of note, the FXR2P target transcriptome has a significant overlap with epilepsy and SE risk genes. In addition, Fxr2 KO mice fail to show sustained ERK1/2 phosphorylation induced by KA and present reduced burst activity in the hippocampus. Taken together, our findings show that the absence of FXR2P decreases the expression of glutamatergic proteins, and this decrease might prevent self-sustained seizures.
Collapse
Affiliation(s)
- Adrian C Lo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Nicholas Rajan
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Denise Gastaldo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Ludovic Telley
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Muna L Hilal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Andrea Buzzi
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Michele Simonato
- Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy.,Division of Neuroscience, IRCCS San Raffaele Hospital, Milan, Italy
| | - Tilmann Achsel
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Claudia Bagni
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium.,Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
16
|
Dyakin VV, Wisniewski TM, Lajtha A. Racemization in Post-Translational Modifications Relevance to Protein Aging, Aggregation and Neurodegeneration: Tip of the Iceberg. Symmetry (Basel) 2021; 13:455. [PMID: 34350031 PMCID: PMC8330555 DOI: 10.3390/sym13030455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Homochirality of DNA and prevalent chirality of free and protein-bound amino acids in a living organism represents the challenge for modern biochemistry and neuroscience. The idea of an association between age-related disease, neurodegeneration, and racemization originated from the studies of fossils and cataract disease. Under the pressure of new results, this concept has a broader significance linking protein folding, aggregation, and disfunction to an organism's cognitive and behavioral functions. The integrity of cognitive function is provided by a delicate balance between the evolutionarily imposed molecular homo-chirality and the epigenetic/developmental impact of spontaneous and enzymatic racemization. The chirality of amino acids is the crucial player in the modulation the structure and function of proteins, lipids, and DNA. The collapse of homochirality by racemization is the result of the conformational phase transition. The racemization of protein-bound amino acids (spontaneous and enzymatic) occurs through thermal activation over the energy barrier or by the tunnel transfer effect under the energy barrier. The phase transition is achieved through the intermediate state, where the chirality of alpha carbon vanished. From a thermodynamic consideration, the system in the homo-chiral (single enantiomeric) state is characterized by a decreased level of entropy. The oscillating protein chirality is suggesting its distinct significance in the neurotransmission and flow of perceptual information, adaptive associative learning, and cognitive laterality. The common pathological hallmarks of neurodegenerative disorders include protein misfolding, aging, and the deposition of protease-resistant protein aggregates. Each of the landmarks is influenced by racemization. The brain region, cell type, and age-dependent racemization critically influence the functions of many intracellular, membrane-bound, and extracellular proteins including amyloid precursor protein (APP), TAU, PrP, Huntingtin, α-synuclein, myelin basic protein (MBP), and collagen. The amyloid cascade hypothesis in Alzheimer's disease (AD) coexists with the failure of amyloid beta (Aβ) targeting drug therapy. According to our view, racemization should be considered as a critical factor of protein conformation with the potential for inducing order, disorder, misfolding, aggregation, toxicity, and malfunctions.
Collapse
Affiliation(s)
- Victor V. Dyakin
- Virtual Reality Perception Lab (VRPL), The Nathan S. Kline Institute for Psychiatric Research (NKI), Orangeburg, NY 10962, USA
| | - Thomas M. Wisniewski
- Departments of Neurology, Pathology and Psychiatry, Center for Cognitive Neurology, New York University School of Medicine, New York, NY 10016, USA
| | - Abel Lajtha
- Center for Neurochemistry, The Nathan S. Kline Institute for Psychiatric Research (NKI), Orangeburg, NY 10962, USA
| |
Collapse
|
17
|
Yamagata K. Astrocyte-induced synapse formation and ischemic stroke. J Neurosci Res 2021; 99:1401-1413. [PMID: 33604930 DOI: 10.1002/jnr.24807] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/26/2021] [Indexed: 12/16/2022]
Abstract
Astrocytes are closely associated with the regulation of synapse formation and function. In addition, astrocytes have been shown to block certain brain impairments, including synaptic damage from stroke and other diseases of the central nervous system (CNS). Although astrocytes do not completely prevent synaptic damage, they appear to be protective and to restore synaptic function following damage. The purpose of this study is to discuss the role of astrocytes in synaptogenesis and synaptic damage in ischemic stroke. I detail the mechanism of action of the multiple factors secreted by astrocytes that are involved in synapse formation. In particular, I describe the characteristics and role in synapse formation of each secreted molecule related to synaptic structure and function. Furthermore, I discuss the effect of astrocytes on synaptogenesis and repair in ischemic stroke and in other CNS diseases. Astrocytes release molecules such as thrombospondin, hevin, secreted protein acidic rich in cysteine, etc., due to activation by ischemia to induce synaptic structure and function, an effect associated with protection of the brain from synaptic damage in ischemic stroke. In conclusion, I show that astrocytes may regulate synaptic transmission while having the potential to block and repair synaptic dysfunction in stroke-associated brain damage.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Department of Food Bioscience & Biotechnology, College of Bioresource Science, Nihon University (UNBS), Fujisawa, Japan
| |
Collapse
|
18
|
Postsynaptic Serine Racemase Regulates NMDA Receptor Function. J Neurosci 2020; 40:9564-9575. [PMID: 33158959 DOI: 10.1523/jneurosci.1525-20.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/23/2020] [Accepted: 11/01/2020] [Indexed: 01/03/2023] Open
Abstract
d-serine is the primary NMDAR coagonist at mature forebrain synapses and is synthesized by the enzyme serine racemase (SR). However, our understanding of the mechanisms regulating the availability of synaptic d-serine remains limited. Though early studies suggested d-serine is synthesized and released from astrocytes, more recent studies have demonstrated a predominantly neuronal localization of SR. More specifically, recent work intriguingly suggests that SR may be found at the postsynaptic density, yet the functional implications of postsynaptic SR on synaptic transmission are not yet known. Here, we show an age-dependent dendritic and postsynaptic localization of SR and d-serine by immunohistochemistry and electron microscopy in mouse CA1 pyramidal neurons. In addition, using a single-neuron genetic approach in SR conditional KO mice from both sexes, we demonstrate a cell-autonomous role for SR in regulating synaptic NMDAR function at Schaffer collateral (CA3)-CA1 synapses. Importantly, single-neuron genetic deletion of SR resulted in the elimination of LTP at 1 month of age, which could be rescued by exogenous d-serine. Interestingly, there was a restoration of LTP by 2 months of age that was associated with an upregulation of synaptic GluN2B. Our findings support a cell-autonomous role for postsynaptic neuronal SR in regulating synaptic NMDAR function and suggests a possible autocrine mode of d-serine action.SIGNIFICANCE STATEMENT NMDARs are key regulators of neurodevelopment and synaptic plasticity and are unique in their requirement for binding of a coagonist, which is d-serine at most forebrain synapses. However, our understanding of the mechanisms regulating synaptic d-serine availability remains limited. d-serine is synthesized in the brain by the neuronal enzyme serine racemase (SR). Here, we show dendritic and postsynaptic localization of SR and d-serine in CA1 pyramidal neurons. In addition, using single-neuron genetic deletion of SR, we establish a role of postsynaptic SR in regulating NMDAR function. These results support an autocrine mode of d-serine action at synapses.
Collapse
|
19
|
Maugard M, Vigneron PA, Bolaños JP, Bonvento G. l-Serine links metabolism with neurotransmission. Prog Neurobiol 2020; 197:101896. [PMID: 32798642 DOI: 10.1016/j.pneurobio.2020.101896] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/20/2020] [Accepted: 08/09/2020] [Indexed: 12/12/2022]
Abstract
Brain energy metabolism is often considered as a succession of biochemical steps that metabolize the fuel (glucose and oxygen) for the unique purpose of providing sufficient ATP to maintain the huge information processing power of the brain. However, a significant fraction (10-15 %) of glucose is shunted away from the ATP-producing pathway (oxidative phosphorylation) and may be used to support other functions. Recent studies have pointed to the marked compartmentation of energy metabolic pathways between neurons and glial cells. Here, we focused our attention on the biosynthesis of l-serine, a non-essential amino acid that is formed exclusively in glial cells (mostly astrocytes) by re-routing the metabolic fate of the glycolytic intermediate, 3-phosphoglycerate (3PG). This metabolic pathway is called the phosphorylated pathway and transforms 3PG into l-serine via three enzymatic reactions. We first compiled the available data on the mechanisms that regulate the flux through this metabolic pathway. We then reviewed the current evidence that is beginning to unravel the roles of l-serine both in the healthy and diseased brain, leading to the notion that this specific metabolic pathway connects glial metabolism with synaptic activity and plasticity. We finally suggest that restoring astrocyte-mediated l-serine homeostasis may provide new therapeutic strategies for brain disorders.
Collapse
Affiliation(s)
- Marianne Maugard
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Pierre-Antoine Vigneron
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Institute of Biomedical Research of Salamanca, 37007, Salamanca, Spain
| | - Gilles Bonvento
- Université Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-aux-Roses, France.
| |
Collapse
|
20
|
Xiong B, Zhang W, Zhang L, Huang X, Zhou W, Zou Q, Manyande A, Wang J, Tian Y, Tian X. Hippocampal glutamatergic synapses impairment mediated novel-object recognition dysfunction in rats with neuropathic pain. Pain 2020; 161:1824-1836. [PMID: 32701842 DOI: 10.1097/j.pain.0000000000001878] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cognitive impairment is one of the most common complications associated with chronic pain. Almost 20% of chronic pain patients suffer from cognitive impairment, which may substantially influence their quality of life. Levels of major excitatory neurotransmitters in the central nervous system and alterations in the glutamatergic system may influence cognitive function and the pain sensory pathway. In this study, we adopted the spared nerve injury model to establish the progress of chronic pain and investigated the mechanism underlying the cognitive aspect related to it. At behavioral level, using the novel-object recognition test, mechanical hypersensitivity was observed in peripheral nerve-injured rats because they exhibited recognition deficits. We showed a dramatic decrease in hippocampal glutamate concentration using nuclear magnetic resonance and reduced glutamatergic synaptic transmission using whole-cell recordings. These were associated with deficient hippocampal long-term potentiation induced by high-frequency stimulation of the Schaffer collateral afferent. Ultra-high-performance liquid chromatography revealed lower levels of D-serine in the hippocampus of the spared nerve injury rats and that D-serine treatment could restore synaptic plasticity and cognitive dysfunction. The reduction of excitatory synapses was also increased by administering D-serine. These findings suggest that chronic pain has a critical effect on synaptic plasticity linked to cognitive function and may built up a new target for the development of cognitive impairment under chronic pain conditions.
Collapse
Affiliation(s)
- Bingrui Xiong
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Anesthesiology, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Wen Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Longqing Zhang
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xian Huang
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenchang Zhou
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qian Zou
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Anne Manyande
- School of Human and Social Sciences, University of West London, London, United Kingdom
| | - Jie Wang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yuke Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuebi Tian
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
21
|
D-Serine Signaling and NMDAR-Mediated Synaptic Plasticity Are Regulated by System A-Type of Glutamine/D-Serine Dual Transporters. J Neurosci 2020; 40:6489-6502. [PMID: 32661027 DOI: 10.1523/jneurosci.0801-20.2020] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/22/2020] [Accepted: 06/25/2020] [Indexed: 02/03/2023] Open
Abstract
D-serine is a physiologic coagonist of NMDA receptors (NMDARs) required for synaptic plasticity, but mechanisms that terminate D-serine signaling are unclear. In particular, the identity of unidirectional plasma membrane transporters that mediate D-serine reuptake has remained elusive. We report that D-serine and glutamine share the same neuronal transport system, consisting of the classic system A transporters Slc38a1 and Slc38a2. We show that these transporters are not saturated with glutamine in vivo and regulate the extracellular levels of D-serine and NMDAR activity. Glutamine increased the NMDAR-dependent long-term potentiation and the isolated NMDAR potentials at the Schaffer collateral-CA1 synapses, but without affecting basal neurotransmission in male mice. Glutamine did not increase the NMDAR potentials in slices from serine racemase knock-out mice, which are devoid of D-serine, indicating that the effect of glutamine is caused by outcompeting D-serine for a dual glutamine-D-serine transport system. Inhibition of the system A reduced the uptake of D-serine in synaptosomes and neuronal cultures of mice of either sex, while increasing the extracellular D-serine concentration in slices and in vivo by microdialysis. When compared with Slc38a2, the Slc38a1 transporter displayed more favorable kinetics toward the D-enantiomer. Biochemical experiments with synaptosomes from Slc38a1 knock-down mice of either sex further support its role as a D-serine reuptake system. Our study identifies the first concentrative and electrogenic transporters mediating D-serine reuptake in vivo In addition to their classical role in the glutamine-glutamate cycle, system A transporters regulate the synaptic turnover of D-serine and its effects on NMDAR synaptic plasticity.SIGNIFICANCE STATEMENT Despite the plethora of roles attributed to D-serine, the regulation of its synaptic turnover is poorly understood. We identified the system A transporters Slc38a1 and Slc38a2 as the main pathway for neuronal reuptake of D-serine. These transporters are not saturated with glutamine in vivo and provide an unexpected link between the serine shuttle pathway, responsible for regulating D-serine synaptic turnover, and the glutamine-glutamate cycle. Our observations suggest that Slc38a1 and Slc38a2 have a dual role in regulating neurotransmission. In addition to their classical role as the glutamine providers, the system A transporters regulate extracellular D-serine and therefore affect NMDAR-dependent synaptic plasticity. Higher glutamine export from astrocytes would increase extracellular D-serine, providing a feedforward mechanism to increase synaptic NMDAR activation.
Collapse
|
22
|
Niu HM, Ma DL, Wang MY, Chen XP, Zhang L, Li YL, Zhang L, Li L. Epimedium flavonoids protect neurons and synapses in the brain via activating NRG1/ErbB4 and BDNF/Fyn signaling pathways in a chronic cerebral hypoperfusion rat model. Brain Res Bull 2020; 162:132-140. [PMID: 32592805 DOI: 10.1016/j.brainresbull.2020.06.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/02/2020] [Accepted: 06/19/2020] [Indexed: 12/20/2022]
Abstract
Cerebral hypoperfusion is a common feature of cerebral small vascular disease (CSVD), which has been considered as one of the causes of cognitive decline in recent years. Epimedium flavonoids (EF) are the main ingredients extracted from Epimedium. The purpose of this study was to investigate the effects of EF on cognitive impairment, and the underlying mechanisms in rats with permanent occlusion of the bilateral common carotid artery (2VO). EF (50, 100, and 200 mg/kg) was intragastrically administered for 12 weeks starting 2 weeks after 2VO surgery. The results showed that EF treatment improved learning and memory impairment in 2VO rats evaluated by novel object recognition and Y-maze tests. NeuN immunohistochemical staining indicated that EF alleviated neuronal loss in the hippocampus and cerebral cortex of 2VO rats. MAP-2 immunofluorescence staining and western blotting showed that EF protected neuronal dendrites and increased the expression of cytoskeleton proteins MAP-2 and NF200 in the hippocampus of 2VO rats. Moreover, EF protected the synapse ultrastructure detected by transmission electron microscopy, and increased the expression of synaptic plasticity-related proteins, including synaptophysin, synaptotagmin-I, synapsin I, PSD-95, p-NMDA2B, and p-CaMKII-α in the hippocampus of 2VO rats. In addition, EF increased the expression of neuregulin-1 (NRG-1), p-ErbB4, brain-derived neurotrophic factor (BDNF), p-Fyn, PI3K, p-Akt, and p-CREB in the hippocampus of 2VO rats. These results suggest that EF may protect neurons and synapses by activating the NRG1/ErbB4, BDNF/Fyn, and P13 K/Akt/CREB pathways in the hippocampus and cerebral cortex, thus improving cognitive impairment induced by chronic cerebral hypoperfusion. EF may be a potential candidate drug for chronic cerebral hypoperfusion and CSVD therapy.
Collapse
Affiliation(s)
- Hong-Mei Niu
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Deng-Lei Ma
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Ming-Yang Wang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Xiao-Ping Chen
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Li Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Ya-Li Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China
| | - Lin Li
- Department of Pharmacy, Xuanwu Hospital of Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing Engineering Research Center for Nerve System Drugs, Beijing Institute for Brain Disorders, Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, 100053, China.
| |
Collapse
|
23
|
Wolosker H, Balu DT. D-Serine as the gatekeeper of NMDA receptor activity: implications for the pharmacologic management of anxiety disorders. Transl Psychiatry 2020; 10:184. [PMID: 32518273 PMCID: PMC7283225 DOI: 10.1038/s41398-020-00870-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/04/2020] [Accepted: 05/14/2020] [Indexed: 12/12/2022] Open
Abstract
Fear, anxiety, and trauma-related disorders, including post-traumatic stress disorder (PTSD), are quite common and debilitating, with an estimated lifetime prevalence of ~28% in Western populations. They are associated with excessive fear reactions, often including an inability to extinguish learned fear, increased avoidance behavior, as well as altered cognition and mood. There is an extensive literature demonstrating the importance of N-methyl-D-aspartate receptor (NMDAR) function in regulating these behaviors. NMDARs require the binding of a co-agonist, D-serine or glycine, at the glycine modulatory site (GMS) to function. D-serine is now garnering attention as the primary NMDAR co-agonist in limbic brain regions implicated in neuropsychiatric disorders. L-serine is synthesized by astrocytes, which is then transported to neurons for conversion to D-serine by serine racemase (SR), a model we term the 'serine shuttle.' The neuronally-released D-serine is what regulates NMDAR activity. Our review discusses how the systems that regulate the synaptic availability of D-serine, a critical gatekeeper of NMDAR-dependent activation, could be targeted to improve the pharmacologic management of anxiety-related disorders where the desired outcomes are the facilitation of fear extinction, as well as mood and cognitive enhancement.
Collapse
Affiliation(s)
- Herman Wolosker
- grid.6451.60000000121102151Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, 31096 Israel
| | - Darrick T. Balu
- grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA 02115 USA ,grid.240206.20000 0000 8795 072XTranslational Psychiatry Laboratory, McLean Hospital, Belmont, MA 02478 USA
| |
Collapse
|
24
|
D-Serine, the Shape-Shifting NMDA Receptor Co-agonist. Neurochem Res 2020; 45:1344-1353. [PMID: 32189130 DOI: 10.1007/s11064-020-03014-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/04/2020] [Accepted: 03/11/2020] [Indexed: 12/17/2022]
Abstract
Shape-shifting, a phenomenon wide-spread in folklore, refers to the ability to physically change from one identity to another, typically from an innocuous entity to a destructive one. The amino acid D-serine over the last 25 years has "shape-shifted" into several identities: a purported glial transmitter activating N-methyl-D-aspartate receptors (NMDARs), a co-transmitter concentrated in excitatory glutamatergic neurons, an autocrine that is released at dendritic spines to prime their post-synaptic NMDARs for an instantaneous response to glutamate and an excitotoxic moiety released from inflammatory (A1) astrocytes. This article will review evidence in support of these scenarios and the artifacts that misled investigators of the true identity of D-serine.
Collapse
|
25
|
Berdenis van Berlekom A, Muflihah CH, Snijders GJLJ, MacGillavry HD, Middeldorp J, Hol EM, Kahn RS, de Witte LD. Synapse Pathology in Schizophrenia: A Meta-analysis of Postsynaptic Elements in Postmortem Brain Studies. Schizophr Bull 2020; 46:374-386. [PMID: 31192350 PMCID: PMC7442385 DOI: 10.1093/schbul/sbz060] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Changed synapse density has been suggested to be involved in the altered brain connectivity underlying schizophrenia (SCZ) pathology. However, postmortem studies addressing this topic are heterogeneous and it is not known whether changes are restricted to specific brain regions. Using meta-analysis, we systematically and quantitatively reviewed literature on the density of postsynaptic elements in postmortem brain tissue of patients with SCZ compared to healthy controls. We included 3 outcome measurements for postsynaptic elements: dendritic spine density (DSD), postsynaptic density (PSD) number, and PSD protein expression levels. Random-effects meta-analysis (31 studies) revealed an overall decrease in density of postsynaptic elements in SCZ (Hedges's g: -0.33; 95% CI: -0.60 to -0.05; P = .020). Subgroup analyses showed reduction of postsynaptic elements in cortical but not subcortical tissues (Hedges's g: -0.44; 95% CI: -0.76 to -0.12; P = .008, Hedges's g: -0.11; 95% CI: -0.54 to 0.35; P = .671) and specifically a decrease for the outcome measure DSD (Hedges's g: -0.81; 95% CI: -1.37 to -0.26; P = .004). Further exploratory analyses showed a significant decrease of postsynaptic elements in the prefrontal cortex and cortical layer 3. In all analyses, substantial heterogeneity was present. Meta-regression analyses showed no influence of age, sex, postmortem interval, or brain bank on the effect size. This meta-analysis shows a region-specific decrease in the density of postsynaptic elements in SCZ. This phenotype provides an important cellular hallmark for future preclinical and neuropathological research in order to increase our understanding of brain dysconnectivity in SCZ.
Collapse
Affiliation(s)
- Amber Berdenis van Berlekom
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands,To whom correspondence should be addressed; tel: +31-88-75-68811, fax: +31(0)887569032, e-mail:
| | - Cita H Muflihah
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands,Faculty of Pharmacy, Universitas Muhammadiyah Surakarta, Sukoharjo, Indonesia
| | - Gijsje J L J Snijders
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands,Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Harold D MacGillavry
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - René S Kahn
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY,Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY
| | - Lot D de Witte
- Department of Psychiatry, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY,Mental Illness Research, Education and Clinical Center (MIRECC), James J Peters VA Medical Center, Bronx, NY
| |
Collapse
|
26
|
Sevoflurane Impairs Short-Term Memory by Affecting PSD-95 and AMPA Receptor in the Hippocampus of a Mouse Model. Behav Neurol 2019; 2019:1068260. [PMID: 31772680 PMCID: PMC6854262 DOI: 10.1155/2019/1068260] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 07/23/2019] [Accepted: 09/10/2019] [Indexed: 11/18/2022] Open
Abstract
Objective To explore the effects of sevoflurane on the latency and error times of the passive avoidance and levels of PSD-95 and AMPA receptors in the hippocampus. We evaluated the effects of sevoflurane on short-term memory in adult mice and explored the possible mechanism. Methods 144 Kunming mice (2-3 months, 30-35 g) were randomly divided into two groups A (n = 64) and B (n = 80) and received the dark-avoidance (DA) and step-down avoidance (SA) tests, respectively. The groups DA and SA were further divided into control (inhaled 40% O2 2 h) and sevoflurane (3.3% sevoflurane and 40% O2 2 h) subgroups. Before inhalation intervention, all mice were trained to be familiar with the Morris water maze (MWM). According to the test points of behavioral indicators, 8 mice were randomly selected from each subgroup at point 12 h (T1), 24 h (T2), 48 h (T3), and 72 h (T4) after inhalation intervention. The step-through latency and error times were measured in 5 min. After the behavioral test, the mice were killed and the tissues of the hippocampus were taken for hematoxylin and eosin (H&E) staining. The expression level of PSD-95 and AMPA receptors in the hippocampus was detected by immunohistochemistry and Western Blot. The changes of synaptic transmission were measured via electrophysiology analysis of hippocampal slices. Results The mice in the control subgroups found the platform in a shorter pathway than those in the sevoflurane subgroups during an MWM test. The step-through latency of T1 and T2 in the sevoflurane subgroup was shorter than baseline time, and the error times were increased in 5 min and higher than baseline time when compared with the control subgroup (P < 0.05) in the A and B groups. Compared with the control subgroup, the expression level of PSD-95 and AMPA receptors in the hippocampus was decreased at T1 and T2 in the sevoflurane subgroup (P < 0.05). The nerve cells were partially swelling. Electrophysiology analysis showed that the levels of PSD-95 and AMPA receptor expression were associated with synaptic transmission. Conclusion Sevoflurane impaired short-term memory in adult mice by inhibiting the expression of PSD-95 and AMPA receptors in the hippocampus, which led to the decrease in synaptic transmission.
Collapse
|
27
|
Nakazawa K, Sapkota K. The origin of NMDA receptor hypofunction in schizophrenia. Pharmacol Ther 2019; 205:107426. [PMID: 31629007 DOI: 10.1016/j.pharmthera.2019.107426] [Citation(s) in RCA: 157] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 10/10/2019] [Indexed: 12/12/2022]
Abstract
N-methyl-d-aspartate (NMDA) receptor (NMDAR) hypofunction plays a key role in pathophysiology of schizophrenia. Since NMDAR hypofunction has also been reported in autism, Alzheimer's disease and cognitive dementia, it is crucial to identify the location, timing, and mechanism of NMDAR hypofunction for schizophrenia for better understanding of disease etiology and for novel therapeutic intervention. In this review, we first discuss the shared underlying mechanisms of NMDAR hypofunction in NMDAR antagonist models and the anti-NMDAR autoantibody model of schizophrenia and suggest that NMDAR hypofunction could occur in GABAergic neurons in both models. Preclinical models using transgenic mice have shown that NMDAR hypofunction in cortical GABAergic neurons, in particular parvalbumin-positive fast-spiking interneurons, in the early postnatal period confers schizophrenia-related phenotypes. Recent studies suggest that NMDAR hypofunction can also occur in PV-positive GABAergic neurons with alterations of NMDAR-associated proteins, such as neuregulin/ErbB4, α7nAChR, and serine racemase. Furthermore, several environmental factors, such as oxidative stress, kynurenic acid and hypoxia, may also potentially elicit NMDAR hypofunction in GABAergic neurons in early postnatal period. Altogether, the studies discussed here support a central role for GABAergic abnormalities in the context of NMDAR hypofunction. We conclude by suggesting potential therapeutic strategies to improve the function of fast-spiking neurons.
Collapse
|
28
|
Balu DT, Pantazopoulos H, Huang CCY, Muszynski K, Harvey TL, Uno Y, Rorabaugh JM, Galloway CR, Botz-Zapp C, Berretta S, Weinshenker D, Coyle JT. Neurotoxic astrocytes express the d-serine synthesizing enzyme, serine racemase, in Alzheimer's disease. Neurobiol Dis 2019; 130:104511. [PMID: 31212068 PMCID: PMC6689433 DOI: 10.1016/j.nbd.2019.104511] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/07/2019] [Accepted: 06/14/2019] [Indexed: 12/21/2022] Open
Abstract
Although β-amyloid plaques are a well-recognized hallmark of Alzheimer's disease (AD) neuropathology, no drugs reducing amyloid burden have shown efficacy in clinical trials, suggesting that once AD symptoms emerge, disease progression becomes independent of Aβ production. Reactive astrocytes are another neuropathological feature of AD, where there is an emergence of neurotoxic (A1) reactive astrocytes. We find that serine racemase (SR), the neuronal enzyme that produces the N-methyl-d-aspartate receptor (NMDAR) co-agonist d-serine, is robustly expressed in A1-reactive neurotoxic astrocytes in the hippocampus and entorhinal cortex of AD subjects and an AD rat model. Furthermore, we observe intracellular signaling changes consistent with increased extra-synaptic NMDAR activation, excitotoxicity and decreased neuronal survival. Thus, reducing neurotoxic d-serine release from A1 inflammatory astrocytes could have therapeutic benefit for mild to advanced AD, when anti-amyloid strategies are ineffective.
Collapse
Affiliation(s)
- Darrick T Balu
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA.
| | - Harry Pantazopoulos
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA, USA
| | - Cathy C Y Huang
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA
| | - Kevin Muszynski
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA
| | - Theresa Lynn Harvey
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA
| | - Yota Uno
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Translational Psychiatry Laboratory, McLean Hospital, Belmont, MA, USA
| | - Jacki M Rorabaugh
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Claire R Galloway
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Christian Botz-Zapp
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Sabina Berretta
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Translational Neuroscience Laboratory, McLean Hospital, Belmont, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - David Weinshenker
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Joseph T Coyle
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA; Laboratory of Psychiatric and Molecular Neuroscience, McLean Hospital, Belmont, MA, USA
| |
Collapse
|
29
|
The NMDA receptor activation by d-serine and glycine is controlled by an astrocytic Phgdh-dependent serine shuttle. Proc Natl Acad Sci U S A 2019; 116:20736-20742. [PMID: 31548413 DOI: 10.1073/pnas.1909458116] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Astrocytes express the 3-phosphoglycerate dehydrogenase (Phgdh) enzyme required for the synthesis of l-serine from glucose. Astrocytic l-serine was proposed to regulate NMDAR activity by shuttling to neurons to sustain d-serine production, but this hypothesis remains untested. We now report that inhibition of astrocytic Phgdh suppressed the de novo synthesis of l-and d-serine and reduced the NMDAR synaptic potentials and long-term potentiation (LTP) at the Schaffer collaterals-CA1 synapse. Likewise, enzymatic removal of extracellular l-serine impaired LTP, supporting an l-serine shuttle mechanism between glia and neurons in generating the NMDAR coagonist d-serine. Moreover, deletion of serine racemase (SR) in glutamatergic neurons abrogated d-serine synthesis to the same extent as Phgdh inhibition, suggesting that neurons are the predominant source of the newly synthesized d-serine. We also found that the synaptic NMDAR activation in adult SR-knockout (KO) mice requires Phgdh-derived glycine, despite the sharp decline in the postnatal glycine levels as a result of the emergence of the glycine cleavage system. Unexpectedly, we also discovered that glycine regulates d-serine metabolism by a dual mechanism. The first consists of tonic inhibition of SR by intracellular glycine observed in vitro, primary cultures, and in vivo microdialysis. The second involves a transient glycine-induce d-serine release through the Asc-1 transporter, an effect abolished in Asc-1 KO mice and diminished by deleting SR in glutamatergic neurons. Our observations suggest that glycine is a multifaceted regulator of d-serine metabolism and implicate both d-serine and glycine in mediating NMDAR synaptic activation at the mature hippocampus through a Phgdh-dependent shuttle mechanism.
Collapse
|
30
|
Chen XQ, Qiu K, Liu H, He Q, Bai JH, Lu W. Application and prospects of butylphthalide for the treatment of neurologic diseases. Chin Med J (Engl) 2019; 132:1467-1477. [PMID: 31205106 PMCID: PMC6629339 DOI: 10.1097/cm9.0000000000000289] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Indexed: 01/30/2023] Open
Abstract
OBJECTIVE The 3-N-butylphthalide (NBP) comprises one of the chemical constituents of celery oil. It has a series of pharmacologic mechanisms including reconstructing microcirculation, protecting mitochondrial function, inhibiting oxidative stress, inhibiting neuronal apoptosis, etc. Based on the complex multi-targets of pharmacologic mechanisms of NBP, the clinical application of NBP is increasing and more clinical researches and animal experiments are also focused on NBP. The aim of this review was to comprehensively and systematically summarize the application of NBP on neurologic diseases and briefly summarize its application to non-neurologic diseases. Moreover, recent progress in experimental models of NBP on animals was summarized. DATA SOURCES Literature was collected from PubMed and Wangfang database until November 2018, using the search terms including "3-N-butylphthalide," "microcirculation," "mitochondria," "ischemic stroke," "Alzheimer disease," "vascular dementia," "Parkinson disease," "brain edema," "CO poisoning," "traumatic central nervous system injury," "autoimmune disease," "amyotrophic lateral sclerosis," "seizures," "diabetes," "diabetic cataract," and "atherosclerosis." STUDY SELECTION Literature was mainly derived from English articles or articles that could be obtained with English abstracts and partly derived from Chinese articles. Article type was not limited. References were also identified from the bibliographies of identified articles and the authors' files. RESULTS NBP has become an important adjunct for ischemic stroke. In vascular dementia, the clinical application of NBP to treat severe cognitive dysfunction syndrome caused by the hypoperfusion of brain tissue during cerebrovascular disease is also increasing. Evidence also suggests that NBP has a therapeutic effect for neurodegenerative diseases. Many animal experiments have found that it can also improve symptoms in other neurologic diseases such as epilepsy, cerebral edema, and decreased cognitive function caused by severe acute carbon monoxide poisoning. Moreover, NBP has therapeutic effects for diabetes, diabetes-induced cataracts, and non-neurologic diseases such as atherosclerosis. Mechanistically, NBP mainly improves microcirculation and protects mitochondria. Its broad pharmacologic effects also include inhibiting oxidative stress, nerve cell apoptosis, inflammatory responses, and anti-platelet and anti-thrombotic effects. CONCLUSIONS The varied pharmacologic mechanisms of NBP involve many complex molecular mechanisms; however, there many unknown pharmacologic effects await further study.
Collapse
Affiliation(s)
- Xi-Qian Chen
- Department of Neurology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | | | | | | | | | | |
Collapse
|
31
|
Luo H, Wu PF, Han QQ, Cao Y, Deng SL, Wang J, Deng Q, Wang F, Chen JG. Reactive Sulfur Species Emerge as Gliotransmitters to Support Memory via Sulfuration-Dependent Gating of NR2A-Containing N-Methyl-d-Aspartate Subtype Glutamate Receptor Function. Antioxid Redox Signal 2019; 30:1880-1899. [PMID: 30187770 DOI: 10.1089/ars.2018.7503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
AIMS Astrocytes have been revealed as a controller of synaptic plasticity and memory via releasing gliotransmitters. Our recent findings showed that reactive sulfur species (RSS), including hydrogen sulfide (H2S) and polysulfide (H2Sn), regulated the availability of d-serine, which is a well-known gliotransmitter that is involved in synaptic plasticity. An interesting question is whether RSS, which are small molecules, can function as direct gliotransmitters to integrate astrocyte-neuron interactions throughout the memory process. RESULTS We found that hippocampal RSS level increased significantly in response to learning. We further demonstrated that the activity-triggered RSS signal controlled memory formation by using pharmacological and genetic approaches. The RSS-supporting memory was primarily conferred by enzymes that were mainly located in astrocytes, including cystathionine β-synthase (CBS) and mercaptopyruvate sulfurtransferase (3-MST), and the memory-promoting effects were mostly dependent on sulfration of the NR2A subunit of N-methyl-d-aspartate subtype glutamate receptors (NMDARs). Further, RSS were demonstrated to buffer the strong inhibitory effect of synaptically released zinc on NR2A-containing NMDARs. Innovation and Conclusion: These results suggest that glial-derived RSS signals can serve as direct gliotransmitters that regulate memory formation through the redox modulation of postsynaptic receptors; this conclusion will enrich the gliotransmission hypothesis.
Collapse
Affiliation(s)
- Han Luo
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng-Fei Wu
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian-Qian Han
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Cao
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Si-Long Deng
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji Wang
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiao Deng
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Wang
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2 Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.,3 The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.,4 Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China.,5 The Collaborative-Innovation Center for Brain Science, Wuhan, China
| | - Jian-Guo Chen
- 1 Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,2 Key Laboratory of Neurological Diseases (HUST), Ministry of Education of China, Wuhan, China.,3 The Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China.,4 Laboratory of Neuropsychiatric Diseases, The Institute of Brain Research, Huazhong University of Science and Technology, Wuhan, China.,5 The Collaborative-Innovation Center for Brain Science, Wuhan, China
| |
Collapse
|
32
|
MacKay MAB, Kravtsenyuk M, Thomas R, Mitchell ND, Dursun SM, Baker GB. D-Serine: Potential Therapeutic Agent and/or Biomarker in Schizophrenia and Depression? Front Psychiatry 2019; 10:25. [PMID: 30787885 PMCID: PMC6372501 DOI: 10.3389/fpsyt.2019.00025] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 01/15/2019] [Indexed: 11/17/2022] Open
Abstract
D-Serine is a potent co-agonist at the NMDA glutamate receptor and has been the object of many preclinical studies to ascertain the nature of its metabolism, its regional and cellular distribution in the brain, its physiological functions and its possible clinical relevance. The enzymes involved in its formation and catabolism are serine racemase (SR) and D-amino acid oxidase (DAAO), respectively, and manipulations of the activity of those enzymes have been useful in developing animal models of schizophrenia and in providing clues to the development of potential new antipsychotic strategies. Clinical studies have been conducted in schizophrenia patients to evaluate body fluid levels of D-serine and/or to use D-serine alone or in combination with antipsychotics to determine its effectiveness as a therapeutic agent. D-serine has also been used in combination with DAAO inhibitors in preclinical investigations, and interesting results have been obtained. Genetic studies and postmortem brain studies have also been conducted on D-serine and the enzymes involved in its metabolism. It is also of considerable interest that in recent years clinical and preclinical investigations have suggested that D-serine may also have antidepressant properties. Clinical studies have also shown that D-serine may be a biomarker for antidepressant response to ketamine. Relevant to both schizophrenia and depression, preclinical and clinical studies with D-serine indicate that it may be effective in reducing cognitive dysfunction.
Collapse
Affiliation(s)
- Mary-Anne B MacKay
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Maryana Kravtsenyuk
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Rejish Thomas
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Nicholas D Mitchell
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Serdar M Dursun
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen B Baker
- Neurochemical Research Unit and Bebensee Schizophrenia Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
33
|
Billard JM. Changes in Serine Racemase-Dependent Modulation of NMDA Receptor: Impact on Physiological and Pathological Brain Aging. Front Mol Biosci 2018; 5:106. [PMID: 30555832 PMCID: PMC6282039 DOI: 10.3389/fmolb.2018.00106] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/09/2018] [Indexed: 02/02/2023] Open
Abstract
The N-methyl-D-Aspartate glutamate receptors (NMDARs) are pivotal for the functional and morphological plasticity that are required in neuronal networks for efficient brain activities and notably for cognitive-related abilities. Because NMDARs are heterogeneous in subunit composition and associated with multiple functional regulatory sites, their efficacy is under the tonic influence of numerous allosteric modulations, whose dysfunction generally represents the first step generating pathological states. Among the enzymatic candidates, serine racemase (SR) has recently gathered an increasing interest considering that it tightly regulates the production of d-serine, an amino acid now viewed as the main endogenous co-agonist necessary for NMDAR activation. Nowadays, SR deregulation is associated with a wide range of neurological and psychiatric diseases including schizophrenia, amyotrophic lateral sclerosis, and depression. This review aims at compelling the most recent experimental evidences indicating that changes in SR-related modulation of NMDARs also govern opposite functional dysfunctions in physiological and pathological (Alzheimer's disease) aging that finally results in memory disabilities in both cases. It also highlights SR as a relevant alternative target for new pharmacological strategies aimed at preventing functional alterations and cognitive impairments linked to the aging process.
Collapse
|
34
|
Neuronal serine racemase associates with Disrupted-In-Schizophrenia-1 and DISC1 agglomerates: Implications for schizophrenia. Neurosci Lett 2018; 692:107-114. [PMID: 30391323 DOI: 10.1016/j.neulet.2018.10.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/18/2018] [Accepted: 10/29/2018] [Indexed: 12/21/2022]
Abstract
D-Serine, an endogenous coagonist of N-methyl-d-aspartate receptors (NMDARs) at the glycine binding site, is synthesized by serine racemase (SR) through conversion of l-Serine. Dysregulation of SR/D-Serine and Disrupted-In-Schizophrenia-1 (DISC1) contributes to the pathogenesis of schizophrenia at converging pathways, as perturbation of SR-DISC1 binding in astrocytes elicits schizophrenia-like behaviors in mice. However, an association of neuronal SR with DISC1 remains elusive. Here we report that SR associates with DISC1 and its agglomerates in cortical neurons, which can be modulated by NMDAR activity. Endogenous SR colocalizes with DISC1 large agglomerates in the soma and with smaller puncta in the nucleus and dendrites of cortical neurons. Co-immunoprecipitation assays demonstrate SR interaction with DISC1 in cortical neuronal lysates, suggesting the physiological presence of functional SR-DISC1 complexes in neurons. Moreover, exogenous d-Serine application significantly increases the interaction of SR with DISC1, the number of DISC1-SR large agglomerates and the levels of DISC1 agglomerated form along with SR in the triton-insoluble pellet fraction, whereas application of glycine with a glycine transporter inhibitor fails to increase their interactions, abundance of DISC1-SR large agglomerates and levels of DISC1 agglomerated form. This increase by d-Serine application is blocked by 7-chlorokynurenic acid, a specific antagonist at the glycine site of NMDARs, suggesting mediation through NMDARs. Our findings thus demonstrate neuronal SR association with DISC1 and its agglomerates, which can be modulated by d-Serine, thereby validating a novel neuronal SR-DISC1 complex responsive to NMDAR activation and providing a molecular mechanism by which pathways implicated in schizophrenia converge.
Collapse
|
35
|
Lv C, Ma Q, Han B, Li J, Geng Y, Zhang X, Wang M. Long-Term DL-3- n-Butylphthalide Treatment Alleviates Cognitive Impairment Correlate With Improving Synaptic Plasticity in SAMP8 Mice. Front Aging Neurosci 2018; 10:200. [PMID: 30026693 PMCID: PMC6041467 DOI: 10.3389/fnagi.2018.00200] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/13/2018] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is the most prevalent form of dementia worldwide. AD is characterized by mild cognitive impairment at onset, irreversibly progressing with age to severe neurodegeneration and cognitive deficits in the late stages. Unfortunately, no effective treatments exist to prevent or delay the cognitive symptoms of AD. Studies have shown that DL-3-n-butylphthalide (DL-NBP) alleviates cognitive impairment induced by amyloid-β in mice by reducing oxidative stress, inhibiting apoptosis, and decreasing tau phosphorylation. In this study, we examined the effects of DL-NBP administration on cognitive function in the senescence-accelerated mouse prone 8 (SAMP8) model of age-related dementia. DL-NBP treatment for 3 months alleviated cognitive impairment in SAMP8 mice as assessed by performance in the Morris water maze test. Moreover, DL-NBP significantly increased the expression of synaptophysin and postsynaptic density protein 95 in the hippocampus of SAMP8 mice, indicative of a protective effect on hippocampal structural synaptic plasticity. In addition, brain-derived neurotrophic factor/tropomyosin receptor kinase B signaling, previously shown to promote synaptic plasticity, was significantly enhanced by the DL-NBP administration. Our findings suggest that DL-NBP is a potential drug candidate for the treatment of cognitive impairment in AD and may serve as the foundation for further research into the development of AD drugs.
Collapse
Affiliation(s)
- Chaonan Lv
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, China
| | - Qinying Ma
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, China
| | - Bing Han
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, China
| | - Jing Li
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, China
| | - Yuan Geng
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, China
| | - Xiaoman Zhang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, China
| | - Mingwei Wang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China.,Brain Aging and Cognitive Neuroscience Key Laboratory of Hebei Province, Shijiazhuang, China
| |
Collapse
|
36
|
Serine Racemase and D-serine in the Amygdala Are Dynamically Involved in Fear Learning. Biol Psychiatry 2018; 83:273-283. [PMID: 29025687 PMCID: PMC5806199 DOI: 10.1016/j.biopsych.2017.08.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 08/15/2017] [Accepted: 08/16/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND The amygdala is a central component of the neural circuitry that underlies fear learning. N-methyl-D-aspartate receptor-dependent plasticity in the amygdala is required for pavlovian fear conditioning and extinction. N-methyl-D-aspartate receptor activation requires the binding of a coagonist, D-serine, which is synthesized from L-serine by the neuronal enzyme serine racemase (SR). However, little is known about SR and D-serine function in the amygdala. METHODS We used immunohistochemical methods to characterize the cellular localization of SR and D-serine in the mouse and human amygdala. Using biochemical and molecular techniques, we determined whether trace fear conditioning and extinction engages the SR/D-serine system in the brain. D-serine was administered systemically to mice to evaluate its effect on fear extinction. Finally, we investigated whether the functional single nucleotide polymorphism rs4523957, which is an expression quantitative trait locus of the human serine racemase (SRR) gene, was associated with fear-related phenotypes in a highly traumatized human cohort. RESULTS We demonstrate that approximately half of the neurons in the amygdala express SR, including both excitatory and inhibitory neurons. We find that the acquisition and extinction of fear memory engages the SR/D-serine system in the mouse amygdala and that D-serine administration facilitates fear extinction. We also demonstrate that the SRR single nucleotide polymorphism, rs4523957, is associated with posttraumatic stress disorder in humans, consistent with the facilitatory effect of D-serine on fear extinction. CONCLUSIONS These new findings have important implications for understanding D-serine-mediated N-methyl-D-aspartate receptor plasticity in the amygdala and how this system could contribute to disorders with maladaptive fear circuitry.
Collapse
|
37
|
|
38
|
Coyle JT, Balu DT. The Role of Serine Racemase in the Pathophysiology of Brain Disorders. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:35-56. [PMID: 29413527 PMCID: PMC5821497 DOI: 10.1016/bs.apha.2017.10.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The N-methyl-d-aspartate receptor (NMDAR) is unique in requiring two agonists to bind simultaneously to open its cation channel: the neurotransmitter, glutamate, and the coagonists, glycine, or d-serine. The Snyder laboratory was the first to clone serine racemase (SR), the enzyme that synthesizes d-serine, and to localize it immunocytochemically. Our laboratory has focused on the role of d-serine in brain disorders. Silencing the expression of SR, a risk gene for schizophrenia (SCZ), in mice (SR-/-), results in a phenotype that closely resembles SCZ including: cortical atrophy, reduced dendritic spine density and complexity, downregulation of parvalbumin-positive cortical GABAergic neurons, and cognitive impairments. This pathology can be reversed by treatment of SR-/- mice with d-serine in adulthood. SR-/- mice also exhibit abnormal response toward abusable substances, such as stimulants. They show reduced behavioral sensitization to d-amphetamine, but fail to extinguish it. Place preference to cocaine is altered, and the hedonic response to it is profoundly impaired as assessed by intracranial self-stimulation. d-cycloserine, a partial agonist at the NMDAR glycine modulatory site, shows therapeutic benefit for treating pathologic anxiety in combination with behavioral therapies. Studies in vitro with cortical culture and in vivo with middle cerebral artery occlusion show that silencing SR provides substantial protection against ischemic neuronal death. Finally, the switch of SR expression from neurons to reactive astrocytes after closed head trauma accounts for the reduced in vivo neuroplasticity, electroencephalogram abnormalities, and cognitive impairments.
Collapse
Affiliation(s)
- Joseph T Coyle
- Harvard Medical School, Boston, MA, United States; McLean Hospital, Belmont, MA, United States.
| | - Darrick T Balu
- Harvard Medical School, Boston, MA, United States; McLean Hospital, Belmont, MA, United States
| |
Collapse
|
39
|
Brain-Derived Neurotrophic Factor Increases Synaptic Protein Levels via the MAPK/Erk Signaling Pathway and Nrf2/Trx Axis Following the Transplantation of Neural Stem Cells in a Rat Model of Traumatic Brain Injury. Neurochem Res 2017; 42:3073-3083. [PMID: 28780733 DOI: 10.1007/s11064-017-2340-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 06/05/2017] [Accepted: 06/20/2017] [Indexed: 01/28/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) plays an important role in promoting the growth, differentiation, survival and synaptic stability of neurons. Presently, the transplantation of neural stem cells (NSCs) is known to induce neural repair to some extent after injury or disease. In this study, to investigate whether NSCs genetically modified to encode the BDNF gene (BDNF/NSCs) would further enhance synaptogenesis, BDNF/NSCs or naive NSCs were directly engrafted into lesions in a rat model of traumatic brain injury (TBI). Immunohistochemistry, western blotting and RT-PCR were performed to detect synaptic proteins, BDNF-TrkB and its downstream signaling pathways, at 1, 2, 3 or 4 weeks after transplantation. Our results showed that BDNF significantly increased the expression levels of the TrkB receptor gene and the phosphorylation of the TrkB protein in the lesions. The expression levels of Ras, phosphorylated Erk1/2 and postsynaptic density protein-95 were elevated in the BDNF/NSCs-transplanted groups compared with those in the NSCs-transplanted groups throughout the experimental period. Moreover, the nuclear factor (erythroid-derived 2)-like 2/Thioredoxin (Nrf2/Trx) axis, which is a specific therapeutic target for the treatment of injury or cell death, was upregulated by BDNF overexpression. Therefore, we determined that the increased synaptic proteins level implicated in synaptogenesis might be associated with the activation of the MAPK/Erk1/2 signaling pathway and the upregulation of the antioxidant agent Trx modified by BDNF-TrkB following the BDNF/NSCs transplantation after TBI.
Collapse
|
40
|
Gorska-Ponikowska M, Perricone U, Kuban-Jankowska A, Lo Bosco G, Barone G. 2-methoxyestradiol impacts on amino acids-mediated metabolic reprogramming in osteosarcoma cells by its interaction with NMDA receptor. J Cell Physiol 2017; 232:3030-3049. [PMID: 28262924 DOI: 10.1002/jcp.25888] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 03/02/2017] [Indexed: 12/28/2022]
Abstract
Deregulation of serine and glycine metabolism, have been identified to function as metabolic regulators in supporting tumor cell growth. The role of serine and glycine in regulation of cancer cell proliferation is complicated, dependent on concentrations of amino acids and tissue-specific. D-serine and glycine are coagonists of N-methyl-D-aspartate (NMDA) receptor subunit GRIN1. Importantly, NMDA receptors are widely expressed in cancer cells and play an important role in regulation of cell death, proliferation, and metabolism of numerous malignancies. The aim of the present work was to associate the metabolism of glycine and D-serine with the anticancer activity of 2-methoxyestradiol. 2-methoxyestradiol is a potent anticancer agent but also a physiological 17β- estradiol metabolite. In the study we have chosen two malignant cell lines expressing functional NMDA receptors, that is osteosarcoma 143B and breast cancer MCF7. We used MTS assay, migration assay, flow cytometric analyses, Western blotting and immunoprecipitation techniques as well as molecular modeling studies. We have demonstrated the extensive crosstalk between the deregulated metabolic network and cancer cell signaling. Herein, we observed an anticancer effect of high concentrations of glycine and D-serine in osteosarcoma cells. In contrast, the amino acids when used at low, physiological concentrations induced the proliferation and migration of osteosarcoma cells. Importantly, the pro-cancergogenic effects of both glycine and D-serine where abrogated by the usage of 2-methoxyestradiol at both physiological and pharmacological relevant concentrations. The obtained data confirmed that 2-methoxyestradiol may be a physiological anticancer molecule.
Collapse
Affiliation(s)
| | - Ugo Perricone
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, Edificio 17, Palermo, Italy.,Fondazione Ri.MED, Palermo, Italy
| | | | - Giosuè Lo Bosco
- Dipartimento di Matematica e Informatica, Università degli Studi di Palermo, Palermo, Italy.,Istituto Euro Mediterraneo di Scienza e Tecnologia, Palermo, Italy
| | - Giampaolo Barone
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Università degli Studi di Palermo, Viale delle Scienze, Edificio 17, Palermo, Italy
| |
Collapse
|