1
|
Kamat PK, Kalani A, Debnath N, Mushtaq Z, Tyagi SC, Tyagi N. Neuroprotective mechanism of hydrogen sulfide in okadaic acid-induced alzheimer-like pathology. GeroScience 2025:10.1007/s11357-025-01662-1. [PMID: 40341601 DOI: 10.1007/s11357-025-01662-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/09/2025] [Indexed: 05/10/2025] Open
Abstract
Okadaic acid (OKA) is a marine biotoxin that accumulates in shellfish and is responsible for causing diarrheic shellfish poisoning. OKA is a powerful and selective inhibitor of serine/threonine phosphatases 1 and 2A, which induces hyperphosphorylation of tau in vitro and in vivo leading to Alzheimer's disease (AD)-like pathology and memory impairment. Hydrogen sulfide (H2S), a gaseous signaling molecule produced endogenously in the brain, has been demonstrated to possess neuroprotective properties in various models of neurodegeneration. The aim of this study was to investigate the potential of H₂S in reducing OKA-induced Alzheimer's disease (AD)-like pathology, focusing on its effects on the GSK3β/Tau and CaMKII/CREB signaling pathways in mice. To test this hypothesis, we used age 8-10 weeks-old male C57BL/6J wild-type mice, divided into the following experimental groups: 1. Control group: Received a single intracerebroventricular (ICV) injection of artificial cerebrospinal fluid (aCSF). 2. WT + OKA group: Received a single ICV injection of OKA (100 ng/5 µl) bilaterally to induce AD-like pathology. OKA was dissolved in artificial cerebrospinal fluid. 3. WT + OKA + GYY4137 group: Received a single ICV injection of OKA (100 ng/5 µl) bilaterally, followed by GYY4137 (30 µM/kg) via drinking water for 21 days. 4. WT + GYY4137 group: Received only GYY4137 per se (30 µM/kg) via drinking water for 21 days. After the treatment period, synaptic proteins and neurodegeneration were evaluated using Western blotting, RT-PCR, and immunohistochemistry techniques. Our results demonstrate that OKA administration results in memory impairment with decreased cerebral blood flow (CBF). OKA also caused a significant decrease in synapse proteins (PSD95, MAP-2, BDNF, CaMKIIα, and Tubulin-3β) levels, along with increased expression of Tau, PHF-1, and GSK-3β and memory-associated signaling molecules and pCREB. Interestingly, IP administration of GYY4137 (30 µM/Kg; an H2S donor) for 21 days significantly improved the level of synapse proteins and memory function in OKA-treated mice. The findings of this study determine the neuroprotective mechanism of H2S in OKA-induced AD-like pathology through the modulation of Tau, GSK3β, and pCREB signaling. Therefore, H2S ameliorates OKA-induced memory impairment by improving synapse function and forgetfulness. As a result, H2S could be used as a promising therapeutic molecule against Alzheimer's disease-like pathology.
Collapse
Affiliation(s)
- Pradip K Kamat
- Departments of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Anuradha Kalani
- Disease Biology Lab and Molecular Oncology Lab, Department of Life Science and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, 208024, India
| | - Nabendu Debnath
- Centre for Molecular Biology, Central University of Jammu, Bagla (Raya-Suchani), District Samba, 181143, Jammu & Kashmir, India
| | - Zayd Mushtaq
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, A- 1201, Louisville, KY, 40202, USA
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, A- 1201, Louisville, KY, 40202, USA
| | - Neetu Tyagi
- Department of Physiology, School of Medicine, Health Sciences Center, University of Louisville, A- 1201, Louisville, KY, 40202, USA.
| |
Collapse
|
2
|
Reinders NR, van der Spek SJF, Klaassen RV, Koymans KJ, MacGillavry HD, Smit AB, Kessels HW. Amyloid-β-Driven Synaptic Deficits Are Mediated by Synaptic Removal of GluA3-Containing AMPA Receptors. J Neurosci 2025; 45:e0393242024. [PMID: 39779375 PMCID: PMC11867010 DOI: 10.1523/jneurosci.0393-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
The detrimental effects of oligomeric amyloid-β (Aβ) on synapses are considered the leading cause for cognitive deficits in Alzheimer's disease. However, through which mechanism Aβ oligomers impair synaptic structure and function remains unknown. Here, we used electrophysiology and amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) imaging on mouse and rat neurons to demonstrate that GluA3 expression in neurons lacking GluA3 is sufficient to resensitize their synapses to the damaging effects of Aβ, indicating that GluA3-containing AMPARs at synapses are necessary and sufficient for Aβ to induce synaptic deficits. We found that Aβ oligomers trigger the endocytosis of GluA3 and promote its translocation toward endolysosomal compartments for degradation. Mechanistically, these Aβ-driven effects critically depend on the PDZ-binding motif of GluA3. A single point mutation in the GluA3 PDZ-binding motif prevented Aβ-driven effects and rendered synapses fully resistant to the effects of Aβ. Correspondingly, proteomics on synaptosome fractions from APP/PS1-transgenic mice revealed a selective reduction of GluA3 at an early age. These findings support a model where the endocytosis and lysosomal degradation of GluA3-containing AMPARs are a critical early step in the cascade of events through which Aβ accumulation causes a loss of synapses.
Collapse
Affiliation(s)
- Niels R Reinders
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105 BA, The Netherlands
- Swammerdam Institute of Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - Sophie J F van der Spek
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Remco V Klaassen
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Karin J Koymans
- Swammerdam Institute of Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
| | - Harold D MacGillavry
- Division of Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht 3584 CH, The Netherlands
| | - August B Smit
- Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Helmut W Kessels
- Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Amsterdam 1105 BA, The Netherlands
- Swammerdam Institute of Life Sciences, Amsterdam Neuroscience, University of Amsterdam, Amsterdam 1098 XH, The Netherlands
| |
Collapse
|
3
|
Chen KH, Yang J, Liu B, Jiang C, Koylass N, Zhang Z, Sun S, Huganir R, Qiu Z. Loss of the proton-activated chloride channel in neurons impairs AMPA receptor endocytosis and LTD via endosomal hyper-acidification. Cell Rep 2025; 44:115302. [PMID: 39946237 PMCID: PMC11938102 DOI: 10.1016/j.celrep.2025.115302] [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: 09/10/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025] Open
Abstract
Hippocampal long-term potentiation (LTP) and long-term depression (LTD) are forms of synaptic plasticity, thought to be the molecular basis of learning and memory, dependent on dynamic α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking. Alteration of endosomal pH negatively affects synaptic transmission and neural development, but it is unclear how pH is involved in AMPAR trafficking. We show that the proton-activated chloride (PAC) channel localizes to early and recycling endosomes in neurons and prevents endosome hyper-acidification. Loss of PAC reduces AMPAR endocytosis during chemical LTD in primary neurons, while basal trafficking and LTP are unaffected. Pyramidal neuron-specific PAC knockout mice have impaired hippocampal LTD, but not LTP, and perform poorly in the Morris water maze reversal test, exhibiting impaired behavioral adaptation. We conclude that proper maintenance of endosomal pH by PAC in neurons is important during LTD to regulate AMPAR trafficking in a manner critical for animal physiology and behavior.
Collapse
Affiliation(s)
- Kevin H Chen
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Junhua Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843, USA
| | - Bian Liu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chaohua Jiang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nicholas Koylass
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhe Zhang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shuying Sun
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhaozhu Qiu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
4
|
Choquet D, Opazo P, Zhang H. AMPA receptor diffusional trapping machinery as an early therapeutic target in neurodegenerative and neuropsychiatric disorders. Transl Neurodegener 2025; 14:8. [PMID: 39934896 PMCID: PMC11817889 DOI: 10.1186/s40035-025-00470-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025] Open
Abstract
Over the past two decades, there has been a growing recognition of the physiological importance and pathological implications surrounding the surface diffusion of AMPA receptors (AMPARs) and their diffusional trapping at synapses. AMPAR surface diffusion entails the thermally powered random Brownian lateral movement of these receptors within the plasma membrane, facilitating dynamic exchanges between synaptic and extrasynaptic compartments. This process also enables the activity-dependent diffusional trapping and accumulation of AMPARs at synapses through transient binding to synaptic anchoring slots. Recent research highlights the critical role of synaptic recruitment of AMPARs via diffusional trapping in fundamental neural processes such as the development of the early phases of long-term potentiation (LTP), contextual fear memory, memory consolidation, and sensory input-induced cortical remapping. Furthermore, studies underscore that regulation of AMPAR diffusional trapping is altered across various neurological disease models, including Huntington's disease (HD), Alzheimer's disease (AD), and stress-related disorders like depression. Notably, pharmacological interventions aimed at correcting deficits in AMPAR diffusional trapping have demonstrated efficacy in restoring synapse numbers, LTP, and memory functions in these diverse disease models, despite their distinct pathogenic mechanisms. This review provides current insights into the molecular mechanisms underlying the dysregulation of AMPAR diffusional trapping, emphasizing its role as a converging point for multiple pathological signaling pathways. We propose that targeting AMPAR diffusional trapping represents a promising early therapeutic strategy to mitigate synaptic plasticity and memory deficits in a spectrum of brain disorders, encompassing but not limited to HD, AD, and stress-related conditions. This approach underscores an integrated therapeutic target amidst the complexity of these neurodegenerative and neuropsychiatric diseases.
Collapse
Affiliation(s)
- Daniel Choquet
- Univ. Bordeaux, CNRS, Interdisciplinary Institute for Neuroscience, IINS, UMR 5297, 33000, Bordeaux, France
- Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UAR 3420, US 4, 33000, Bordeaux, France
| | - Patricio Opazo
- UK Dementia Research Institute, Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh, EH16 4SB, UK
| | - Hongyu Zhang
- Department of Biomedicine, University of Bergen, 5009, Bergen, Norway.
- Mohn Research Center for the Brain, University of Bergen, 5009, Bergen, Norway.
- Department of Radiology, Haukeland University Hospital, 5021, Bergen, Norway.
| |
Collapse
|
5
|
Chen Y, Chen S, Wang H, Cao P. Artemisinin antagonizes cognitive disorder in hyperuricemia through glutamate receptor-interacting protein 1-suppressed pyroptosis. Neuroreport 2025; 36:145-152. [PMID: 39976052 DOI: 10.1097/wnr.0000000000002131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The prevalence of hyperuricemia (HUA) is climbing worldwide and persistent elevation of serum uric acid impairs cognitive function. This study aimed to explore the mechanisms of Artemisinin (Art) antagonizing cognitive disorder in HUA by suppressing pyroptosis. A mouse model of HUA was established by intraperitoneal injection of 300 mg/kg potassium oxonate (PO) in C57BL/6 mice for 14 days. The mice were simultaneously treated with Art, an agonist of pyroptosis Polyphyllin VI (PPVI), or glutamate receptor-interacting protein 1 (GRIP1) knockdown lentiviral plasmid. After treatment, serum uric acid, IL-6, and TNF-ɑ levels were examined, as well as hippocampal IL-1β and IL-18 levels, and the cognitive function of mice was assessed by the Morris water maze test. Pathological changes in the CA1 of the hippocampus were observed. Cleave-caspase-1, GSDMD-N, and GRIP1 protein level in the hippocampus was quantified by western blot. After PO induction, the escape latency and the time spent in the target quadrant increased in mice, cell arrangement in CA1 hippocampus was loose and disorganized, with obvious inflammatory infiltration and serious damage being observed, and the mouse hippocampus had elevated cleaved-caspase-1, GSDMD-N, IL-1β, and IL-18. Art treatment reduced pyroptosis in the hippocampus and improved cognitive disorder in HUA mice. Administration of PPVI aggravated cognitive disorder in Art-treated HUA mice, and Art improved cognitive dysfunction in HUA mice by inhibiting pyroptosis through upregulation of GRIP1. Art blunts pyroptosis in the hippocampus of HUA mice suffering from cognitive disorder by upregulating GRIP1.
Collapse
Affiliation(s)
| | | | - Huiqing Wang
- Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Peng Cao
- Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
6
|
Xiong LL, Niu RZ, Chen L, Huangfu LR, Li J, Xue LL, Sun YF, Wang LM, Li YP, Liu J, Wang TH. Cross-Species Insights from Single-Nucleus Sequencing Highlight Aging-Related Hippocampal Features in Tree Shrew. Mol Biol Evol 2025; 42:msaf020. [PMID: 40036868 PMCID: PMC11879083 DOI: 10.1093/molbev/msaf020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 12/17/2024] [Accepted: 01/13/2025] [Indexed: 03/06/2025] Open
Abstract
The tree shrew brain has garnered considerable attention due to its remarkable similarities to human brain. However, the cellular composition and genetic signatures of tree shrew hippocampus across postnatal life remain poorly characterized. Here, we establish the first single-nucleus transcriptomic atlas of tree shrew hippocampus spanning postnatal life, detailing the dynamics and diversity of the neurogenic lineage, oligodendrocytes, microglia, and endothelial cells. Notably, cross-species transcriptomic comparison among humans, macaques, tree shrews, and mice reveals that the tree shrew transcriptome resembles that of macaques, making it a promising model for simulating human neurological diseases. More interestingly, we identified a unique class of tree shrew-specific neural stem cells and established SOX6, ADAMTS19, and MAP2 as their markers. Furthermore, aberrant gene expression and cellular dysfunction in the tree shrew hippocampus are linked to neuroinflammation and cognitive impairment during tree shrew aging. Our study provides extensive resources on cell composition and transcriptomic profiles, serving as a foundation for future research on neurodevelopmental and neurological disorders in tree shrews.
Collapse
Affiliation(s)
- Liu-Lin Xiong
- Department of Neurosurgery, Institute of Neurological Disease, National-Local Joint Engineering Research Center of Translational Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- Department of Anesthesiology, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi 563000, Guizhou, China
- Translational Neuromedicine Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi 563003, Guizhou, China
| | - Rui-Ze Niu
- Science and Education Department, Mental Health Center of Kunming Medical University, Kunming 650034, Yunnan, China
| | - Li Chen
- Department of Neurosurgery, Institute of Neurological Disease, National-Local Joint Engineering Research Center of Translational Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Li-Ren Huangfu
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Jing Li
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Lu-Lu Xue
- State Key Lab of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yi-Fei Sun
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Li-Mei Wang
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Yong-Ping Li
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Jia Liu
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Ting-Hua Wang
- Department of Neurosurgery, Institute of Neurological Disease, National-Local Joint Engineering Research Center of Translational Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
7
|
Zhang Y, Li T, Miao J, Zhang Z, Yang M, Wang Z, Yang B, Zhang J, Li H, Su Q, Guo J. Gamma-glutamyl transferase 5 overexpression in cerebrovascular endothelial cells improves brain pathology, cognition, and behavior in APP/PS1 mice. Neural Regen Res 2025; 20:533-547. [PMID: 38819065 PMCID: PMC11317949 DOI: 10.4103/nrr.nrr-d-23-01525] [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: 09/10/2023] [Revised: 12/18/2023] [Accepted: 02/21/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00030/figure1/v/2024-05-28T214302Z/r/image-tiff In patients with Alzheimer's disease, gamma-glutamyl transferase 5 (GGT5) expression has been observed to be downregulated in cerebrovascular endothelial cells. However, the functional role of GGT5 in the development of Alzheimer's disease remains unclear. This study aimed to explore the effect of GGT5 on cognitive function and brain pathology in an APP/PS1 mouse model of Alzheimer's disease, as well as the underlying mechanism. We observed a significant reduction in GGT5 expression in two in vitro models of Alzheimer's disease (Aβ1-42-treated hCMEC/D3 and bEnd.3 cells), as well as in the APP/PS1 mouse model. Additionally, injection of APP/PS1 mice with an adeno-associated virus encoding GGT5 enhanced hippocampal synaptic plasticity and mitigated cognitive deficits. Interestingly, increasing GGT5 expression in cerebrovascular endothelial cells reduced levels of both soluble and insoluble amyloid-β in the brains of APP/PS1 mice. This effect may be attributable to inhibition of the expression of β-site APP cleaving enzyme 1, which is mediated by nuclear factor-kappa B. Our findings demonstrate that GGT5 expression in cerebrovascular endothelial cells is inversely associated with Alzheimer's disease pathogenesis, and that GGT5 upregulation mitigates cognitive deficits in APP/PS1 mice. These findings suggest that GGT5 expression in cerebrovascular endothelial cells is a potential therapeutic target and biomarker for Alzheimer's disease.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Neurology, Sixth Hospital of Shanxi Medical University (General Hospital of Tisco), Taiyuan, Shanxi Province, China
| | - Tian Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jie Miao
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhina Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Mingxuan Yang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Zhuoran Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Bo Yang
- Department of Hernia and Abdominal Wall Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Jiawei Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Haiting Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Qiang Su
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi Province, China
- Department of Laboratory Medicine of Fenyang College, Shanxi Medical University, Fenyang, Shanxi Province, China
| | - Junhong Guo
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| |
Collapse
|
8
|
Zimmerman AJ, Serrano-Rodriguez A, Sun M, Wilson SJ, Linsenbardt DN, Brigman JL, Weick JP. Knockout of AMPA receptor binding protein Neuron-specific gene 2 (NSG2) enhances associative learning and cognitive flexibility. Mol Brain 2024; 17:95. [PMID: 39695712 DOI: 10.1186/s13041-024-01158-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
The vast majority of gene mutations and/or gene knockouts result in either no observable changes, or significant deficits in molecular, cellular, or organismal function. However, in a small number of cases, mutant animal models display enhancements in specific behaviors such as learning and memory. To date, most gene deletions shown to enhance cognitive ability generally affect a limited number of pathways such as NMDA receptor- and translation-dependent plasticity, or GABA receptor- and potassium channel-mediated inhibition. While endolysosomal trafficking of AMPA receptors is a critical mediator of synaptic plasticity, mutations in genes that affect AMPAR trafficking either have no effect or are deleterious for synaptic plasticity, learning and memory. NSG2 is one of the three-member family of Neuron-specific genes (NSG1-3), which have been shown to regulate endolysosomal trafficking of a number of proteins critical for neuronal function, including AMPAR subunits (GluA1-2). Based on these findings and the largely universal expression throughout mammalian brain, we predicted that genetic knockout of NSG2 would result in significant impairments across multiple behavioral modalities including motor, affective, and learning/memory paradigms. However, in the current study we show that loss of NSG2 had highly selective effects on associative learning and memory, leaving motor and affective behaviors intact. For instance, NSG2 KO animals performed equivalent to wild-type C57Bl/6n mice on rotarod and Catwalk motor tasks, and did not display alterations in anxiety-like behavior on open field and elevated zero maze tasks. However, NSG2 KO animals demonstrated enhanced recall in the Morris water maze, accelerated reversal learning in a touch-screen task, and accelerated acquisition and enhanced recall on a Trace Fear Conditioning task. Together, these data point to a specific involvement of NSG2 on multiple types of associative learning, and expand the repertoire of pathways that can be targeted for cognitive enhancement.
Collapse
Affiliation(s)
- Amber J Zimmerman
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
- Present Address: Division of Sleep Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Pennsylvania, PA, 19104, USA
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Antonio Serrano-Rodriguez
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - Melody Sun
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - Sandy J Wilson
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - David N Linsenbardt
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - Jonathan L Brigman
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA
| | - Jason P Weick
- Department of Neurosciences, University of New Mexico School of Medicine, 915 Camino de Salud NE, Fitz Hall 145, Albuquerque, NM, 87131, USA.
| |
Collapse
|
9
|
Li Y, Wu C, Wen X, Hu W, Diao M. LncRNA MSTRG.13,871/miR155-5p/Grip1 network involved in the post-cardiac arrest brain injury. Sci Rep 2024; 14:25108. [PMID: 39443577 PMCID: PMC11499652 DOI: 10.1038/s41598-024-75875-3] [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: 06/12/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Post-cardiac arrest brain (PCABI) is a severe medical condition characterized by a significant risk of neurological impairment and death. Nevertheless, the specific process and essential molecules responsible for its development are not fully understood. Profiling based on competing endogenous RNAs (ceRNA) has been implicated in the onset and progression of neurological disorders, yet its role in PCABI remains unclear. In this study, we performed RNA transcriptome sequencing analysis to identify differentially expressed genes in the rat model for cardiac arrest and cardiopulmonary resuscitation (CA/CPR). A hub ceRNA regulatory network was constructed using miRWalk 2.0 and Cytohubba plug-in in Cytoscape. Subsequently, real-time quantitative reverse transcription-polymerase chain reaction and dual-luciferase activity assays validated MSTRG.13,871, miR-155-5p, and Grip1 as differentially expressed in CA/CPR group, with MSTRG.13,871 capable of targeting both miR-155-5p and Grip1. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses revealed the ceRNA network enrichment in immunoregulation mechanisms such as mitochondrion, apoptotic process, and negative regulation cell death. Our research highlights the mechanism of PCABI by revealing a critical regulatory axis involving MSTRG.13,871-miR-155-5p-Grip1 in the hippocampus CA1 region after CA/CPR in rats, proposing a feasible controlled mechanism, which may serve as a theoretical basis for designing innovative therapies.
Collapse
Affiliation(s)
- Yiwei Li
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Chenghao Wu
- Department of Emergency Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin Wen
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Wei Hu
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| | - Mengyuan Diao
- Department of Critical Care Medicine, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China.
| |
Collapse
|
10
|
Dunham TL, Wilkerson JR, Johnson RC, Huganir RL, Volk LJ. WWC2 modulates GABA A-receptor-mediated synaptic transmission, revealing class-specific mechanisms of synapse regulation by WWC family proteins. Cell Rep 2024; 43:114841. [PMID: 39388350 PMCID: PMC11913214 DOI: 10.1016/j.celrep.2024.114841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 07/22/2024] [Accepted: 09/21/2024] [Indexed: 10/12/2024] Open
Abstract
The WW and C2 domain-containing protein (WWC2) is implicated in several neurological disorders. Here, we demonstrate that WWC2 interacts with inhibitory, but not excitatory, postsynaptic scaffolds, consistent with prior proteomic identification of WWC2 as a putative component of the inhibitory postsynaptic density. Using mice lacking WWC2 expression in excitatory forebrain neurons, we show that WWC2 suppresses γ-aminobutyric acid type-A receptor (GABAAR) incorporation into the plasma membrane and regulates HAP1 and GRIP1, which form a complex promoting GABAAR recycling to the membrane. Inhibitory synaptic transmission is increased in CA1 pyramidal cells lacking WWC2. Furthermore, unlike the WWC2 homolog KIBRA (kidney/brain protein; WWC1), a key regulator of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking at excitatory synapses, the deletion of WWC2 does not affect synaptic AMPAR expression. In contrast, loss of KIBRA does not affect GABAAR membrane expression. These data reveal synapse class-selective functions for WWC proteins as regulators of ionotropic neurotransmitter receptors and provide insight into mechanisms regulating GABAAR membrane expression.
Collapse
Affiliation(s)
- Thomas L Dunham
- Neuroscience Graduate Program, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Julia R Wilkerson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Richard C Johnson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard L Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lenora J Volk
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Psychiatry UT Southwestern Medical Center, Dallas, TX 75390, USA; Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
11
|
Romeo D, Ramirez-Calero S, Ravasi T, Rodolfo-Metalpa R, Schunter C. Neural mechanisms of mutualistic fish cleaning behaviour: a study in the wild. Biol Lett 2024; 20:20240339. [PMID: 39406338 PMCID: PMC11479757 DOI: 10.1098/rsbl.2024.0339] [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: 06/15/2024] [Revised: 08/09/2024] [Accepted: 09/09/2024] [Indexed: 10/20/2024] Open
Abstract
One crucial interaction for the health of fish communities in coral reefs is performed by cleaner fish by removing ectoparasites from the body of other fish, so-called clients. Studying the underlying mechanisms of this behaviour is essential to understanding how species react to social stimuli and defining the drivers of mutualistic social behaviour. Here, we pinpoint the neural molecular mechanisms in the cleaning behaviour of Labroides dimidiatus in the wild through an in situ interaction experiment at a coral reef in New Caledonia. Five cleaners and clients (Abudefduf saxatilis) were placed into underwater aquaria to interact, while five were not presented with a client. The brain transcriptomes revealed 233 differentially expressed genes in cleaners that were interacting with a client. Among these genes, grin2d, npy, slc6a3 and immediate early genes (IEGs; fosb and fosl1) were related to learning and memory, glutamate and dopamine pathways, which confirm molecular pathways observed in laboratory studies. However, a new potential mechanism was found with npy (neuropeptide Y) as a driver of feeding behaviour. These results show the role of neurotransmitters and IEGs in mutualistic social behaviour, unveiling the mechanism behind the feeding stimulus that leads the cleaner fish to establish mutualistic interactions in coral reefs.
Collapse
Affiliation(s)
- Daniele Romeo
- Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People’s Republic of China
| | - Sandra Ramirez-Calero
- Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People’s Republic of China
- Departament de Biologia Marina, Institut de Ciències del Mar (CSIC), Barcelona, Spain
- Departament de Biologia Evolutiva, Ecologia i Ciències Ambientals, Universitat de Barcelona (UB), Av. Diagonal 645, Barcelona08028, Spain
| | - Timothy Ravasi
- Marine Climate Change Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Japan
| | | | - Celia Schunter
- Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, People’s Republic of China
- State Key Laboratory of Marine Pollution, City University of Hong Kong, Hong Kong SAR, People’s Republic of China
| |
Collapse
|
12
|
Zimmerman AJ, Serrano-Rodriguez A, Wilson SJ, Linsenbardt DN, Brigman JL, Weick J. Knockout of AMPA receptor binding protein Neuron-Specific Gene 2 (NSG2) enhances associative learning and cognitive flexibility. RESEARCH SQUARE 2024:rs.3.rs-4790348. [PMID: 39257983 PMCID: PMC11384823 DOI: 10.21203/rs.3.rs-4790348/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The vast majority of gene mutations and/or gene knockouts result in either no observable changes, or significant deficits in molecular, cellular, or organismal function. However, in a small number of cases, mutant animal models display enhancements in specific behaviors such as learning and memory. To date, most gene deletions shown to enhance cognitive ability generally affect a limited number of pathways such as NMDA receptor- and translation-dependent plasticity, or GABA receptor- and potassium channel-mediated inhibition. While endolysosomal trafficking of AMPA receptors is a critical mediator of synaptic plasticity, mutations in genes that affect AMPAR trafficking either have no effect or are deleterious for synaptic plasticity, learning and memory. NSG2 is one of the three-member family of Neuron-specific genes (NSG1-3), which have been shown to regulate endolysosomal trafficking of a number of proteins critical for neuronal function, including AMPAR subunits (GluA1-2). Based on these findings and the largely universal expression throughout mammalian brain, we predicted that genetic knockout of NSG2 would result in significant impairments across multiple behavioral modalities including motor, affective, and learning/memory paradigms. However, in the current study we show that loss of NSG2 had highly selective effects on associative learning and memory, leaving motor and affective behaviors intact. For instance, NSG2 KO animals performed equivalent to wild-type C57Bl/6n mice on rotarod and Catwalk motor tasks, and did not display alterations in anxiety-like behavior on open field and elevated zero maze tasks. However, NSG2 KO animals demonstrated enhanced recall in the Morris water maze, accelerated reversal learning in a touch-screen task, and accelerated acquisition and enhanced recall on a Trace Fear Conditioning task. Together, these data point to a specific involvement of NSG2 on multiple types of associative learning, and expand the repertoire of pathways that can be targeted for cognitive enhancement.
Collapse
|
13
|
Lauby SC, Lapp HE, Salazar M, Semyrenko S, Chauhan D, Margolis AE, Champagne FA. Postnatal maternal care moderates the effects of prenatal bisphenol exposure on offspring neurodevelopmental, behavioral, and transcriptomic outcomes. PLoS One 2024; 19:e0305256. [PMID: 38861567 PMCID: PMC11166292 DOI: 10.1371/journal.pone.0305256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 05/28/2024] [Indexed: 06/13/2024] Open
Abstract
Bisphenols (BP), including BPA and "BPA-free" structural analogs, are commonly used plasticizers that are present in many plastics and are known endocrine disrupting chemicals. Prenatal exposure to BPA has been associated with negative neurodevelopmental and behavioral outcomes in children and in rodent models. Prenatal BPA exposure has also been shown to impair postnatal maternal care provisioning, which can also affect offspring neurodevelopment and behavior. However, there is limited knowledge regarding the biological effects of prenatal exposure to bisphenols other than BPA and the interplay between prenatal bisphenol exposure and postnatal maternal care on adult behavior. The purpose of the current study was to determine the interactive impact of prenatal bisphenol exposure and postnatal maternal care on neurodevelopment and behavior in rats. Our findings suggest that the effects of prenatal bisphenol exposure on eye-opening, adult attentional set shifting and anxiety-like behavior in the open field are dependent on maternal care in the first five days of life. Interestingly, maternal care might also attenuate the effects of prenatal bisphenol exposure on eye opening and adult attentional set shifting. Finally, transcriptomic profiles in male and female medial prefrontal cortex and amygdala suggest that the interactive effects of prenatal bisphenol exposure and postnatal maternal care converge on estrogen receptor signaling and are involved in biological processes related to gene expression and protein translation and synthesis. Overall, these findings indicate that postnatal maternal care plays a critical role in the expression of the effects of prenatal bisphenol exposure on neurodevelopment and adult behavior. Understanding the underlying biological mechanisms involved might allow us to identify potential avenues to mitigate the adverse effects of prenatal bisphenol exposure and improve health and well-being in human populations.
Collapse
Affiliation(s)
- Samantha C. Lauby
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
- Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin, Austin, Texas, United States of America
| | - Hannah E. Lapp
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
| | - Melissa Salazar
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
| | - Sofiia Semyrenko
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
| | - Danyal Chauhan
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
| | - Amy E. Margolis
- Department of Psychiatry, Columbia University Irving Medical Center, New York City, New York, United States of America
| | - Frances A. Champagne
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, Austin, Texas, United States of America
- Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin, Austin, Texas, United States of America
| |
Collapse
|
14
|
Szabó D, Franke V, Bianco S, Batiuk MY, Paul EJ, Kukalev A, Pfisterer UG, Irastorza-Azcarate I, Chiariello AM, Demharter S, Zea-Redondo L, Lopez-Atalaya JP, Nicodemi M, Akalin A, Khodosevich K, Ungless MA, Winick-Ng W, Pombo A. A single dose of cocaine rewires the 3D genome structure of midbrain dopamine neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593308. [PMID: 38766140 PMCID: PMC11100777 DOI: 10.1101/2024.05.10.593308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Midbrain dopamine neurons (DNs) respond to a first exposure to addictive drugs and play key roles in chronic drug usage1-3. As the synaptic and transcriptional changes that follow an acute cocaine exposure are mostly resolved within a few days4,5, the molecular changes that encode the long-term cellular memory of the exposure within DNs remain unknown. To investigate whether a single cocaine exposure induces long-term changes in the 3D genome structure of DNs, we applied Genome Architecture Mapping and single nucleus transcriptomic analyses in the mouse midbrain. We found extensive rewiring of 3D genome architecture at 24 hours past exposure which remains or worsens by 14 days, outlasting transcriptional responses. The cocaine-induced chromatin rewiring occurs at all genomic scales and affects genes with major roles in cocaine-induced synaptic changes. A single cocaine exposure triggers extensive long-lasting changes in chromatin condensation in post-synaptic and post-transcriptional regulatory genes, for example the unfolding of Rbfox1 which becomes most prominent 14 days post exposure. Finally, structurally remodeled genes are most expressed in a specific DN sub-type characterized by low expression of the dopamine auto-receptor Drd2, a key feature of highly cocaine-sensitive cells. These results reveal an important role for long-lasting 3D genome remodelling in the cellular memory of a single cocaine exposure, providing new hypotheses for understanding the inception of drug addiction and 3D genome plasticity.
Collapse
Affiliation(s)
- Dominik Szabó
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
- Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Vedran Franke
- Bioinformatics & Omics Data Science platform, Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, 10115 Berlin, Germany
| | - Simona Bianco
- Dipartimento di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant’Angelo, 80126 Naples, Italy
| | - Mykhailo Y. Batiuk
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Eleanor J. Paul
- MRC London Institute of Medical Sciences (LMS), London W12 0HS, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Alexander Kukalev
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
| | - Ulrich G. Pfisterer
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Ibai Irastorza-Azcarate
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
| | - Andrea M. Chiariello
- Dipartimento di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant’Angelo, 80126 Naples, Italy
| | - Samuel Demharter
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Luna Zea-Redondo
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
- Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Jose P. Lopez-Atalaya
- Instituto de Neurociencias, Universidad Miguel Hernández-Consejo Superior de Investigaciones Científicas (UMH-CSIC), 03550, Sant Joan d’Alacant, Spain
| | - Mario Nicodemi
- Dipartimento di Fisica, Università di Napoli Federico II, and INFN Napoli, Complesso Universitario di Monte Sant’Angelo, 80126 Naples, Italy
- Berlin Institute of Health, 10178 Berlin, Germany
| | - Altuna Akalin
- Bioinformatics & Omics Data Science platform, Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, 10115 Berlin, Germany
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Mark A. Ungless
- MRC London Institute of Medical Sciences (LMS), London W12 0HS, UK
- Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London SW7 2AZ, UK
| | - Warren Winick-Ng
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
- Donnelly Centre, University of Toronto, Toronto, Ontario M5S 3E1, Toronto, Canada
| | - Ana Pombo
- Max-Delbrück Centre for Molecular Medicine, Berlin Institute for Medical Systems Biology, Epigenetic Regulation and Chromatin Architecture Group, 10115 Berlin, Germany
- Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| |
Collapse
|
15
|
Bandara D, Riccardi K. Graph Node Classification to Predict Autism Risk in Genes. Genes (Basel) 2024; 15:447. [PMID: 38674382 PMCID: PMC11049455 DOI: 10.3390/genes15040447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
This study explores the genetic risk associations with autism spectrum disorder (ASD) using graph neural networks (GNNs), leveraging the Sfari dataset and protein interaction network (PIN) data. We built a gene network with genes as nodes, chromosome band location as node features, and gene interactions as edges. Graph models were employed to classify the autism risk associated with newly introduced genes (test set). Three classification tasks were undertaken to test the ability of our models: binary risk association, multi-class risk association, and syndromic gene association. We tested graph convolutional networks, Graph Sage, graph transformer, and Multi-Layer Perceptron (Baseline) architectures on this problem. The Graph Sage model consistently outperformed the other models, showcasing its utility in classifying ASD-related genes. Our ablation studies show that the chromosome band location and protein interactions contain useful information for this problem. The models achieved 85.80% accuracy on the binary risk classification, 81.68% accuracy on the multi-class risk classification, and 90.22% on the syndromic classification.
Collapse
Affiliation(s)
- Danushka Bandara
- Department of Computer Science and Engineering, Fairfield University, Fairfield, CT 06824, USA;
| | | |
Collapse
|
16
|
Dunham TL, Wilkerson JR, Johnson RC, Huganir RL, Volk LJ. Modulation of GABA A receptor trafficking by WWC2 reveals class-specific mechanisms of synapse regulation by WWC family proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584487. [PMID: 38559047 PMCID: PMC10979870 DOI: 10.1101/2024.03.11.584487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
WWC2 (WW and C2 domain-containing protein) is implicated in several neurological disorders, however its function in the brain has yet to be determined. Here, we demonstrate that WWC2 interacts with inhibitory but not excitatory postsynaptic scaffolds, consistent with prior proteomic identification of WWC2 as a putative component of the inhibitory postsynaptic density. Using mice lacking WWC2 expression in excitatory forebrain neurons, we show that WWC2 suppresses GABA A R incorporation into the plasma membrane and regulates HAP1 and GRIP1, which form a complex promoting GABA A R recycling to the membrane. Inhibitory synaptic transmission is dysregulated in CA1 pyramidal cells lacking WWC2. Furthermore, unlike the WWC2 homolog KIBRA (WWC1), a key regulator of AMPA receptor trafficking at excitatory synapses, deletion of WWC2 does not affect synaptic AMPAR expression. In contrast, loss of KIBRA does not affect GABA A R membrane expression. These data reveal unique, synapse class-selective functions for WWC proteins as regulators of ionotropic neurotransmitter receptors and provide insight into mechanisms regulating GABA A R membrane expression.
Collapse
|
17
|
Lu P, Gao CX, Luo FJ, Huang YT, Gao MM, Long YS. Hippocampal proteomic changes in high-fat diet-induced obese mice associated with memory decline. J Nutr Biochem 2024; 125:109554. [PMID: 38142716 DOI: 10.1016/j.jnutbio.2023.109554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/24/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Substantial evidence suggest that chronic consumption of high-fat diets (HFDs) can lead to obesity, abnormal metabolism, as well as cognitive impairment. Molecular and cellular changes regarding hippocampal dysfunctions have been identified in multiple HFD animal models. Therefore, in-depth identification of expression changes of hippocampal proteins is critical for understanding the mechanism of HFD-induced cognitive deficits. In this study, we fed 3-week-old male mice with HFD for 3 months to generate obese mice who exhibit systemic metabolic abnormality and learning and memory decline. Using an iTRAQ-labeled proteomic analysis, we identified a total of 82 differentially expressed proteins (DEPs) in the hippocampus upon HFD with 35 up-regulated proteins and 47 down-regulated proteins. Functional enrichment indicated that these DEPs were predominantly enriched in regulation of catabolic process, dendritic shaft, neuron projection morphogenesis and GTPase regulator activity. Protein-protein interaction enrichment showed that the DEPs are mostly enriched in postsynaptic functions; and of them, six proteins (i.e., DLG3, SYNGAP1, DCLK1, GRIA4, GRIP1, and ARHGAP32) were involved in several functional assemblies of the postsynaptic density including G-protein signaling, scaffolding and adaptor, kinase and AMPA signaling, respectively. Collectively, our findings suggest that these DEPs upon HFD might contribute to memory decline by disturbing neuronal and postsynaptic functions in the hippocampus.
Collapse
Affiliation(s)
- Ping Lu
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Cun-Xiu Gao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Fei-Jian Luo
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yu-Ting Huang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Mei-Mei Gao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yue-Sheng Long
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
18
|
Bahbahani H, Alfoudari A, Al-Ateeqi A, Al Abri M, Almathen F. Positive selection footprints and haplotype distribution in the genome of dromedary camels. Animal 2024; 18:101098. [PMID: 38377812 DOI: 10.1016/j.animal.2024.101098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/22/2024] Open
Abstract
Dromedary camels are a domestic species characterized by various adaptive traits. Limited efforts have been employed toward identifying genetic regions and haplotypes under selection that might be related to such adaptations. These genetic elements are considered valuable sources that should be conserved to maintain the dromedaries' adaptability. Here, we have analyzed whole genome sequences of 40 dromedary camels from different Arabian Peninsula populations to assess their genetic relationship and define regions with signatures of selection. Genetic distinction based on geography was observed, classifying the populations into four groups: (1) North and Central, (2) West, (3) Southwest, and (4) Southeast, with substantial levels of genetic admixture. Using the de-correlated composite of multiple signal approach, which combines four intra-population analyses (Tajima's D index, nucleotide diversity, integrated haplotype score, and number of segregating sites by length), a total of 36 candidate regions harboring 87 genes were identified to be under positive selection. These regions overlapped with 185 haplotype blocks encompassing 1 340 haplotypes, of which 30 (∼2%) were found to be approaching fixation. The defined candidate genes are associated with different biological processes related to the dromedaries' adaptive physiologies, including neurological pathways, musculoskeletal development, fertility, fat distribution, immunity, visual development, and kidney physiology. The results of this study highlight opportunities for further investigations at the whole-genome level to enhance our understanding of the evolutionary pressures shaping the dromedary genome.
Collapse
Affiliation(s)
- H Bahbahani
- Department of Biological Sciences, Faculty of Science, Kuwait University, Sh. Sabah Al-Salem campus, Kuwait.
| | - A Alfoudari
- Department of Biological Sciences, Faculty of Science, Kuwait University, Sh. Sabah Al-Salem campus, Kuwait
| | - A Al-Ateeqi
- Environment and Life Sciences Research Center, Kuwait Institute for Scientific Research, Kuwait City, Kuwait
| | - M Al Abri
- Department of Animal and Veterinary Sciences, Sultan Qaboos University, Muscat, Oman
| | - F Almathen
- Department of Public Health, King Faisal University, 400 Al-Ahsa, Kingdom of Saudi Arabia; Camel Research Center, King Faisal University, 400 Al-Ahsa, Saudi Arabia
| |
Collapse
|
19
|
Araki Y, Rajkovich KE, Gerber EE, Gamache TR, Johnson RC, Tran THN, Liu B, Zhu Q, Hong I, Kirkwood A, Huganir R. SynGAP regulates synaptic plasticity and cognition independently of its catalytic activity. Science 2024; 383:eadk1291. [PMID: 38422154 PMCID: PMC11188940 DOI: 10.1126/science.adk1291] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/28/2023] [Indexed: 03/02/2024]
Abstract
SynGAP is an abundant synaptic GTPase-activating protein (GAP) critical for synaptic plasticity, learning, memory, and cognition. Mutations in SYNGAP1 in humans result in intellectual disability, autistic-like behaviors, and epilepsy. Heterozygous Syngap1-knockout mice display deficits in synaptic plasticity, learning, and memory and exhibit seizures. It is unclear whether SynGAP imparts structural properties at synapses independently of its GAP activity. Here, we report that inactivating mutations within the GAP domain do not inhibit synaptic plasticity or cause behavioral deficits. Instead, SynGAP modulates synaptic strength by physically competing with the AMPA-receptor-TARP excitatory receptor complex in the formation of molecular condensates with synaptic scaffolding proteins. These results have major implications for developing therapeutic treatments for SYNGAP1-related neurodevelopmental disorders.
Collapse
Affiliation(s)
| | | | | | | | - Richard C. Johnson
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thanh Hai N. Tran
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bian Liu
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Qianwen Zhu
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ingie Hong
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Alfredo Kirkwood
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard Huganir
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
20
|
Zhou P, Wu Z, Zhang Q, Wang L, Zhang W, Han X. A novel link between circPDE3B and ferroptosis in esophageal squamous cell carcinoma progression. Genomics 2024; 116:110761. [PMID: 38092323 DOI: 10.1016/j.ygeno.2023.110761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/30/2023] [Accepted: 12/10/2023] [Indexed: 12/23/2023]
Abstract
AIM To unravel whether ferroptosis involves with the actions by circPDE3B-mediated facilitation of esophageal squamous cell carcinoma (ESCC) progression. METHODS Human ESCC tissues and cell lines were prepared for the evaluation of ferroptosis. Cellular iron, ROS, GSH, and MDA levels were measured to assess ferroptosis. Flow cytometry was employed to analyze apoptosis and cell cycle. Subcellular fractionation and fluorescence in situ hybridization (FISH) were conducted to validate the localization of circPDE3B. RNA pull-down, RNA immunoprecipitation (RIP), and luciferase assay were subjected to identify the molecular mechanisms. Nude mouse xenograft model was carried out to evaluate the function of circPDE3B/SLC7A11/CBS in vivo. RESULTS Increased circPDE3B in human ESCC specimens was positively correlated with ferroptosis-related molecules, SLC7A11 and CBS. Functionally, circPDE3B knockdown triggered ferroptosis, apoptosis, and cell cycle arrest in ESCC cells. Whereas, these effects were obviously blocked by miR-516b-5p inhibitor. Mechanistically, not only circPDE3B sponged miR-516b-5p to upregulate CBS, but also directly bound with HNRNPK to stabilize SLC7A11. In mice, depletion of circPDE3B restrained ESCC growth, while this was abolished by overexpression of CBS or SLC7A11. CONCLUSION In summary, circPDE3B promotes ESCC progression by suppressing ferroptosis through recruiting HNRNPK/SLC7A11 and miR-516b-5p/CBS axes.
Collapse
Affiliation(s)
- Pengli Zhou
- Intervention Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Zhengyang Wu
- Intervention Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Qinghui Zhang
- Intervention Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Ling Wang
- Intervention Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Wenguang Zhang
- Intervention Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China
| | - Xinwei Han
- Intervention Department, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, PR China.
| |
Collapse
|
21
|
Cao YY, Wu LL, Li XN, Yuan YL, Zhao WW, Qi JX, Zhao XY, Ward N, Wang J. Molecular Mechanisms of AMPA Receptor Trafficking in the Nervous System. Int J Mol Sci 2023; 25:111. [PMID: 38203282 PMCID: PMC10779435 DOI: 10.3390/ijms25010111] [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: 11/24/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Synaptic plasticity enhances or reduces connections between neurons, affecting learning and memory. Postsynaptic AMPARs mediate greater than 90% of the rapid excitatory synaptic transmission in glutamatergic neurons. The number and subunit composition of AMPARs are fundamental to synaptic plasticity and the formation of entire neural networks. Accordingly, the insertion and functionalization of AMPARs at the postsynaptic membrane have become a core issue related to neural circuit formation and information processing in the central nervous system. In this review, we summarize current knowledge regarding the related mechanisms of AMPAR expression and trafficking. The proteins related to AMPAR trafficking are discussed in detail, including vesicle-related proteins, cytoskeletal proteins, synaptic proteins, and protein kinases. Furthermore, significant emphasis was placed on the pivotal role of the actin cytoskeleton, which spans throughout the entire transport process in AMPAR transport, indicating that the actin cytoskeleton may serve as a fundamental basis for AMPAR trafficking. Additionally, we summarize the proteases involved in AMPAR post-translational modifications. Moreover, we provide an overview of AMPAR transport and localization to the postsynaptic membrane. Understanding the assembly, trafficking, and dynamic synaptic expression mechanisms of AMPAR may provide valuable insights into the cognitive decline associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Yang Cao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Ling-Ling Wu
- School of Medicine, Shanghai University, Shanghai 200444, China;
| | - Xiao-Nan Li
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Yu-Lian Yuan
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Wan-Wei Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Jing-Xuan Qi
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Xu-Yu Zhao
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| | - Natalie Ward
- Medical Laboratory, Exceptional Community Hospital, 19060 N John Wayne Pkwy, Maricopa, AZ 85139, USA;
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.-Y.C.); (X.-N.L.); (Y.-L.Y.); (W.-W.Z.); (J.-X.Q.); (X.-Y.Z.)
| |
Collapse
|
22
|
Xie Y, Zhao J, Li X, Sun J, Yang H. Effects of Cyfluthrin Exposure on Neurobehaviour, Hippocampal Tissue and Synaptic Plasticity in Wistar Rats. TOXICS 2023; 11:999. [PMID: 38133400 PMCID: PMC10748044 DOI: 10.3390/toxics11120999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
This experiment was conducted to study the effects of Cyfluthrin (Cy) exposure on neurobehaviour, hippocampal tissue and synaptic plasticity in Wistar rats. First, it was found that high-dose Cy exposure could cause nerve injury, resulting in symptoms such as deficits in learning and memory ability, spatial exploration and autonomic motor function. Moreover, it was found that medium- and high-dose Cy exposure could cause an abnormal release of the neurotransmitter Glu. Second, brain tissue pathology showed that the middle and high doses of Cy caused tissue deformation, reduced the number of hippocampal puramidal cells, caused a disorder of these cells, decreased the number of Nissl bodies, and caused pyknosis of the hippocampal cell nuclear membrane and serious damage to organelles, indicating that exposure to these doses of Cy may cause hippocampal tissue damage in rats. Third, as the exposure dose increased, morphological changes in hippocampal synapses, including blurred synaptic spaces, a decreased number of synaptic vesicles and a decreased number of synapses, became more obvious. Moreover, the expression levels of the key synaptic proteins PSD-95 and SYP also decreased in a dose-dependent manner, indicating obvious synaptic damage. Finally, the study found that medium and high doses of Cy could upregulate the expression of A2AR in the hippocampus and that the expression levels of inflammatory factors and apoptosis-related proteins increased in a dose-dependent manner. Moreover, the expression of A2AR mRNA was correlated with neurobehavioural indicators and the levels of inflammatory factors, synaptic plasticity-related factors and apoptosis-related factors, suggesting that Cy may cause nerve damage in rats and that this effect is closely related to A2AR.
Collapse
Affiliation(s)
- Yongxin Xie
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China; (Y.X.); (J.Z.); (X.L.)
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750004, China
| | - Ji Zhao
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China; (Y.X.); (J.Z.); (X.L.)
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750004, China
| | - Xiaoyu Li
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China; (Y.X.); (J.Z.); (X.L.)
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750004, China
| | - Jian Sun
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China; (Y.X.); (J.Z.); (X.L.)
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750004, China
| | - Huifang Yang
- School of Public Health, Ningxia Medical University, Yinchuan 750004, China; (Y.X.); (J.Z.); (X.L.)
- Key Laboratory of Environmental Factors and Chronic Disease Control, No. 1160, Shengli Street, Xingqing District, Yinchuan 750004, China
| |
Collapse
|
23
|
Plitt MH, Kaganovsky K, Südhof TC, Giocomo LM. Hippocampal place code plasticity in CA1 requires postsynaptic membrane fusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567978. [PMID: 38045362 PMCID: PMC10690209 DOI: 10.1101/2023.11.20.567978] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Rapid delivery of glutamate receptors to the postsynaptic membrane via vesicle fusion is a central component of synaptic plasticity. However, it is unknown how this process supports specific neural computations during behavior. To bridge this gap, we combined conditional genetic deletion of a component of the postsynaptic membrane fusion machinery, Syntaxin3 (Stx3), in hippocampal CA1 neurons of mice with population in vivo calcium imaging. This approach revealed that Stx3 is necessary for forming the neural dynamics that support novelty processing, spatial reward memory and offline memory consolidation. In contrast, CA1 Stx3 was dispensable for maintaining aspects of the neural code that exist presynaptic to CA1 such as representations of context and space. Thus, manipulating postsynaptic membrane fusion identified computations that specifically require synaptic restructuring via membrane trafficking in CA1 and distinguished them from neural representation that could be inherited from upstream brain regions or learned through other mechanisms.
Collapse
Affiliation(s)
- Mark H. Plitt
- Department of Neurobiology, Stanford University School of Medicine; Stanford, CA, USA
- These authors contributed equally to this work
- Present address: Department of Molecular and Cell Biology, University of California Berkeley; Berkeley, CA, USA
| | - Konstantin Kaganovsky
- Department of Neurosurgery, Stanford University School of Medicine; Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine; Stanford, CA, USA
- These authors contributed equally to this work
- Present address: Department of Psychiatry and Behavioral Sciences, Center for Sleep Sciences and Medicine, Stanford University School of Medicine; Stanford, CA, USA
| | - Thomas C. Südhof
- Department of Neurosurgery, Stanford University School of Medicine; Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine; Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine; Stanford, CA, USA
| | - Lisa M. Giocomo
- Department of Neurobiology, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
24
|
Lauby SC, Lapp HE, Salazar M, Semyrenko S, Chauhan D, Margolis AE, Champagne FA. Postnatal maternal care moderates the effects of prenatal bisphenol exposure on offspring neurodevelopmental, behavioral, and transcriptomic outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.19.558481. [PMID: 37786706 PMCID: PMC10541647 DOI: 10.1101/2023.09.19.558481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Bisphenols (BPs), including BPA and "BPA-free" structural analogs, are commonly used plasticizers that are present in many plastics and are known endocrine disrupting chemicals. Prenatal exposure to BPA has been associated with negative neurodevelopmental and behavioral outcomes in children and rodent models. Prenatal BPA exposure has also been shown to impair postnatal maternal care provisioning, which can also affect offspring neurodevelopment and behavior. However, there is limited knowledge regarding the biological effects of prenatal exposure to bisphenols other than BPA and the interplay between prenatal BP exposure and postnatal maternal care on adult behavior. The purpose of the current study was to determine the interactive impact of prenatal BP exposure and postnatal maternal care on neurodevelopment and behavior. Our findings suggest that the effects of prenatal BP exposure on eye-opening, adult attentional set shifting and anxiety-like behavior in the open field are dependent on maternal care in the first five days of life. Interestingly, maternal care might also attenuate the effects of prenatal BP exposure on eye opening and adult attentional set shifting. Finally, transcriptomic profiles in male and female medial prefrontal cortex and amygdala suggest that the interactive effects of prenatal BP exposure and postnatal maternal care converge on estrogen receptor signaling and are involved in biological processes related to gene expression and protein translation and synthesis. Overall, these findings indicate that postnatal maternal care plays a critical role in the expression of the effects of prenatal BP exposure on neurodevelopment and adult behavior. Understanding the underlying biological mechanisms involved might allow us to identify potential avenues to mitigate the adverse effects of prenatal BP exposure and improve health and well-being in human populations.
Collapse
Affiliation(s)
- Samantha C Lauby
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
- Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin
| | - Hannah E Lapp
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
| | - Melissa Salazar
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
| | - Sofiia Semyrenko
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
| | - Danyal Chauhan
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
| | - Amy E Margolis
- Department of Psychiatry, Columbia University Irving Medical Center
| | - Frances A Champagne
- Department of Psychology, College of Liberal Arts, University of Texas at Austin
- Center for Molecular Carcinogenesis and Toxicology, University of Texas at Austin
| |
Collapse
|
25
|
Dong W, Weng JF, Zhu JB, Zheng YF, Liu LL, Dong C, Ruan Y, Fang X, Chen J, Liu WY, Peng XP, Chen XY. CREB-binding protein and HIF-1α/β-catenin to upregulate miR-322 and alleviate myocardial ischemia-reperfusion injury. FASEB J 2023; 37:e22996. [PMID: 37566526 DOI: 10.1096/fj.202200596rrrrrr] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 08/13/2023]
Abstract
Myocardial ischemia/reperfusion injury (MIRI) is a prevalent condition associated with numerous critical clinical conditions. miR-322 has been implicated in MIRI through poorly understood mechanisms. Our preliminary analysis indicated potential interaction of CREB-binding protein (CBP), a transcriptional coactivator and acetyltransferase, with HIF-1α/β-catenin, which might regulate miR-322 expression. We, therefore, hypothesized that CBP/HIF-1α/β-catenin/miR-322 axis might play a role in MIRI. Rat cardiomyocytes subjected to oxygen-glucose deprivation /reperfusion (OGD/R) and Langendorff perfused heart model were used to model MIRI in vitro and in vivo, respectively. We used various techniques such as CCK-8 assay, transferase dUTP nick end labeling staining, western blotting, RT-qPCR, chromatin immunoprecipitation (ChIP), dual-luciferase assay, co-immunoprecipitation (Co-IP), hematoxylin and eosin staining, and TTC staining to assess cell viability, apoptosis, and the levels of CBP, HIF-1α, β-catenin, miR-322, and acetylation. Our results indicate that OGD/R in cardiomyocytes decreased CBP/HIF-1α/β-catenin/miR-322 expression, increased cell apoptosis and cytokines, and reduced cell viability. However, overexpression of CBP or miR-322 suppressed OGD/R-induced cell injury, while knockdown of HIF-1α/β-catenin further exacerbated the damage. HIF-1α/β-catenin bound to miR-322 promoter to promote its expression, while CBP acetylated HIF-1α/β-catenin for stabilization. Overexpression of CBP attenuated MIRI in rats by acetylating HIF-1α/β-catenin to stabilize their expression, resulting in stronger binding of HIF-1α/β-catenin with the miR-322 promoter and subsequent increased miR-322 levels. Therefore, activating CBP/HIF-1α/β-catenin/miR-322 signaling may be a potential approach to treat MIRI.
Collapse
Affiliation(s)
- Wei Dong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jun-Fei Weng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jian-Bing Zhu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Yao-Fu Zheng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Lei-Lei Liu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Chen Dong
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Yang Ruan
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Xu Fang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jin Chen
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Wen-Yu Liu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Xiao-Ping Peng
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Xuan-Ying Chen
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
26
|
Tan LM, Chen P, Nie ZY, Liu XF, Wang B. Circular RNA XRCC5 aggravates glioma progression by activating CLC3/SGK1 axis via recruiting IGF2BP2. Neurochem Int 2023; 166:105534. [PMID: 37061192 DOI: 10.1016/j.neuint.2023.105534] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND Increasing evidences have reported the critical roles of circular RNA (circRNA) in gliomas. Whereas, the role of circXRCC5 in glioma and its underlying molecular mechanism has not been reported. METHODS The RNA transcripts and protein levels were detected using qRT-PCR, immunohistochemistry (IHC) and in situ hybridization (ISH) assays. Cell proliferation was characterized by CCK-8 and clone formation assays. The formation of NLRP3-inflammasomes was identified using immunofluorescence (IF) and Western blot assays. The cytokines were determined using immunosorbent assay (ELISA) and Western blot assays. The molecular interactions were validated using RIP and pull-down assays. RESULTS circXRCC5 was over-expressed in glioma and positively related to the shorter survival rate, advanced TNM stage and larger tumor volume. circXRCC5 knockdown inhibited cell proliferation and NLRP3-mediated inflammasome activation of glioma cells. Subsequently, we found that circXRCC5 maintained mRNA stability of CLC3 by binding to IGF2BP2. Furthermore, CLC3 accelerated SGK1 expression via PI3K/PDK1/AKT pathway. The rescue experiments showed that both overexpression of CLC3 or SGK1 dramatically alleviated circXRCC5 knockdown-induced inhibition of cell proliferation and NLRP3-mediated inflammasome activation of glioma cells. In vivo, our study proved that circXRCC5 accelerated glioma growth by regulating CLC3/SGK1 axis. CONCLUSION Our data concluded that circXRCC5 formed a complex with IGF2BP2 to regulate inflammasome activation and tumor growth via CLC3/SGK1 axis.
Collapse
Affiliation(s)
- Li-Ming Tan
- Department of Neurosurgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| | - Ping Chen
- Department of Neurosurgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| | - Zhen-Yu Nie
- Department of Neurosurgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| | - Xiao-Fei Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| | - Bing Wang
- Department of Neurosurgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China.
| |
Collapse
|
27
|
Zhang YY, Li XS, Ren KD, Peng J, Luo XJ. Restoration of metal homeostasis: a potential strategy against neurodegenerative diseases. Ageing Res Rev 2023; 87:101931. [PMID: 37031723 DOI: 10.1016/j.arr.2023.101931] [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: 01/31/2023] [Revised: 04/04/2023] [Accepted: 04/06/2023] [Indexed: 04/11/2023]
Abstract
Metal homeostasis is critical to normal neurophysiological activity. Metal ions are involved in the development, metabolism, redox and neurotransmitter transmission of the central nervous system (CNS). Thus, disturbance of homeostasis (such as metal deficiency or excess) can result in serious consequences, including neurooxidative stress, excitotoxicity, neuroinflammation, and nerve cell death. The uptake, transport and metabolism of metal ions are highly regulated by ion channels. There is growing evidence that metal ion disorders and/or the dysfunction of ion channels contribute to the progression of neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). Therefore, metal homeostasis-related signaling pathways are emerging as promising therapeutic targets for diverse neurological diseases. This review summarizes recent advances in the studies regarding the physiological and pathophysiological functions of metal ions and their channels, as well as their role in neurodegenerative diseases. In addition, currently available metal ion modulators and in vivo quantitative metal ion imaging methods are also discussed. Current work provides certain recommendations based on literatures and in-depth reflections to improve neurodegenerative diseases. Future studies should turn to crosstalk and interactions between different metal ions and their channels. Concomitant pharmacological interventions for two or more metal signaling pathways may offer clinical advantages in treating the neurodegenerative diseases.
Collapse
Affiliation(s)
- Yi-Yue Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Xi-Sheng Li
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013,China
| | - Kai-Di Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha 410013,China.
| |
Collapse
|
28
|
Li HY, Zhu MZ, Yuan XR, Guo ZX, Pan YD, Li YQ, Zhu XH. A thalamic-primary auditory cortex circuit mediates resilience to stress. Cell 2023; 186:1352-1368.e18. [PMID: 37001500 DOI: 10.1016/j.cell.2023.02.036] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 01/09/2023] [Accepted: 02/23/2023] [Indexed: 04/01/2023]
Abstract
Resilience enables mental elasticity in individuals when rebounding from adversity. In this study, we identified a microcircuit and relevant molecular adaptations that play a role in natural resilience. We found that activation of parvalbumin (PV) interneurons in the primary auditory cortex (A1) by thalamic inputs from the ipsilateral medial geniculate body (MG) is essential for resilience in mice exposed to chronic social defeat stress. Early attacks during chronic social defeat stress induced short-term hyperpolarizations of MG neurons projecting to the A1 (MGA1 neurons) in resilient mice. In addition, this temporal neural plasticity of MGA1 neurons initiated synaptogenesis onto thalamic PV neurons via presynaptic BDNF-TrkB signaling in subsequent stress responses. Moreover, optogenetic mimicking of the short-term hyperpolarization of MGA1 neurons, rather than merely activating MGA1 neurons, elicited innate resilience mechanisms in response to stress and achieved sustained antidepressant-like effects in multiple animal models, representing a new strategy for targeted neuromodulation.
Collapse
Affiliation(s)
- Huan-Yu Li
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Min-Zhen Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Xin-Rui Yuan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Zhi-Xin Guo
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Yi-Da Pan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| | - Yuan-Qing Li
- Research Center for Brain-Computer Interface, Pazhou Lab, Guangzhou 510330, China; School of Automation Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Xin-Hong Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China; School of Psychology, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
29
|
Zhang B, Wang ML, Huang SM, Cui Y, Li Y. Kaixin-San improves Aβ-induced synaptic plasticity inhibition by affecting the expression of regulation proteins associated with postsynaptic AMPAR expression. Front Pharmacol 2023; 14:1079400. [PMID: 36865910 PMCID: PMC9970989 DOI: 10.3389/fphar.2023.1079400] [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: 11/18/2022] [Accepted: 01/26/2023] [Indexed: 02/16/2023] Open
Abstract
Objective: To explore the mechanism underlying Kaixin-San (KXS) regulation of postsynaptic AMPA receptor (AMPAR) expression to mitigate toxic effects of the amyloid-β protein (Aβ). Methods: An animal model was established via intracerebroventricular injection of Aβ1-42. The Morris water maze test was conducted to evaluate learning and memory, while electrophysiological recording was conducted to assess the hippocampal long-term potentiation (LTP). Western blotting was used to detect expression levels of the hippocampal postsynaptic AMPAR and its accessory proteins. Results: The time spent to find the platform was significantly prolonged, the number of mice crossing the target site was significantly reduced, and the maintenance of LTP was inhibited in the Aβ group than in the control group. In the Aβ/KXS group, the time taken to find the platform was significantly shortened and the number of mice crossing the target site was significantly increased than in the Aβ group; furthermore, the inhibition of LTP induced by Aβ was reversed. The expression of GluR1, GluR2, ABP, GRIP1, NSF, and pGluR1-Ser845 was upregulated, while that of pGluR2-Ser880 and PKC δ was downregulated in the Aβ/KXS group. Conclusion: The increased expression of ABP, GRIP1, NSF, and pGluR1-Ser845 and the decreased expression of pGluR2-Ser880 and PKC δ under the influence of KXS, followed by the upregulation of postsynaptic GluR1 and GluR2, alleviated the inhibition of LTP induced by Aβ. Ultimately, the memory function of model animals was improved by KXS. Our study provides novel insights into the mechanism underlying KXS mitigation of Aβ-induced synaptic plasticity inhibition and memory impairment by altering the levels of accessory proteins associated with AMPAR expression.
Collapse
Affiliation(s)
- Bo Zhang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Meng-Lu Wang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shu-Ming Huang
- Institute of Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Cui
- Department of Veterinary Medicine, Institute of tropical agriculture and forestry, Hainan University, Haikou, China,*Correspondence: Yu Cui, ; Yan Li,
| | - Yan Li
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College of Yangzhou University, Yangzhou, China,*Correspondence: Yu Cui, ; Yan Li,
| |
Collapse
|
30
|
Brown JC, Higgins ES, George MS. Synaptic Plasticity 101: The Story of the AMPA Receptor for the Brain Stimulation Practitioner. Neuromodulation 2022; 25:1289-1298. [PMID: 35088731 PMCID: PMC10479373 DOI: 10.1016/j.neurom.2021.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/10/2021] [Accepted: 09/08/2021] [Indexed: 02/04/2023]
Abstract
The fields of Neurobiology and Neuromodulation have never been closer. Consequently, the phrase "synaptic plasticity" has become very familiar to non-basic scientists, without actually being very familiar. We present the "Story of the AMPA receptor," an easy-to-understand "10,000 ft" narrative overview of synaptic plasticity, oriented toward the brain stimulation clinician or scientist without basic science training. Neuromodulation is unparalleled in its capacity to both modulate and probe plasticity, yet many are not comfortable with their grasp of the topic. Here, we describe the seminal discoveries that defined the canonical mechanisms of long-term potentiation (LTP), long-term depression (LTD), and homeostatic plasticity. We then provide a conceptual framework for how plasticity at the synapse is accomplished, describing the functional roles of N-methyl-d-aspartate (NMDA) receptors and calcium, their effect on calmodulin, phosphatases (ie, calcineurin), kinases (ie, calcium/calmodulin-dependent protein kinase [CaMKII]), and structural "scaffolding" proteins (ie, post-synaptic density protein [PSD-95]). Ultimately, we describe how these affect the α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor. More specifically, AMPA receptor delivery to (LTP induction), removal from (LTD), or recycling within (LTP maintenance) the synapse is determined by the status of phosphorylation and protein binding at specific sites on the tails of AMPA receptor subunits: GluA1 and GluA2. Finally, we relate these to transcranial magnetic stimulation (TMS) treatment, highlighting evidences for LTP as the basis of high-frequency TMS therapy, and briefly touch on the role of plasticity for other brain stimulation modalities. In summary, we present Synaptic Plasticity 101 as a singular introductory reference for those less familiar with the mechanisms of synaptic plasticity.
Collapse
Affiliation(s)
- Joshua C Brown
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA; Department of Neurology, Medical University of South Carolina, Charleston, SC, USA; Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, USA.
| | - Edmund S Higgins
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Mark S George
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA; Ralph Johnson VA Medical Center, Charleston, SC, USA
| |
Collapse
|
31
|
Amoah DK. Advances in the understanding and enhancement of the human cognitive functions of learning and memory. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Learning and memory are among the key cognitive functions that drive the human experience. As such, any defective condition associated with these cognitive domains could affect our navigation through everyday life. For years, researchers have been working toward having a clear understanding of how learning and memory work, as well as ways to improve them. Many advances have been made, as well as some challenges that have also been faced in the process. That notwithstanding, there are prospects with regards to the frontier of the enhancement of learning and memory in humans. This review article selectively highlights four broad areas of focus in research into the understanding and enhancement of learning and memory. Brain stimulation, effects of sleep, effects of stress and emotion, and synaptic plasticity are the main focal areas of this review, in terms of some pivotal research works, findings and theories.
Collapse
Affiliation(s)
- Daniel Kofi Amoah
- Noguchi Memorial Institute for Medical Research, University of Ghana, Accra LG 25, Ghana
| |
Collapse
|
32
|
Okano Y, Kase Y, Okano H. A set-theoretic definition of cell types with an algebraic structure on gene regulatory networks and application in annotation of RNA-seq data. Stem Cell Reports 2022; 18:113-130. [PMID: 36400029 PMCID: PMC9859932 DOI: 10.1016/j.stemcr.2022.10.015] [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: 04/09/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/18/2022] Open
Abstract
The emergence of single-cell RNA sequencing (RNA-seq) has radically changed the observation of cellular diversity. Although annotations of RNA-seq data require preserved properties among cells of an identity, annotations using conventional methods have not been able to capture universal characters of a cell type. Analysis of expression levels cannot be accurately annotated for cells because differences in transcription do not necessarily explain biological characteristics in terms of cellular functions and because the data themselves do not inform about the correct mapping between cell types and genes. Hence, in this study, we developed a new representation of cellular identities that can be compared over different datasets while preserving nontrivial biological semantics. To generalize the notion of cell types, we developed a new framework to manage cellular identities in terms of set theory. We provided further insights into cells by installing mathematical descriptions of cell biology. We also performed experiments that could correspond to practical applications in annotations of RNA-seq data.
Collapse
Affiliation(s)
- Yuji Okano
- Department of Physiology, Keio University School of Medicine, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Yoshitaka Kase
- Department of Physiology, Keio University School of Medicine, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.
| |
Collapse
|
33
|
Chen H, Chen L, Yuan Z, Yuan J, Li Y, Xu Y, Wu J, Zhang L, Wang G, Li J. Glutamate receptor-interacting protein 1 in D1- and D2-dopamine receptor-expressing medium spiny neurons differentially regulates cocaine acquisition, reinstatement, and associated spine plasticity. Front Cell Neurosci 2022; 16:979078. [PMID: 36406750 PMCID: PMC9669444 DOI: 10.3389/fncel.2022.979078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/17/2022] [Indexed: 09/12/2023] Open
Abstract
BACKGROUND The nucleus accumbens (NAc) is involved in the expression of cocaine addictive phenotypes, including acquisition, extinction, and reinstatement. In the NAc, D1-medium spiny neurons (MSNs) encode cocaine reward, whereas D2-MSNs encode aversive responses in drug addiction. Glutamate receptor-interacting protein 1 (GRIP1) is known to be associated with cocaine addiction, but the role of GRIP1 in D1-MSNs and D2-MSNs of the NAc in cocaine acquisition and reinstatement remains unknown. METHODS A conditioned place preference apparatus was used to establish cocaine acquisition, extinction, and reinstatement in mouse models. GRIP1 expression was evaluated using Western blotting. Furthermore, GRIP1-siRNA and GRIP1 overexpression lentivirus were used to interfere with GRIP1 in the NAc. After the behavioral test, green fluorescent protein immunostaining of brain slices was used to detect spine density. RESULTS GRIP1 expression decreased during cocaine acquisition and reinstatement. GRIP1-siRNA enhanced cocaine-induced CPP behavior in acquisition and reinstatement and regulated associated spine plasticity. Importantly, the decreased GRIP1 expression that mediated cocaine acquisition and reinstatement was mainly driven by the interference of the GRIP1-GluA2 interaction in D1-MSNs and could be blocked by the interference of the GRIP1-GluA2 interaction in D2-MSNs. Interference with the GRIP1-GluA2 interaction in D1- and D2-MSNs decreased spine density in D1- and D2-MSNs, respectively. CONCLUSION GRIP1 in D1- and D2-MSNs of the NAc differentially modulates cocaine acquisition and reinstatement. GRIP1 downregulation in D1-MSNs has a positive effect on cocaine acquisition and reinstatement, while GRIP1 downregulation in D2-MSNs has a negative effect. Additionally, GRIP1 downregulation in D1-MSNs plays a leading role in cocaine acquisition and reinstatement.
Collapse
Affiliation(s)
- He Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Limei Chen
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zhirong Yuan
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiajie Yuan
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yitong Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuesi Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jieyi Wu
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lu Zhang
- Key Laboratory of Functional Proteomics of Guangdong Province, Key Laboratory of Mental Health of the Ministry of Education, School of Basic Medical Sciences, Pediatric Center of Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guohua Wang
- School of Food and Biotechnology, Guangdong Industry Polytechnic, Guangzhou, China
| | - Juan Li
- Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
34
|
circXRCC5 foster gastric cancer growth and metastasis by the HNRNPC/circXRCC5/miR-655-3p/RREB1/UBA2 positive feedback loop. Cancer Gene Ther 2022; 29:1648-1661. [PMID: 35661832 DOI: 10.1038/s41417-022-00482-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/22/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023]
Abstract
Gastric cancer (GC) is one of the most common malignancies, leading to millions of deaths each year. Here, we investigated the molecular mechanisms of GC, with a focus on circXRCC5/miR-655-3p/RREB1/UBA2 axis. circXRCC5 was identified in 62 paired cancer specimens and adjacent normal tissues by genome-wide bioinformatics analysis and verified by qRT-PCR and Sanger sequencing. Knockdown or exogenous expression of circXRCC5 was performed to validate the functional significance of circXRCC5 using both in vitro and in vivo assays, including CCK-8, colony formation, EdU incorporation, transwell system, as well as animal experiments. RNA immunoprecipitation, biotinylated RNA pull-down, ChIP, and dual-luciferase assays were employed to validate the regulatory network of circXRCC5/miR-655-3p/RREB1/UBA2. Frequently elevated circXRCC5 in GC tissues and cell lines was associated with poor prognosis of GC patients. Functionally, circXRCC5 overexpression facilitated GC cell proliferation, migration, and invasion, as well as promoted tumor growth and metastasis in vivo. Mechanistically, circXRCC5 served as a sponge of miR-655-3p to induce upregulation of RREB1. RREB1 was identified as a transcriptional activator of UBA2, thus contributing to GC tumorigenesis. Moreover, RNA binding protein (RBP) HNRNPC was proved to interact with circXRCC5 to promote circXRCC5 biogenesis. Collectively, circXRCC5 facilitates GC progression through the HNRNPC/circXRCC5/miR-655-3p/RREB1/UBA2 axis, which might bring novel therapeutic strategies for GC treatment.
Collapse
|
35
|
Learning and memory impairment and transcriptomic profile in hippocampus of offspring after maternal fructose exposure during gestation and lactation. Food Chem Toxicol 2022; 169:113394. [PMID: 36049592 DOI: 10.1016/j.fct.2022.113394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/06/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022]
Abstract
Increased fructose intake is a global issue, especially in mothers. Maternal fructose exposure during gestation and lactation can affect learning and memory in offspring; however, the detailed mechanism is still unknown. The hippocampus is a mind locale liable for learning and memory. Here, we established a maternal high-fructose diet model by administering 13% and 40% fructose water, applied the Morris Water Maze test on postnatal day 60 offspring, and performed full-length RNA sequencing using the Oxford Nanopore Technologies platform to explore the changes in gene expression in the hippocampus. The results showed that learning and memory in offspring were negatively affected. Compared with the control group, 369 differentially expressed transcripts (DETs) were identified in the 13% fructose group, and 501 DETs were identified in the 40% fructose group. Gene Ontology enriched term and Kyoto Encyclopedia of Genes and Genomes enriched pathway analyses identified several terms and pathways related to brain development and cognitive function. Furthermore, we confirmed that the Wnt/β-catenin signaling pathway was down-regulated and neuron degeneration was enhanced. In summary, our results indicate that maternal fructose exposure during gestation and lactation can impair learning and memory in offspring and affect brain function at the transcriptome level.
Collapse
|
36
|
Glutamate Receptor Interacting Protein 1 in the Dorsal CA1 Drives Alpha-amino-3-hydroxy-5-methyl-4-Isoxazolepropionic Acid Receptor Endocytosis and Exocytosis Bidirectionally and Mediates Forgetting, Exploratory, and Anxiety-like Behavior. Neuroscience 2022; 498:235-248. [PMID: 35863680 DOI: 10.1016/j.neuroscience.2022.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 01/21/2023]
Abstract
Endocytosis of GluA2-containing alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) in CA1 of the hippocampus regulates forgetting; deficits in forgetting nociceptive memory can induce serious stress disorders. As a transporter of GluA2-containing AMPAR, the functions of glutamate receptor interacting protein 1 (GRIP1) in forgetting and possible stress responses remain unclear. Lentivirus-mediated interference of GRIP1 expression or function in the dorsal CA1 of the hippocampus in mice indicated that GRIP1 overexpression enhanced spatial memory, impaired forgetting in a Barnes maze, and induced anxiety-like behavior in the open field and elevated plus-maze test. In contrast, GRIP1 knockdown impaired learning capacity. Furthermore, inhibition of the PDZ2 and PDZ4/5 domains of GRIP1 and GluA2-dn enhanced learning capacity, whereas GluA2-dn impaired spatial memory; inhibition of the PDZ2 and PDZ4/5 domains of GRIP1 also decreased forgetting capacity to some extent. Importantly, inhibition of both the PDZ2 and PDZ4/5 domains of GRIP1 induced anxiety-like behavior but not GluA2-dn. Furthermore, optogenetic control of both GluA1 and GluA2 insertion into the postsynaptic membrane impaired memory and induced anxiety-like behavior. In vitro experiments showed that GRIP1-ov and -dn, inhibition of PDZ2 and PDZ4/5 domains of GRIP1, and GluA2-dn decreased glycine-induced GluA1 and GluA2 exocytosis; meanwhile, GRIP1-ov and -dn, and interference of PDZ2 and PDZ4/5 domains of GRIP1 blocked AMPA- and NMDA-induced GluA1 and GluA2 endocytosis. Overall, these results suggest that GRIP1 drives AMPA receptor endocytosis and exocytosis bidirectionally; furthermore, GRIP1-induced stabilization of anchoring postsynaptic GluA1 and GluA2 impairs forgetting and induces anxiety-like behavior. GRIP1 may provide a potential therapeutic target in posttraumatic syndromes and anxiety disorders.
Collapse
|
37
|
Dong L, Huang J, Gao X, Du J, Wang Y, Zhao L. CircPCBP2 promotes the stemness and chemoresistance of DLBCL via targeting miR-33a/b to disinhibit PD-L1. Cancer Sci 2022; 113:2888-2903. [PMID: 35579082 PMCID: PMC9357607 DOI: 10.1111/cas.15402] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 05/07/2022] [Accepted: 05/09/2022] [Indexed: 11/28/2022] Open
Abstract
Diffuse large B‐cell lymphoma (DLBCL) is the most common lymphoid malignancy with a high relapse rate of up to 40%. The prognosis of the disease needs improvement and requires a understanding of its molecular mechanism. We investigated the mechanisms of DLBCL development and its sensitivity to chemotherapy by focusing on circPCBP2/miR‐33a/b/PD‐L1 axis. Human DLBCL specimens and cultured cancer cell lines were used. Features of circPCBP2 were systematically characterized through Sanger sequencing, Actinomycin D, RNase R treatment, and FISH. The expression levels of circPCBP2, miR‐33a/b, PD‐L1, stemness‐related markers, ERK/AKT and JAK2/STAT3 signaling were measured using qRT‐PCR, western blotting, and immunohistochemistry. Stemness of DLBCL cells was assessed through spheroid formation assay and flow cytometry. Cell viability and apoptosis upon cyclophosphamide, doxorubicin, vincristine, and prednisone (CHOP) treatment were determined using MTT assay and flow cytometry, respectively. Interactions of circPCBP2‐miR‐33a/b and miR‐33a/b‐PD‐L1 were validated using dual luciferase activity assay and RNA‐RIP. Nude mouse xenograft model was used to assess the function of circPCBP2 in DLBCL growth in vivo. circPCBP2 was upregulated in human DLBCL specimens and cultured DLBCL cells while miR‐33a/b was reduced. Knockdown of circPCBP2 or miR‐33a/b overexpression inhibited the stemness of DLBCL cells and promoted cancer cell apoptosis upon CHOP treatment. circPCBP2 directly bound with miR‐33a/b while miR‐33a/b targeted PD‐L1 3’‐UTR. circPCBP2 disinhibited PD‐L1 signaling via sponging miR‐33a/b. miR‐33a/b inhibitor and activating PD‐L1 reversed the effects of circPCBP2 knockdown and miR‐33a/b mimics, respectively. circPBCP2 knockdown restrained DLBCL growth in vivo and potentiated the anti‐tumor effects of CHOP. In conclusion, circPCBP2 enhances DLBCL cell stemness but suppresses its sensitivity to CHOP via sponging miR‐33a/b to disinhibit PD‐L1 expression. circPCBP2/miR‐33a/b/PD‐L1 axis could serve as a diagnosis marker or therapeutic target for DLBCL.
Collapse
Affiliation(s)
- Lihua Dong
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan Province, P.R. China
| | - Jingjing Huang
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan Province, P.R. China
| | - Xue Gao
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan Province, P.R. China
| | - Jianwei Du
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan Province, P.R. China
| | - Yesheng Wang
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou 450008, Henan Province, P.R. China
| | - Lingdi Zhao
- Department of Immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou 450008, Henan Province, P.R. China
| |
Collapse
|
38
|
Choquet D, Opazo P. The role of AMPAR lateral diffusion in memory. Semin Cell Dev Biol 2022; 125:76-83. [PMID: 35123863 DOI: 10.1016/j.semcdb.2022.01.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/17/2022]
Abstract
The accumulation of AMPARs to synapses is a fundamental step in Long-term potentiation (LTP) of synaptic transmission, a well-established cellular correlate of learning and memory. The discovery of a sizeable and highly mobile population of extrasynaptic AMPARs - randomly scanning the synaptic surface under basal conditions - provided a conceptual framework for a simplified model: LTP can be induced by the capture, and hence accumulation, of laterally diffusing extrasynaptic AMPARs. Here, we review the evidence supporting a rate-limiting role of AMPAR lateral diffusion in LTP and as consequence, in learning and memory. We propose that there are "multiple solutions" for achieving the diffusional trapping of AMPAR during LTP, mainly mediated by the interaction between interchangeable AMPAR auxiliary subunits and cell-adhesion molecules containing PDZ-binding domains and synaptic scaffolds containing PDZ-domains. We believe that this molecular degeneracy in the diffusional trapping of AMPAR during LTP serve to ensure the robustness of this crucial step in the making of memories. All in all, the role of AMPAR lateral diffusion in LTP is not only a conceptual leap in our understanding of memory, but it might also hold the keys for the development of therapeutics against disorders associated with memory deficits such as Alzheimer's disease.
Collapse
Affiliation(s)
- Daniel Choquet
- Interdisciplinary Institute for Neuroscience, CNRS, Univ. Bordeaux, IINS, UMR 5297, Bordeaux, France; Univ. Bordeaux, CNRS, INSERM, Bordeaux Imaging Center, BIC, UMS 3420, Bordeaux, France.
| | - Patricio Opazo
- UK Dementia Research Institute, Centre for Discovery Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh Medical School, Edinburgh EH16 4SB, UK; Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
39
|
Chater TE, Goda Y. The Shaping of AMPA Receptor Surface Distribution by Neuronal Activity. Front Synaptic Neurosci 2022; 14:833782. [PMID: 35387308 PMCID: PMC8979068 DOI: 10.3389/fnsyn.2022.833782] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 02/25/2022] [Indexed: 12/29/2022] Open
Abstract
Neurotransmission is critically dependent on the number, position, and composition of receptor proteins on the postsynaptic neuron. Of these, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs) are responsible for the majority of postsynaptic depolarization at excitatory mammalian synapses following glutamate release. AMPARs are continually trafficked to and from the cell surface, and once at the surface, AMPARs laterally diffuse in and out of synaptic domains. Moreover, the subcellular distribution of AMPARs is shaped by patterns of activity, as classically demonstrated by the synaptic insertion or removal of AMPARs following the induction of long-term potentiation (LTP) and long-term depression (LTD), respectively. Crucially, there are many subtleties in the regulation of AMPARs, and exactly how local and global synaptic activity drives the trafficking and retention of synaptic AMPARs of different subtypes continues to attract attention. Here we will review how activity can have differential effects on AMPAR distribution and trafficking along with its subunit composition and phosphorylation state, and we highlight some of the controversies and remaining questions. As the AMPAR field is extensive, to say the least, this review will focus primarily on cellular and molecular studies in the hippocampus. We apologise to authors whose work could not be cited directly owing to space limitations.
Collapse
|
40
|
circDlgap4 Alleviates Cerebral Ischaemic Injury by Binding to AUF1 to Suppress Oxidative Stress and Neuroinflammation. Mol Neurobiol 2022; 59:3218-3232. [PMID: 35294732 DOI: 10.1007/s12035-022-02796-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 03/09/2022] [Indexed: 10/18/2022]
Abstract
Ischaemic stroke is one of the most common causes of mortality and morbidity.circDlgap4 has been implicated in ischemia/reperfusion injury through an unknown mechanism. Here, we studied the function of circDlgap4/AUF1 in ischaemic stroke and its underlying molecular mechanism. N2a cells and primary mouse cortical neurons were subjected to OGD to mimic neuronal injury during ischemia. BV-2 cells were treated with LPS to mimic neuroinflammation. The MTT assay was used to assess cell viability, while flow cytometry was used to measure cell apoptosis. qRT-PCR, western blotting, immunohistochemistry, and immunostaining were employed to determine the levels of circDlgap4, AUF1, NRF2/HO-1, proinflammatory cytokines, NF-κB pathway-related proteins, and IBA-1. RIP and RNA pulldown assays were employed to validate the interactions of circDlgap4/AUF1, AUF1/NRF2, and AUF1/cytokine mRNAs. mRNA degradation was used to determine the effects on mRNA stability. The tMCAO model was used as an in vivo model of ischaemic stroke. TCC staining and neurological scoring were performed to evaluate ischaemic injury. circDlgap4 was decreased following OGD and during tMCAO. circDlgap4 overexpression inhibited OGD-induced cell death and oxidative stress and LPS-induced increases in proinflammatory cytokines by increasing NRF2/HO-1. Knockdown of AUF1 blocked the effects of circDlgap4 overexpression. Mechanistically, RIP, RNA pulldown, and mRNA degradation assay results showed circDlgap4/AUF1/NRF2 mRNA formed a complex to stabilize NRF2 mRNA. Furthermore, AUF1 directly interacted with TNF-α, IL-1β, and COX-2 mRNAs, and circDlgap4/AUF1 binding promoted the degradation of these mRNAs. Finally, circDlgap4 ameliorated ischaemic injury in vivo. circDlgap4 alleviates ischaemic stroke injury by suppressing oxidative stress and neuroinflammation by binding to AUF1.
Collapse
|
41
|
He JG, Zhou HY, Wang F, Chen JG. Dysfunction of Glutamatergic Synaptic Transmission in Depression: Focus on AMPA Receptor Trafficking. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2022; 3:187-196. [PMID: 37124348 PMCID: PMC10140449 DOI: 10.1016/j.bpsgos.2022.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/06/2022] [Accepted: 02/22/2022] [Indexed: 11/26/2022] Open
Abstract
Pharmacological and anatomical evidence suggests that abnormal glutamatergic neurotransmission may be associated with the pathophysiology of depression. Compounds that act as NMDA receptor antagonists may be a potential treatment for depression, notably the rapid-acting agent ketamine. The rapid-acting and sustained antidepressant effects of ketamine rely on the activation of AMPA receptors (AMPARs). As the key elements of fast excitatory neurotransmission in the brain, AMPARs are crucially involved in synaptic plasticity and memory. Recent efforts have been directed toward investigating the bidirectional dysregulation of AMPAR-mediated synaptic transmission in depression. Here, we summarize the published evidence relevant to the dysfunction of AMPAR in stress conditions and review the recent progress toward the understanding of the involvement of AMPAR trafficking in the pathophysiology of depression, focusing on the roles of AMPAR auxiliary subunits, key AMPAR-interacting proteins, and posttranslational regulation of AMPARs. We also discuss new prospects for the development of improved therapeutics for depression.
Collapse
|
42
|
Xing W, Zhou PC, Zhang HY, Chen LM, Zhou YM, Cui XF, Liu ZG. Circular RNA circ_GLIS2 suppresses hepatocellular carcinoma growth and metastasis. Liver Int 2022; 42:682-695. [PMID: 34743403 DOI: 10.1111/liv.15097] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/12/2021] [Accepted: 11/02/2021] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) is one of the leading causes of tumour-related death. Here, we investigated the molecular mechanism of HCC by studying the function of circ_GLIS2. METHODS Human HCC specimens and cell lines were used. Sanger sequencing, actinomycin D and RNase R treatment were performed to validate circular RNA features of circ_GLIS2. qRT-PCR, western blotting, immunostaining, and IHC were employed to examine levels of circ_GLIS2, GLIS2 mRNA, and EMT-related markers. CCK-8, colony formation, flow cytometry, wound healing assay, and transwell assays were performed to evaluate cancer cell proliferation, apoptosis, migration, and invasion. RIP and RNA pull-down assay were used to validate EIF4A3/GLIS2 mRNA interaction. MSP was performed to measure the methylation status of GLIS2 promoter. Nude mouse xenograft model was used to examine tumour growth and metastasis in vivo. RESULTS Circ_GLIS2 and linear GLIS2 mRNA were reduced in human HCC tissues and cells. Their low levels correlated with a poor survival rate of HCC patients. Overexpression of circ_GLIS2 and GLIS2 suppressed HCC cell proliferation, migration, and invasion but promoted cell apoptosis. GLIS2 promoter region was hypermethylated in HCC cells. EIF4A3 was directly bound with GLIS2 mRNA and promoted circ_GLIS2/GLIS2 expression. Moreover, overexpression of circ_GLIS2 restrained HCC tumour growth and metastasis in vivo. CONCLUSION Circ_GLIS2 suppresses HCC growth and metastasis by inhibiting cell proliferation, migration, and invasion, but promoting cell apoptosis. These findings provide molecular insights into the mechanism of HCC and indicate that circ_GLIS2 could serve as a diagnosis marker or therapeutic target for HCC.
Collapse
Affiliation(s)
- Wu Xing
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Peng-Cheng Zhou
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Hao-Ye Zhang
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Chen
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yang-Mei Zhou
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xue-Fei Cui
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Zhen-Guo Liu
- Department of Infectious Disease, The Third Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
43
|
Potier B, Lallemant L, Parrot S, Huguet-Lachon A, Gourdon G, Dutar P, Gomes-Pereira M. DM1 Transgenic Mice Exhibit Abnormal Neurotransmitter Homeostasis and Synaptic Plasticity in Association with RNA Foci and Mis-Splicing in the Hippocampus. Int J Mol Sci 2022; 23:ijms23020592. [PMID: 35054778 PMCID: PMC8775431 DOI: 10.3390/ijms23020592] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 12/31/2021] [Accepted: 01/02/2022] [Indexed: 02/01/2023] Open
Abstract
Myotonic dystrophy type 1 (DM1) is a severe neuromuscular disease mediated by a toxic gain of function of mutant RNAs. The neuropsychological manifestations affect multiple domains of cognition and behavior, but their etiology remains elusive. Transgenic DMSXL mice carry the DM1 mutation, show behavioral abnormalities, and express low levels of GLT1, a critical regulator of glutamate concentration in the synaptic cleft. However, the impact of glutamate homeostasis on neurotransmission in DM1 remains unknown. We confirmed reduced glutamate uptake in the DMSXL hippocampus. Patch clamp recordings in hippocampal slices revealed increased amplitude of tonic glutamate currents in DMSXL CA1 pyramidal neurons and DG granule cells, likely mediated by higher levels of ambient glutamate. Unexpectedly, extracellular GABA levels and tonic current were also elevated in DMSXL mice. Finally, we found evidence of synaptic dysfunction in DMSXL mice, suggestive of abnormal short-term plasticity, illustrated by an altered LTP time course in DG and in CA1. Synaptic dysfunction was accompanied by RNA foci accumulation in localized areas of the hippocampus and by the mis-splicing of candidate genes with relevant functions in neurotransmission. Molecular and functional changes triggered by toxic RNA may induce synaptic abnormalities in restricted brain areas that favor neuronal dysfunction.
Collapse
Affiliation(s)
- Brigitte Potier
- LuMIn, CNRS FRE2036, ENS Paris-Saclay, CentraleSupelec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (B.P.); (P.D.)
| | - Louison Lallemant
- Centre de Recherche en Myologie, Institut de Myologie, Inserm, Sorbonne Université, 75013 Paris, France; (L.L.); (A.H.-L.)
| | - Sandrine Parrot
- Lyon Neuroscience Research Center, Inserm U1028, CNRS UMR5292, Université Lyon 1, 69500 Bron, France;
| | - Aline Huguet-Lachon
- Centre de Recherche en Myologie, Institut de Myologie, Inserm, Sorbonne Université, 75013 Paris, France; (L.L.); (A.H.-L.)
| | - Geneviève Gourdon
- Centre de Recherche en Myologie, Institut de Myologie, Inserm, Sorbonne Université, 75013 Paris, France; (L.L.); (A.H.-L.)
- Correspondence: (G.G.); (M.G.-P.)
| | - Patrick Dutar
- LuMIn, CNRS FRE2036, ENS Paris-Saclay, CentraleSupelec, Université Paris-Saclay, 91190 Gif-sur-Yvette, France; (B.P.); (P.D.)
| | - Mário Gomes-Pereira
- Centre de Recherche en Myologie, Institut de Myologie, Inserm, Sorbonne Université, 75013 Paris, France; (L.L.); (A.H.-L.)
- Correspondence: (G.G.); (M.G.-P.)
| |
Collapse
|
44
|
Yu Y, Ou-Yang WX, Zhang H, Jiang T, Tang L, Tan YF, Luo HY, Xiao ZH, Li SJ. MiR-125b enhances autophagic flux to improve septic cardiomyopathy via targeting STAT3/HMGB1. Exp Cell Res 2021; 409:112842. [PMID: 34563514 DOI: 10.1016/j.yexcr.2021.112842] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 01/05/2023]
Abstract
We explore the role of miR-125b in septic cardiomyopathy, focusing on miR-125b/STAT3/HMGB1 axis. CLP mouse model and LPS-stimulated primary rat cardiomyocytes (CMs) and H9C2 cell were used as in vivo and in vitro models of septic cardiomyopathy, respectively. qRT-PCR and western blot were performed to measure expression levels of miR-125b, STAT3, HMGB1, and autophagy-related proteins. MTT assay was employed to examine LPS toxicity. Dual luciferase activity assay and CHIP were performed to validate interactions of miR-125b/STAT3 and STAT3/HMGB1 promoter. Immunostaining was used to assess the level of autophagic flux. ROS level was measured by fluorescence assay. Heart functions were examined via intracoronary Doppler ultrasound. miR-125b was diminished while STAT3 and HMGB1 were elevated in the heart tissue following CLP surgery and in LPS-treated H9C2 cells. LPS treatment up-regulated ROS generation and suppressed autophagic flux. Overexpression of miR-125b mimics or knockdown of STAT3 or HMGB1 alleviated LPS-induced hindrance of autophagic flux and ROS production. miR-125b directly targeted STAT3 mRNA and STAT3 bound with HMGB1 promoter. Overexpression of miR-125b mitigated myocardial dysfunction induced by CLP in vivo. Hyperactivation of STAT3/HMGB1 caused by reduced miR-125b contributes to ROS generation and the hindrance of autophagic flux during septic cardiomyopathy, leading to myocardial dysfunction.
Collapse
Affiliation(s)
- Ying Yu
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Wen-Xian Ou-Yang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Hui Zhang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Tao Jiang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Lian Tang
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Yan-Fang Tan
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Hai-Yan Luo
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Zheng-Hui Xiao
- Emergence Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China
| | - Shuang-Jie Li
- Liver Disease Center, Hunan Children's Hospital, Changsha, 410007, Hunan Province, PR China.
| |
Collapse
|
45
|
Meftahi GH, Jangravi Z, Taghdir M, Sepandi M, Bahari Z. Micro-injection of propranolol within basolateral amygdala impaired fear and spatial memory and dysregulated evoked responses of CA1 neurons following foot shock stress in rats. Brain Res Bull 2021; 177:12-21. [PMID: 34534638 DOI: 10.1016/j.brainresbull.2021.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 11/18/2022]
Abstract
The basolateral nucleus of the amygdala (BLA) is responsible for memory retrieval after stress. It regulates hippocampal long-term potentiation (LTP) during stress. Although β-adrenoceptors of the BLA have a critical role in memory, few studies have addressed this question in the BLA, and the results still have been contradictory. The present study was designed to investigate the involvement of β-adrenoceptors of the BLA on hippocampus memory, anxiety, and plasticity in intact and stressed rats. Male Wistar rats were submitted to the electrical foot-shock stress for four consecutive days. Over four consecutive days, animals received bilateral micro-injections of either vehicle or propranolol (4 µg in 1 µl/side) into the BLA (5 min before foot-shock stress). Behavioral (memory, as well as anxiety-like behaviors), electrophysiological, and histological (neural arborization in the hippocampal CA1 pyramidal neurons) studies were performed. Results showed that inhibition of β-adrenoceptors of BLA by propranolol significantly further impaired fear and spatial memory in stressed rats. Similarly, propranolol effectively impaired both memories in the intact animals. Propranolol significantly amplified the slope and amplitude of fEPSP in the CA1 area of the hippocampus only in stressed rats. Foot-shock stress significantly increased the number of dendritic branches in the hippocampus, and propranolol suppressed this effect of stress. It is suggested that β-adrenoceptors in the BLA promote memory and reduce anxiety-like behaviors under tonic and stress conditions. Propranolol dysregulated LTP parameters and reduced dendritic branches, resulting in memory impairment. Probably β-adrenoceptors of BLA regulate evoked responses of CA1 neurons only in stress- and not the tonic condition.
Collapse
Affiliation(s)
| | - Zohreh Jangravi
- Department of Biochemistry, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Taghdir
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mojtaba Sepandi
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Epidemiology and Biostatistics, Faculty of Health, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Zahra Bahari
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran; Department of Physiology and Medical Physics, Faculty of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
46
|
Henley JM, Nair JD, Seager R, Yucel BP, Woodhall G, Henley BS, Talandyte K, Needs HI, Wilkinson KA. Kainate and AMPA receptors in epilepsy: Cell biology, signalling pathways and possible crosstalk. Neuropharmacology 2021; 195:108569. [PMID: 33915142 DOI: 10.1016/j.neuropharm.2021.108569] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/13/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023]
Abstract
Epilepsy is caused when rhythmic neuronal network activity escapes normal control mechanisms, resulting in seizures. There is an extensive and growing body of evidence that the onset and maintenance of epilepsy involves alterations in the trafficking, synaptic surface expression and signalling of kainate and AMPA receptors (KARs and AMPARs). The KAR subunit GluK2 and AMPAR subunit GluA2 are key determinants of the properties of their respective assembled receptors. Both subunits are subject to extensive protein interactions, RNA editing and post-translational modifications. In this review we focus on the cell biology of GluK2-containing KARs and GluA2-containing AMPARs and outline how their regulation and dysregulation is implicated in, and affected by, seizure activity. Further, we discuss role of KARs in regulating AMPAR surface expression and plasticity, and the relevance of this to epilepsy. This article is part of the special issue on 'Glutamate Receptors - Kainate receptors'.
Collapse
Affiliation(s)
- Jeremy M Henley
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK; Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia.
| | - Jithin D Nair
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Richard Seager
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Busra P Yucel
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Gavin Woodhall
- School of Life and Health Sciences, Aston University, Aston Triangle, Birmingham, B4 7ET, UK
| | - Benjamin S Henley
- Faculty of Medical Sciences, The Medical School, Newcastle University, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Karolina Talandyte
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Hope I Needs
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Kevin A Wilkinson
- School of Biochemistry, Centre for Synaptic Plasticity, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK.
| |
Collapse
|
47
|
Matthews PM, Pinggera A, Kampjut D, Greger IH. Biology of AMPA receptor interacting proteins - From biogenesis to synaptic plasticity. Neuropharmacology 2021; 197:108709. [PMID: 34271020 DOI: 10.1016/j.neuropharm.2021.108709] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/19/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022]
Abstract
AMPA-type glutamate receptors mediate the majority of excitatory synaptic transmission in the central nervous system. Their signaling properties and abundance at synapses are both crucial determinants of synapse efficacy and plasticity, and are therefore under sophisticated control. Unique to this ionotropic glutamate receptor (iGluR) is the abundance of interacting proteins that contribute to its complex regulation. These include transient interactions with the receptor cytoplasmic tail as well as the N-terminal domain locating to the synaptic cleft, both of which are involved in AMPAR trafficking and receptor stabilization at the synapse. Moreover, an array of transmembrane proteins operate as auxiliary subunits that in addition to receptor trafficking and stabilization also substantially impact AMPAR gating and pharmacology. Here, we provide an overview of the catalogue of AMPAR interacting proteins, and how they contribute to the complex biology of this central glutamate receptor.
Collapse
Affiliation(s)
- Peter M Matthews
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Alexandra Pinggera
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Domen Kampjut
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
48
|
Long non-coding RNA CIR inhibits chondrogenic differentiation of mesenchymal stem cells by epigenetically suppressing ATOH8 via methyltransferase EZH2. Mol Med 2021; 27:12. [PMID: 33546582 PMCID: PMC7866678 DOI: 10.1186/s10020-021-00272-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/13/2021] [Indexed: 12/19/2022] Open
Abstract
Background Osteoarthritis (OA) is the most common articular disorder, leading to joint malfunction and disability. Although the incidence of OA is increasing globally, the treatment of OA is very limited. LncRNA CIR has been implicated in OA through unclear mechanisms. Here, we investigated the role of lncRNA CIR in chondrogenic differentiation. Methods Human umbilical-cord-derived mesenchymal stem cells (hUC-MSCs) were obtained from human umbilical cords. Flow cytometry was used to analyze the surface markers of hUC-MSCs. Various culture conditions and corresponding staining assays were employed to assess the differentiation abilities of hUC-MSC. qRT-PCR, western blot, and immunostaining were used to measure expression levels of related genes and proteins such as lncRNA CIR, ATOH8, EZH2, and H3K27me3. RNA immunoprecipitation assay, biotin pull-down, and chromatin immunoprecipitaion assay were performed to analyze the interactions of lncRNA CIR, EZH2, H3K27me3 and ATOH8 promoter. Results hUC-MSCs exhibited MSCs features and could differentiate into chondrocytes under specific conditions. LncRNA CIR was downregulated while ATOH8 was upregulated during the chondrogenic differentiation of hUC-MSCs. Knockdown lncRNA CIR or overexpression of ATOH8 promoted chondrogenic differentiation. Further, lncRNA CIR bound to EZH2 and repressed ATOH8 expression via EZH2-mediated H3K27me3, which promotes the methylation of ATOH8. Inhibition of ATOH8 reversed the effects of knockdown lncRNA CIR on chondrogenic differentiation. Conclusion LncRNA CIR suppresses chondrogenic differentiation of hUC-MSCs. Mechanistically, lncRNA CIR could inhibit ATOH8 expression that functions to promote chondrogenic differentiation through EZH2-mediated epigenetic modifications.
Collapse
|
49
|
Nong W, Wei ZQ, Mo XN, Wu L, Tang N. miR-137 overexpression protects neurons from Aβ-induced neurotoxicity via ERK1/2. ALL LIFE 2021. [DOI: 10.1080/26895293.2021.1932612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Wei Nong
- Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Zhi-quan Wei
- Scientific Experimental Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Xue-Ni Mo
- Guangxi Key Laboratory of Basic Research in Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Lin Wu
- Guangxi Key Laboratory of Basic Research in Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| | - Nong Tang
- Guangxi Key Laboratory of Basic Research in Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, People’s Republic of China
| |
Collapse
|