1
|
Illanes-González J, Flores-Muñoz C, Vitureira N, Ardiles ÁO. Pannexin 1 channels: A bridge between synaptic plasticity and learning and memory processes. Neurosci Biobehav Rev 2025; 174:106173. [PMID: 40274202 DOI: 10.1016/j.neubiorev.2025.106173] [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: 02/11/2025] [Revised: 04/13/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
The Pannexin 1 channel is a membrane protein widely expressed in various vertebrate cell types, including microglia, astrocytes, and neurons within the central nervous system. Growing research has demonstrated the significant involvement of Panx1 in synaptic physiology, such as its contribution to long-term synaptic plasticity, with a particular focus on the hippocampus, an essential structure for learning and memory. Investigations studying the role of Panx1 in synaptic plasticity have utilized knockout animal models and channel inhibition techniques, revealing that the absence or blockade of Panx1 channels in this region promotes synaptic potentiation, dendritic arborization, and spine formation. Despite substantial progress, the precise mechanism by which Panx1 regulates synaptic plasticity remains to be determined. Nevertheless, evidence suggests that Panx1 may exert its influence by releasing signaling molecules, such as adenosine triphosphate (ATP), or through the clearance of endocannabinoids (eCBs). This review aims to comprehensively explore the current literature on the role of Panx1 in synapses. By examining relevant articles, we seek to enhance our understanding of Panx1's contribution to synaptic fundamental processes and the potential implications for cognitive function.
Collapse
Affiliation(s)
- Javiera Illanes-González
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile
| | - Carolina Flores-Muñoz
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile
| | - Nathalia Vitureira
- Unidad Académica de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Álvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile; Centro para la Investigación Traslacional en Neurofarmacología, CItNe, Universidad de Valparaíso, Valparaíso, Chile; Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
2
|
Bartol TM, Ordyan M, Sejnowski TJ, Rangamani P, Kennedy MB. A spatial model of autophosphorylation of CaMKII predicts that the lifetime of phospho-CaMKII after induction of synaptic plasticity is greatly prolonged by CaM-trapping. Front Synaptic Neurosci 2025; 17:1547948. [PMID: 40255983 PMCID: PMC12006173 DOI: 10.3389/fnsyn.2025.1547948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/18/2025] [Indexed: 04/22/2025] Open
Abstract
Long-term potentiation (LTP) is a biochemical process that underlies learning in excitatory glutamatergic synapses in the Central Nervous System (CNS). A critical early driver of LTP is autophosphorylation of the abundant postsynaptic enzyme, Ca2+/calmodulin-dependent protein kinase II (CaMKII). Autophosphorylation is initiated by Ca2+ flowing through NMDA receptors activated by strong synaptic activity. Its lifetime is ultimately determined by the balance of the rates of autophosphorylation and of dephosphorylation by protein phosphatase 1 (PP1). Here we have modeled the autophosphorylation and dephosphorylation of CaMKII during synaptic activity in a spine synapse using MCell4, an open source computer program for creating particle-based stochastic, and spatially realistic models of cellular microchemistry. The model integrates four earlier detailed models of separate aspects of regulation of spine Ca2+ and CaMKII activity, each of which incorporate experimentally measured biochemical parameters and have been validated against experimental data. We validate the composite model by showing that it accurately predicts previous experimental measurements of effects of NMDA receptor activation, including high sensitivity of induction of LTP to phosphatase activity in vivo, and persistence of autophosphorylation for a period of minutes after the end of synaptic stimulation. We then use the model to probe aspects of the mechanism of regulation of autophosphorylation of CaMKII that are difficult to measure in vivo. We examine the effects of "CaM-trapping," a process in which the affinity for Ca2+/CaM increases several hundred-fold after autophosphorylation. We find that CaM-trapping does not increase the proportion of autophosphorylated subunits in holoenzymes after a complex stimulus, as previously hypothesized. Instead, CaM-trapping may dramatically prolong the lifetime of autophosphorylated CaMKII through steric hindrance of dephosphorylation by protein phosphatase 1. The results provide motivation for experimental measurement of the extent of suppression of dephosphorylation of CaMKII by bound Ca2+/CaM. The composite MCell4 model of biochemical effects of complex stimuli in synaptic spines is a powerful new tool for realistic, detailed dissection of mechanisms of synaptic plasticity.
Collapse
Affiliation(s)
- Thomas M. Bartol
- The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Mariam Ordyan
- The Salk Institute for Biological Studies, La Jolla, CA, United States
| | - Terrence J. Sejnowski
- The Salk Institute for Biological Studies, La Jolla, CA, United States
- Department of Neurobiology, University of California, San Diego, La Jolla, CA, United States
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, La Jolla, CA, United States
| | - Mary B. Kennedy
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
3
|
Mesa MH, McCabe KJ, Rangamani P. Synaptic cleft geometry modulates NMDAR opening probability by tuning neurotransmitter residence time. Biophys J 2025; 124:1058-1072. [PMID: 39876560 PMCID: PMC11993924 DOI: 10.1016/j.bpj.2025.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/27/2024] [Accepted: 01/23/2025] [Indexed: 01/30/2025] Open
Abstract
Synaptic morphology plays a critical role in modulating the dynamics of neurotransmitter diffusion and receptor activation in interneuron communication. Central physical aspects of synaptic geometry, such as the curvature of the synaptic cleft, the distance between the presynaptic and postsynaptic membranes, and the surface-area-to-volume ratio of the cleft, crucially influence glutamate diffusion and N-methyl-D-aspartate receptor (NMDAR) opening probabilities. In this study, we developed a stochastic model for receptor activation using realistic synaptic geometries. Our simulations revealed substantial variability in NMDAR activation, showing a significant impact of synaptic structure on receptor activation. Next, we designed a theoretical study with idealized cleft geometries to understand the impact of different biophysical properties on receptor activation. Specifically, we found that increasing the curvature of the synaptic membranes could compensate for reduced NMDAR activation when the synaptic cleft width was large. Additionally, nonparallel membrane configurations, such as convex presynapses or concave postsynaptic densities, maximize NMDAR activation by increasing the surface-area-to-volume ratio, thereby increasing glutamate residence time and reducing glutamate escape. Furthermore, clustering NMDARs within the postsynaptic density significantly increased receptor activation across different geometric conditions and mitigated the effects of synaptic morphology on NMDAR opening probabilities. These findings highlight the complex interplay between synaptic geometry and receptor dynamics and provide important insights into how structural modifications can influence synaptic efficacy and plasticity. By considering the major physical factors that affect neurotransmitter diffusion and receptor activation, our work offers a comprehensive understanding of how variations in synaptic geometry may regulate neurotransmission.
Collapse
Affiliation(s)
- María Hernández Mesa
- Department of Computational Physiology, Simula Research Laboratory, 0164 Oslo, Norway; Department of Informatics, University of Oslo, 0373 Oslo, Norway
| | - Kimberly J McCabe
- Department of Computational Physiology, Simula Research Laboratory, 0164 Oslo, Norway
| | - Padmini Rangamani
- Department of Pharmacology, School of Medicine, University of California, San Diego, San Diego, California; Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, California.
| |
Collapse
|
4
|
Bartol TM, Ordyan M, Sejnowski TJ, Rangamani P, Kennedy MB. A spatial model of autophosphorylation of Ca 2+/calmodulin-dependent protein kinase II (CaMKII) predicts that the lifetime of phospho-CaMKII after induction of synaptic plasticity is greatly prolonged by CaM-trapping. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578696. [PMID: 38352446 PMCID: PMC10862815 DOI: 10.1101/2024.02.02.578696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Long-term potentiation (LTP) is a biochemical process that underlies learning in excitatory glutamatergic synapses in the Central Nervous System (CNS). The critical early driver of LTP is autophosphorylation of the abundant postsynaptic enzyme, Ca2+/calmodulin-dependent protein kinase II (CaMKII). Autophosphorylation is initiated by Ca2+ flowing through NMDA receptors activated by strong synaptic activity. Its lifetime is ultimately determined by the balance of the rates of autophosphorylation and of dephosphorylation by protein phosphatase 1 (PP1). Here we have modeled the autophosphorylation and dephosphorylation of CaMKII during synaptic activity in a spine synapse using MCell4, an open source computer program for creating particle-based stochastic, and spatially realistic models of cellular microchemistry. The model integrates four earlier detailed models of separate aspects of regulation of spine Ca2+ and CaMKII activity, each of which incorporate experimentally measured biochemical parameters and have been validated against experimental data. We validate the composite model by showing that it accurately predicts previous experimental measurements of effects of NMDA receptor activation, including high sensitivity of induction of LTP to phosphatase activity in vivo, and persistence of autophosphorylation for a period of minutes after the end of synaptic stimulation. We then use the model to probe aspects of the mechanism of regulation of autophosphorylation of CaMKII that are difficult to measure in vivo. We examine the effects of "CaM-trapping," a process in which the affinity for Ca2+/CaM increases several hundred-fold after autophosphorylation. We find that CaM-trapping does not increase the proportion of autophosphorylated subunits in holoenzymes after a complex stimulus, as previously hypothesized. Instead, CaM-trapping may dramatically prolong the lifetime of autophosphorylated CaMKII through steric hindrance of dephosphorylation by protein phosphatase 1. The results provide motivation for experimental measurement of the extent of suppression of dephosphorylation of CaMKII by bound Ca2+/CaM. The composite MCell4 model of biochemical effects of complex stimuli in synaptic spines is a powerful new tool for realistic, detailed dissection of mechanisms of synaptic plasticity.
Collapse
Affiliation(s)
| | - Mariam Ordyan
- The Salk Institute for Biological Studies, La Jolla, CA
| | - Terrence J Sejnowski
- The Salk Institute for Biological Studies, La Jolla, CA
- Department of Neurobiology, University of California at San Diego, La Jolla, CA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA
| | - Mary B Kennedy
- Department of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| |
Collapse
|
5
|
Yuan X, Li W, Yuan Y, Zhu X, Meng Y, Wu Q, Yan Q, Zhang P. Characterization of neuronal differentiation in human adipose-derived stromal cells: morphological, molecular, and ultrastructural insights. J Neurosci Methods 2024; 412:110296. [PMID: 39357604 DOI: 10.1016/j.jneumeth.2024.110296] [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: 04/26/2024] [Revised: 09/19/2024] [Accepted: 09/28/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE Adipose-derived stromal cells (ADSCs) have shown promise as a potential source of neural differentiation. In this study, we investigated the morphological, molecular and ultrastructural features of ADSCs during neuronal differentiation. METHODS ADSCs were induced in vitro and their differentiation was examined at different time points. Immunocytochemical staining was performed to detect the expression of neuron-specific markers NSE and MAP-2. Immunofluorescence double labeling and Western blot detected the co-expression of presynaptic markers (CaMKII, SynCAM1, SYN) and postsynaptic markers (PSD-95, Synapsin I). Scanning electron microscopy (SEM) was performed to detect the synaptic structural features of differentiated neurons. RESULTS ADSCs showed diverse morphological features during differentiation, gradually acquiring a neuron-like spindle shape and organized arrangement. The expression of neuron-specific markers and synaptic markers peaked at 5 h of induction. Scanning electron microscopy showed polygonal protrusions of ADSC-derived neurons, and transmission electron microscopy showed characteristic ultrastructures such as nidus, synaptic vesicle-like structures, and tight junctions. CONCLUSION Our findings suggest that ADSCs differentiated for 5 h have neuronal features, including morphological, molecular, and ultrastructural resemblance to neurons, as well as the formation of synaptic structures. These insights contribute to a better understanding of ADSC-based neuronal differentiation and pave the way for future applications in regenerative medicine and neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China.
| | - Wen Li
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Yi Yuan
- Children's hospital of Capital institute of pediatrics, department of pediatric othopedic, Beijing 100000, China
| | - Xuhong Zhu
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Yan Meng
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Qi Wu
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Qi Yan
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China
| | - Pingshu Zhang
- Department of Neurology, Kailuan General Hospital affiliated to North China University of Science and Technology, Tangshan, Hebei Province 063000, China; Hebei Provincial Key Laboratory of Neurobiological Function, Tangshan, Hebei Province 063000, China.
| |
Collapse
|
6
|
Chen Y, Huang J, Zhou Z, Zhang J, Jin C, Zeng X, Jia J, Li L. Noise exposure-induced the cerebral alterations: From emerging evidence to antioxidant-mediated prevention and treatment. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117411. [PMID: 39591731 DOI: 10.1016/j.ecoenv.2024.117411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/27/2024] [Accepted: 11/23/2024] [Indexed: 11/28/2024]
Abstract
It's well acknowledged that noise exposure has become a major environmental risk factor of public health. The previous standpoint holds that the main harm of noise exposure is to cause hearing loss of human. However, in the past two decades a large number of studies have linked the noise exposure to various cerebral changes. In this review, we summarized that noise exposure led to cerebral changes through breaking the redox balance, inducing neuroinflammation and neuronal apoptosis and altering the neurotransmission in numerous brain areas, including cortex, thalamus, hippocampus, amygdala, striatum and cerebellum. Those cerebral changes finally result in a variety of disorders, such as tinnitus, anxiety, depression, cognitive impairment and motor dysfunction. Furthermore, we reviewed several antioxidants, such as resveratrol, vitamin C, curcumin, N-acetylcysteine and α-asarone, and highlighted their protective mechanisms against noise exposure, aiming to provide a promising strategy to prevent and treat noise exposure-induced diseases. Taken together, noise exposure induces various cerebral changes and further leads to disorders in the central nervous system, which can be ameliorated by the treatment with antioxidants.
Collapse
Affiliation(s)
- Yuyan Chen
- Research Center of Neuroscience, Jiaxing University, Jiaxing, China
| | - Jie Huang
- Research Center of Neuroscience, Jiaxing University, Jiaxing, China
| | - Zhiying Zhou
- Research Center of Neuroscience, Jiaxing University, Jiaxing, China
| | - Jiaping Zhang
- Research Center of Neuroscience, Jiaxing University, Jiaxing, China
| | - Chaohui Jin
- Research Center of Neuroscience, Jiaxing University, Jiaxing, China
| | - Xiansi Zeng
- Research Center of Neuroscience, Jiaxing University, Jiaxing, China; Department of Biochemistry and Molecular Biology, Jiaxing University Medical College, Jiaxing, China; Judicial Expertise Center, Jiaxing University, Jiaxing, China.
| | - Jinjing Jia
- Research Center of Neuroscience, Jiaxing University, Jiaxing, China; Department of Physiology, Jiaxing University Medical College, Jiaxing, China.
| | - Li Li
- Research Center of Neuroscience, Jiaxing University, Jiaxing, China; Department of Physiology, Jiaxing University Medical College, Jiaxing, China.
| |
Collapse
|
7
|
Xu SF, Cui JH, Liu X, Pang ZQ, Bai CY, Jiang C, Luan C, Li YP, Zhao Y, You YM, Guo C. Astrocytic lactoferrin deficiency augments MPTP-induced dopaminergic neuron loss by disturbing glutamate/calcium and ER-mitochondria signaling. Free Radic Biol Med 2024; 225:374-387. [PMID: 39406276 DOI: 10.1016/j.freeradbiomed.2024.10.284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/05/2024] [Accepted: 10/12/2024] [Indexed: 10/20/2024]
Abstract
Increased levels of lactoferrin (Lf) are present in the aged brain and in the lesions of various neurodegenerative diseases, including Parkinson's disease (PD), and may contribute to the cascade of events involved in neurodevelopment and neuroprotection. However, whether Lf originates from astrocytes and functions within either the normal or pathological brain are unknown. Here, we employed mice with specific knockout of the astrocyte lactoferrin gene (named Lf-cKO) to explore its specific roles in the pathological process of PD. We observed a decrease in tyrosine hydroxylase-positive cells, mitochondrial dysfunction of residual dopaminergic neurons, and motor deficits in Lf-cKO mice, which were significantly aggravated after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment. To further explore how astrocytic lactoferrin deficiency exacerbated PD-like manifestation in MPTP-treated mice, the critical molecules involved in endoplasmic reticulum (ER)-mitochondria contacts and signaling pathways were investigated. In vitro and in vivo models, we found an aberrant level of effects implicated in glutamate and calcium homeostasis, mitochondrial morphology and functions, mitochondrial dynamics, and mitochondria-associated ER membranes, accompanied by signs of oxidative stress and ER stress, which increase the fragility of dopaminergic neurons. These findings confirm the existence of astrocytic Lf and its influence on the fate of dopaminergic neurons by regulating glutamate/calcium metabolism and ER-mitochondria signaling. Our findings may be a promising target for the treatment of PD.
Collapse
Affiliation(s)
- Shuang-Feng Xu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Jun-He Cui
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Xin Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Zhong-Qiu Pang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chen-Yang Bai
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chao Jiang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chuang Luan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Yun-Peng Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Yan Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Yi-Ming You
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China
| | - Chuang Guo
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110169, China.
| |
Collapse
|
8
|
Zhong YJ, Liu LL, Zhao Y, Feng Z, Liu Y. Elucidating the molecular mechanisms behind the therapeutic impact of median nerve stimulation on cognitive dysfunction post-traumatic brain injury. Exp Gerontol 2024; 194:112500. [PMID: 38901771 DOI: 10.1016/j.exger.2024.112500] [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: 11/05/2023] [Revised: 05/22/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVE Ferroptosis represents a form of regulated cellular death dependent upon iron and lipid peroxidation derivatives, holding considerable implications for cerebral and neurologic pathologies. In the present study, we endeavored to elucidate the molecular mechanisms governing ferroptosis and appraise the therapeutic value of electrical stimulation of median nerve in addressing cognitive impairments following traumatic brain injury (TBI), employing a rodent model. METHODS In this study, we established a rat model to investigate the cognitive impairments resulting from TBI, followed by the application of median nerve stimulation (MNS). Initially, rats received an intraperitoneal injection of Erastin (2 mg/kg) prior to undergoing MNS. After 24 h of MNS treatment, the rats were subjected to an open field test to evaluate their cognitive and motor functions. Subsequently, we conducted biochemical assays to measure the serum levels of GSH, MDA and SOD. The structural integrity and cellular morphology of hippocampal tissue were examined through H&E staining, Nissl staining and transmission electron microscopy. Additionally, we assessed the expression levels of proteins crucial for neuronal health and function in the hippocampus, including VEGF, SLC7A11, GPX4, Nrf2, α-syn, NEUN and PSD95. RESULTS Compared to the control group, rats in the stimulation group demonstrated enhanced mobility, reduced levels of tissue damage, a decrease in MDA concentration, and increased levels of GSH and SOD. Additionally, there was a significant upregulation in the expression of proteins critical for cellular defense and neuronal health, including GPX4, SLC7A11, Nrf2, VEGF, α-syn, NEUN, and PSD95 proteins. Conversely, rats in the Erastin group demonstrated decreased mobility, exacerbated pathological tissue damage, elevated MDA concentration, and decreased levels of GSH and SOD. There was also a notable decrease in the expression of GPX4, SLC7CA11, Nrf2, and VEGF proteins. The expression levels of α-syn, NEUN, and PSD95 were similarly diminished in the Erastin group. Each of these findings was statistically significant, indicating that MNS exerts neuroprotective effect in the hippocampal tissue of rats with TBI by inhibiting the ferroptosis pathway. CONCLUSION (1) MNS may enhance the cognitive and behavioral performance of rats after TBI; (2) MNS can play a neuroprotective role by promoting the expression of nerve injury-related proteins, alleviating oxidative stress and ferroptosis process; (3) MNS may inhibit ferroptosis of neuronal cells by activating Nrf2/ GPX4 signaling pathway, thereby improving cognitive impairment in TBI rats.
Collapse
Affiliation(s)
- Ying-Jun Zhong
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Ling-Ling Liu
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Yue Zhao
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China
| | - Zhen Feng
- Department of Rehabilitation Medicine, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China.
| | - Yuan Liu
- Department of Orthopedics, The 1(st) Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330000, China.
| |
Collapse
|
9
|
Wang P, Shao Y, Al-Nusaif M, Zhang J, Yang H, Yang Y, Kim K, Li S, Liu C, Cai H, Le W. Pathological characteristics of axons and alterations of proteomic and lipidomic profiles in midbrain dopaminergic neurodegeneration induced by WDR45-deficiency. Mol Neurodegener 2024; 19:62. [PMID: 39183331 PMCID: PMC11346282 DOI: 10.1186/s13024-024-00746-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/17/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Although WD repeat domain 45 (WDR45) mutations have been linked to β -propeller protein-associated neurodegeneration (BPAN), the precise molecular and cellular mechanisms behind this disease remain elusive. This study aims to shed light on the impacts of WDR45-deficiency on neurodegeneration, specifically axonal degeneration, within the midbrain dopaminergic (DAergic) system. We hope to better understand the disease process by examining pathological and molecular alterations, especially within the DAergic system. METHODS To investigate the impacts of WDR45 dysfunction on mouse behaviors and DAergic neurons, we developed a mouse model in which WDR45 was conditionally knocked out in the midbrain DAergic neurons (WDR45cKO). Through a longitudinal study, we assessed alterations in the mouse behaviors using open field, rotarod, Y-maze, and 3-chamber social approach tests. We utilized a combination of immunofluorescence staining and transmission electron microscopy to examine the pathological changes in DAergic neuron soma and axons. Additionally, we performed proteomic and lipidomic analyses of the striatum from young and aged mice to identify the molecules and processes potentially involved in the striatal pathology during aging. Further more, primary midbrain neuronal culture was employed to explore the molecular mechanisms leading to axonal degeneration. RESULTS Our study of WDR45cKO mice revealed a range of deficits, including impaired motor function, emotional instability, and memory loss, coinciding with the profound reduction of midbrain DAergic neurons. The neuronal loss, we observed massive axonal enlargements in the dorsal and ventral striatum. These enlargements were characterized by the accumulation of extensively fragmented tubular endoplasmic reticulum (ER), a hallmark of axonal degeneration. Proteomic analysis of the striatum showed that the differentially expressed proteins were enriched in metabolic processes. The carbohydrate metabolic and protein catabolic processes appeared earlier, and amino acid, lipid, and tricarboxylic acid metabolisms were increased during aging. Of note, we observed a tremendous increase in the expression of lysophosphatidylcholine acyltransferase 1 (Lpcat1) that regulates phospholipid metabolism, specifically in the conversion of lysophosphatidylcholine (LPC) to phosphatidylcholine (PC) in the presence of acyl-CoA. The lipidomic results consistently suggested that differential lipids were concentrated on PC and LPC. Axonal degeneration was effectively ameliorated by interfering Lpcat1 expression in primary cultured WDR45-deficient DAergic neurons, proving that Lpcat1 and its regulated lipid metabolism, especially PC and LPC metabolism, participate in controlling the axonal degeneration induced by WDR45 deficits. CONCLUSIONS In this study, we uncovered the molecular mechanisms underlying the contribution of WDR45 deficiency to axonal degeneration, which involves complex relationships between phospholipid metabolism, autophagy, and tubular ER. These findings greatly advance our understanding of the fundamental molecular mechanisms driving axonal degeneration and may provide a foundation for developing novel mechanistically based therapeutic interventions for BPAN and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Panpan Wang
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Yaping Shao
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Murad Al-Nusaif
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Jun Zhang
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Huijia Yang
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Yuting Yang
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Kunhyok Kim
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Song Li
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Cong Liu
- Interdisciplinary Research Center On Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research On the Pathogenic Mechanisms of Neurological Diseases, The First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China.
- Institute of Neurology, Sichuan Academy of Medical Science, Sichuan Provincial Hospital, Chengdu, 610072, China.
| |
Collapse
|
10
|
Ray NR, Kunkle BW, Hamilton‐Nelson K, Kurup JT, Rajabli F, Qiao M, Vardarajan BN, Cosacak MI, Kizil C, Jean‐Francois M, Cuccaro M, Reyes‐Dumeyer D, Cantwell L, Kuzma A, Vance JM, Gao S, Hendrie HC, Baiyewu O, Ogunniyi A, Akinyemi RO, Alzheimer's Disease Genetics Consortium, Lee W, Martin ER, Wang L, Beecham GW, Bush WS, Xu W, Jin F, Wang L, Farrer LA, Haines JL, Byrd GS, Schellenberg GD, Mayeux R, Pericak‐Vance MA, Reitz C. Extended genome-wide association study employing the African genome resources panel identifies novel susceptibility loci for Alzheimer's disease in individuals of African ancestry. Alzheimers Dement 2024; 20:5247-5261. [PMID: 38958117 PMCID: PMC11350055 DOI: 10.1002/alz.13880] [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: 09/20/2023] [Revised: 04/03/2024] [Accepted: 04/12/2024] [Indexed: 07/04/2024]
Abstract
INTRODUCTION Despite a two-fold risk, individuals of African ancestry have been underrepresented in Alzheimer's disease (AD) genomics efforts. METHODS Genome-wide association studies (GWAS) of 2,903 AD cases and 6,265 controls of African ancestry. Within-dataset results were meta-analyzed, followed by functional genomics analyses. RESULTS A novel AD-risk locus was identified in MPDZ on chromosome (chr) 9p23 (rs141610415, MAF = 0.002, p = 3.68×10-9). Two additional novel common and nine rare loci were identified with suggestive associations (P < 9×10-7). Comparison of association and linkage disequilibrium (LD) patterns between datasets with higher and lower degrees of African ancestry showed differential association patterns at chr12q23.2 (ASCL1), suggesting that this association is modulated by regional origin of local African ancestry. DISCUSSION These analyses identified novel AD-associated loci in individuals of African ancestry and suggest that degree of African ancestry modulates some associations. Increased sample sets covering as much African genetic diversity as possible will be critical to identify additional loci and deconvolute local genetic ancestry effects. HIGHLIGHTS Genetic ancestry significantly impacts risk of Alzheimer's Disease (AD). Although individuals of African ancestry are twice as likely to develop AD, they are vastly underrepresented in AD genomics studies. The Alzheimer's Disease Genetics Consortium has previously identified 16 common and rare genetic loci associated with AD in African American individuals. The current analyses significantly expand this effort by increasing the sample size and extending ancestral diversity by including populations from continental Africa. Single variant meta-analysis identified a novel genome-wide significant AD-risk locus in individuals of African ancestry at the MPDZ gene, and 11 additional novel loci with suggestive genome-wide significance at p < 9×10-7. Comparison of African American datasets with samples of higher degree of African ancestry demonstrated differing patterns of association and linkage disequilibrium at one of these loci, suggesting that degree and/or geographic origin of African ancestry modulates the effect at this locus. These findings illustrate the importance of increasing number and ancestral diversity of African ancestry samples in AD genomics studies to fully disentangle the genetic architecture underlying AD, and yield more effective ancestry-informed genetic screening tools and therapeutic interventions.
Collapse
Grants
- P30 AG013854 NIA NIH HHS
- International Parkinson Fonds
- P50 MH060451 NIMH NIH HHS
- P30 AG066444 NIA NIH HHS
- R01 AG28786-01A1 North Carolina A&T University
- U01AG46161 NIA NIH HHS
- AG05128 Duke University
- Medical Research Council
- U01AG057659 NIH HHS
- R01 DK131437 NIDDK NIH HHS
- R01 AG022374 NIA NIH HHS
- U19 AG074865 NIA NIH HHS
- P50 AG023501 NIA NIH HHS
- U01 AG046152 NIA NIH HHS
- P30 AG010124 NIA NIH HHS
- U01 HG006375 NHGRI NIH HHS
- Biogen
- U01 AG058654 NIA NIH HHS
- NIMH MH60451 NINDS NIH HHS
- U54 AG052427 NIA NIH HHS
- P30 AG066518 NIA NIH HHS
- UO1 HG004610 Group Health Research Institute
- RC2 AG036528 NIA NIH HHS
- P30 AG028377 NIA NIH HHS
- R01AG048927 NIH HHS
- UO1 HG006375 Group Health Research Institute
- R01 AG22018 Rush University
- U01AG46152 NIA NIH HHS
- P50 AG008671 NIA NIH HHS
- P30 AG10133 Indiana University
- P50 AG005142 NIA NIH HHS
- U01 AG10483 Boston University
- Higher Education Funding Council for England
- R01 AG035137 NIA NIH HHS
- R01 AG009029 NIA NIH HHS
- P50 AG005131 NIA NIH HHS
- P50 AG005128 NIA NIH HHS
- P30 AG010133 NIA NIH HHS
- U24 AG021886 NIA NIH HHS
- R01 AG031581 NIA NIH HHS
- 5R01AG012101 New York University
- R01 AG009956 NIA NIH HHS
- P50 AG016574 NIA NIH HHS
- P50 AG005146 NIA NIH HHS
- U01AG058654 NIH HHS
- AG025688 Emory University
- P30AG10161 NIA NIH HHS
- Alzheimer's Drug Discovery Foundation
- U01 AG061356 NIA NIH HHS
- RC2 AG036650 NIA NIH HHS
- Servier
- Janssen Alzheimer Immunotherapy Research & Development, LLC.
- U01 AG032984 NIA NIH HHS
- U01 HG008657 NHGRI NIH HHS
- Brain Net Europe
- R01 AG019085 NIA NIH HHS
- Lumosity
- R01 AG013616 NIA NIH HHS
- U01 AG024904 NIA NIH HHS
- R01 HG012384 NHGRI NIH HHS
- Translational Genomics Research Institute
- P50 AG008702 NIA NIH HHS
- Bristol-Myers Squibb Company
- R01 AG030146 NIA NIH HHS
- R01AG041797 NIA FBS (Columbia University)
- U01 AG072579 NIA NIH HHS
- Piramal Imaging
- DeNDRoN
- UL1 RR029893 NCRR NIH HHS
- Takeda Pharmaceutical Company
- 1R01AG035137 New York University
- R01 AG15819 Rush University
- R01AG30146 NIA NIH HHS
- R01AG15819 NIA NIH HHS
- P50 NS039764 NINDS NIH HHS
- P01 AG003991 NIA NIH HHS
- Office of Research and Development
- Genentech, Inc.
- U01 AG016976 NIA NIH HHS
- US Department of Veterans Affairs Administration
- P30 AG008051 NIA NIH HHS
- P50 AG005681 NIA NIH HHS
- P30 AG013846 NIA NIH HHS
- U24 AG056270 NIA NIH HHS
- RC2 AG036502 NIA NIH HHS
- P01 AG026276 NIA NIH HHS
- R01 AG017917 NIA NIH HHS
- Araclon Biotech
- U01 AG057659 NIA NIH HHS
- R01 MH080295 NIMH NIH HHS
- Hersenstichting Nederland Breinbrekend Werk
- R01 CA267872 NCI NIH HHS
- R01 AG026390 NIA NIH HHS
- R01 AG028786 NIA NIH HHS
- KL2 RR024151 NCRR NIH HHS
- Internationale Stiching Alzheimer Onderzoek
- P30AG066462 NIH HHS
- U24 AG026390 NIA FBS (Columbia University)
- Novartis Pharmaceuticals Corporation
- P50 AG005136 NIA NIH HHS
- Meso Scale Diagnostics, LLC.
- CereSpir, Inc.
- P30 AG012300 NIA NIH HHS
- P01 AG03991 University of Washington
- RF1AG059018 NIH HHS
- Canadian Institute of Health Research
- RF1 AG059018 NIA NIH HHS
- BioClinica, Inc.
- UG3 NS132061 NINDS NIH HHS
- U01 AG062943 NIA NIH HHS
- R01 AG012101 NIA NIH HHS
- GE Healthcare
- P50 AG016573 NIA NIH HHS
- U24 AG21886 National Cell Repository for Alzheimer's Disease (NCRAD)
- P50 AG016570 NIA NIH HHS
- P50 AG005134 NIA NIH HHS
- P30 AG066462 NIA NIH HHS
- Stichting MS Research
- P30 AG008017 NIA NIH HHS
- R01AG33193 Boston University
- Howard Hughes Medical Institute
- R01 AG042437 NIA NIH HHS
- U24 AG041689 NIA NIH HHS
- P01 AG019724 NIA NIH HHS
- R01AG36042 NIA NIH HHS
- RC2AG036547 NIA NIH HHS
- R01 AG036042 NIA NIH HHS
- P30 AG010161 NIA NIH HHS
- AG019757 University of Miami
- Kronos Science
- P30 AG08051 New York University
- IIRG-05-14147 Alzheimer's Association
- AG010491 University of Miami
- R01 AG033193 NIA NIH HHS
- P50 AG025688 NIA NIH HHS
- IIRG-08-89720 Alzheimer's Association
- AbbVie
- R37 AG015473 NIA NIH HHS
- U24 AG026395 NIA NIH HHS
- R01 AG032990 NIA NIH HHS
- North Bristol NHS Trust Research and Innovation Department
- AG021547 University of Miami
- R01 AG01101 Rush University
- Transition Therapeutics
- R01 AG072547 NIA NIH HHS
- AG027944 University of Miami
- AG041232 NIA NIH HHS
- A2111048 BrightFocus Foundation
- U01 AG052410 NIA NIH HHS
- Johnson & Johnson Pharmaceutical Research & Development LLC.
- R01 CA129769 NCI NIH HHS
- P50 AG005133 NIA NIH HHS
- U01 AG010483 NIA NIH HHS
- UO1 AG006781 Group Health Research Institute
- Merck & Co., Inc.
- U01AG32984 NIA NIH HHS
- U01 AG024904 NIH HHS
- RC2 AG036547 NIA NIH HHS
- P01 AG002219 NIA NIH HHS
- R01 AG17917 Rush University
- U01 AG006781 NIA NIH HHS
- R01 AG041797 NIA NIH HHS
- NIBIB NIH HHS
- P01 AG010491 NIA NIH HHS
- P50 AG005144 NIA NIH HHS
- U01AG062943 NIH HHS
- R01 AG064614 NIA NIH HHS
- Glaxo Smith Kline
- U01AG072579 NIH HHS
- Biomedical Laboratory Research Program
- U19AG074865 NIH HHS
- R01 AG048927 NIA NIH HHS
- RF1 AG057473 NIA NIH HHS
- R01 AG037212 NIA NIH HHS
- R01 AG022018 NIA NIH HHS
- U24AG056270 NIH HHS
- R01 AG021547 NIA NIH HHS
- R01 AG041232 NIA NIH HHS
- P50 AG005138 NIA NIH HHS
- RF1AG57473 NIA NIH HHS
- R01 AG019757 NIA NIH HHS
- R01 AG020688 NIA NIH HHS
- AG07562 University of Pittsburgh
- R01AG072547 NIH HHS
- Alzheimer's Research Trust
- Pfizer Inc.
- Illinois Department of Public Health
- Elan Pharmaceuticals, Inc.
- NHS trusts
- R01 AG030653 NIA NIH HHS
- R01 HG009658 NHGRI NIH HHS
- AG052410 NIA NIH HHS
- P20 MD000546 NIMHD NIH HHS
- R01 AG027944 NIA NIH HHS
- Eli Lilly and Company
- R01 AG017173 NIA NIH HHS
- R01 AG025259 NIA NIH HHS
- U01 HG004610 NHGRI NIH HHS
- U24-AG041689 University of Pennsylvania
- P30 AG010129 NIA NIH HHS
- U01 AG046161 NIA NIH HHS
- Wellcome Trust
- P30 AG019610 NIA NIH HHS
- IXICO Ltd.
- P50 AG016582 NIA NIH HHS
- R01 AG048015 NIA NIH HHS
- NeuroRx Research
- R01AG17917 NIA NIH HHS
- U01AG61356 NIA NIH HHS
- R01AG36836 NIA NIH HHS
- 5R01AG022374 New York University
- EuroImmun; F. Hoffmann-La Roche Ltd
- R01 AG041718 NIA NIH HHS
- 1RC2AG036502 New York University
- Newcastle University
- R01 AG072474 NIA NIH HHS
- AG041718 University of Pittsburgh
- P30 AG028383 NIA NIH HHS
- AG05144 University of Kentucky
- AG030653 University of Pittsburgh
- R01AG48015 NIA NIH HHS
- R01 AG026916 NIA NIH HHS
- P50 AG033514 NIA NIH HHS
- R01 NS059873 NINDS NIH HHS
- # NS39764 NINDS NIH HHS
- ADGC National Institutes of Health, National Institute on Aging (NIH-NIA)
- Neurotrack Technologies
- Fujirebio
- Lundbeck
- MP-V BrightFocus Foundation
- BRACE
- R01 AG015819 NIA NIH HHS
- R01 AG036836 NIA NIH HHS
- Eisai Inc.
- 5R01AG013616 New York University
- W81XWH-12-2-0012 Department of Defense
- R01AG064614 NIH HHS
- AG02365 University of Pittsburgh
- NIH
- University of Pennsylvania
- NACC
- Boston University
- Columbia University
- Duke University
- Emory University
- Indiana University
- Johns Hopkins University
- Massachusetts General Hospital
- Mayo Clinic
- New York University
- Northwestern University
- Oregon Health & Science University
- Rush University
- NIA
- University of Alabama at Birmingham
- University of Arizona
- University of California, Davis
- University of California, Irvine
- University of California, Los Angeles
- University of California, San Diego
- University of California, San Francisco
- University of Kentucky
- University of Michigan
- University of Pittsburgh
- University of Southern California
- University of Miami
- University of Washington
- Vanderbilt University
- NINDS
- Alzheimer's Association
- Office of Research and Development
- BrightFocus Foundation
- Wellcome Trust
- Howard Hughes Medical Institute
- Medical Research Council
- Newcastle University
- Higher Education Funding Council for England
- Alzheimer's Research Trust
- BRACE
- Stichting MS Research
- Department of Defense
- National Institute of Biomedical Imaging and Bioengineering
- AbbVie
- Alzheimer's Drug Discovery Foundation
- BioClinica, Inc.
- Biogen
- Bristol‐Myers Squibb Company
- Eli Lilly and Company
- Genentech, Inc.
- Fujirebio
- GE Healthcare
- Lundbeck
- Merck & Co., Inc.
- Novartis Pharmaceuticals Corporation
- Pfizer Inc.
- Servier
- Takeda Pharmaceutical Company
- Illinois Department of Public Health
- Translational Genomics Research Institute
Collapse
|
11
|
Wang L, Yang Z, Satoshi F, Prasanna X, Yan Z, Vihinen H, Chen Y, Zhao Y, He X, Bu Q, Li H, Zhao Y, Jiang L, Qin F, Dai Y, Zhang N, Qin M, Kuang W, Zhao Y, Jokitalo E, Vattulainen I, Kajander T, Zhao H, Cen X. Membrane remodeling by FAM92A1 during brain development regulates neuronal morphology, synaptic function, and cognition. Nat Commun 2024; 15:6209. [PMID: 39043703 PMCID: PMC11266426 DOI: 10.1038/s41467-024-50565-w] [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: 09/18/2023] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
The Bin/Amphiphysin/Rvs (BAR) domain protein FAM92A1 is a multifunctional protein engaged in regulating mitochondrial ultrastructure and ciliogenesis, but its physiological role in the brain remains unclear. Here, we show that FAM92A1 is expressed in neurons starting from embryonic development. FAM92A1 knockout in mice results in altered brain morphology and age-associated cognitive deficits, potentially due to neuronal degeneration and disrupted synaptic plasticity. Specifically, FAM92A1 deficiency impairs diverse neuronal membrane morphology, including the mitochondrial inner membrane, myelin sheath, and synapses, indicating its roles in membrane remodeling and maintenance. By determining the crystal structure of the FAM92A1 BAR domain, combined with atomistic molecular dynamics simulations, we uncover that FAM92A1 interacts with phosphoinositide- and cardiolipin-containing membranes to induce lipid-clustering and membrane curvature. Altogether, these findings reveal the physiological role of FAM92A1 in the brain, highlighting its impact on synaptic plasticity and neural function through the regulation of membrane remodeling and endocytic processes.
Collapse
Affiliation(s)
- Liang Wang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Ziyun Yang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Fudo Satoshi
- Helsinki Institute of Life Science - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Xavier Prasanna
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Ziyi Yan
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland
| | - Helena Vihinen
- Helsinki Institute of Life Science (HiLIFE) - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Yaxing Chen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yue Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xiumei He
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, 541004, China
| | - Qian Bu
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Hongchun Li
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ying Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Linhong Jiang
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Feng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Yanping Dai
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Ni Zhang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Meng Qin
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Weihong Kuang
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yinglan Zhao
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Eija Jokitalo
- Helsinki Institute of Life Science (HiLIFE) - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Tommi Kajander
- Helsinki Institute of Life Science - Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Hongxia Zhao
- Faculty of Biological and Environmental Sciences, University of Helsinki, 00014, Helsinki, Finland.
- School of Life Sciences, Guangxi Normal University, Guilin, China.
- Guangxi Universities Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, 541004, China.
| | - Xiaobo Cen
- Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
12
|
Yu K, Yao KR, Aguinaga MA, Choquette JM, Liu C, Wang Y, Liao D. G272V and P301L Mutations Induce Isoform Specific Tau Mislocalization to Dendritic Spines and Synaptic Dysfunctions in Cellular Models of 3R and 4R Tau Frontotemporal Dementia. J Neurosci 2024; 44:e1215232024. [PMID: 38858079 PMCID: PMC11236579 DOI: 10.1523/jneurosci.1215-23.2024] [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: 06/27/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 06/12/2024] Open
Abstract
Tau pathologies are detected in the brains of some of the most common neurodegenerative diseases including Alzheimer's disease (AD), Lewy body dementia (LBD), chronic traumatic encephalopathy (CTE), and frontotemporal dementia (FTD). Tau proteins are expressed in six isoforms with either three or four microtubule-binding repeats (3R tau or 4R tau) due to alternative RNA splicing. AD, LBD, and CTE brains contain pathological deposits of both 3R and 4R tau. FTD patients can exhibit either 4R tau pathologies in most cases or 3R tau pathologies less commonly in Pick's disease, which is a subfamily of FTD. Here, we report the isoform-specific roles of tau in FTD. The P301L mutation, linked to familial 4R tau FTD, induces mislocalization of 4R tau to dendritic spines in primary hippocampal cultures that were prepared from neonatal rat pups of both sexes. Contrastingly, the G272V mutation, linked to familial Pick's disease, induces phosphorylation-dependent mislocalization of 3R tau but not 4R tau proteins to dendritic spines. The overexpression of G272V 3R tau but not 4R tau proteins leads to the reduction of dendritic spine density and suppression of mEPSCs in 5-week-old primary rat hippocampal cultures. The decrease in mEPSC amplitude caused by G272V 3R tau is dynamin-dependent whereas that caused by P301L 4R tau is dynamin-independent, indicating that the two tau isoforms activate different signaling pathways responsible for excitatory synaptic dysfunction. Our 3R and 4R tau studies here will shed new light on diverse mechanisms underlying FTD, AD, LBD, and CTE.
Collapse
Affiliation(s)
- Ke Yu
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Department of General Practice, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Katherine R Yao
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- College of Biological Sciences, University of Minnesota, St Paul, Minnesota 55108
| | - Miguel A Aguinaga
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- College of Biological Sciences, University of Minnesota, St Paul, Minnesota 55108
| | - Jessica M Choquette
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Chengliang Liu
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Yuxin Wang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Dezhi Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
13
|
Tang Y, Wu X, Li J, Li Y, Xu X, Li G, Zhang P, Qin C, Wu LJ, Tang Z, Tian DS. The Emerging Role of Microglial Hv1 as a Target for Immunomodulation in Myelin Repair. Aging Dis 2024; 15:1176-1203. [PMID: 38029392 PMCID: PMC11081154 DOI: 10.14336/ad.2023.1107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
In the central nervous system (CNS), the myelin sheath ensures efficient interconnection between neurons and contributes to the regulation of the proper function of neuronal networks. The maintenance of myelin and the well-organized subtle process of myelin plasticity requires cooperation among myelin-forming cells, glial cells, and neural networks. The process of cooperation is fragile, and the balance is highly susceptible to disruption by microenvironment influences. Reactive microglia play a critical and complicated role in the demyelination and remyelination process. Recent studies have shown that the voltage-gated proton channel Hv1 is selectively expressed in microglia in CNS, which regulates intracellular pH and is involved in the production of reactive oxygen species, underlying multifaceted roles in maintaining microglia function. This paper begins by examining the molecular mechanisms of demyelination and emphasizes the crucial role of the microenvironment in demyelination. It focuses specifically on the role of Hv1 in myelin repair and its therapeutic potential in CNS demyelinating diseases.
Collapse
Affiliation(s)
- Yingxin Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xuan Wu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Jiarui Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Yuanwei Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoxiao Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Gaigai Li
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ping Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Zhouping Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
Bamford RA, Zuko A, Eve M, Sprengers JJ, Post H, Taggenbrock RLRE, Fäβler D, Mehr A, Jones OJR, Kudzinskas A, Gandawijaya J, Müller UC, Kas MJH, Burbach JPH, Oguro-Ando A. CNTN4 modulates neural elongation through interplay with APP. Open Biol 2024; 14:240018. [PMID: 38745463 PMCID: PMC11293442 DOI: 10.1098/rsob.240018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 05/16/2024] Open
Abstract
The neuronal cell adhesion molecule contactin-4 (CNTN4) is genetically associated with autism spectrum disorder (ASD) and other psychiatric disorders. Cntn4-deficient mouse models have previously shown that CNTN4 plays important roles in axon guidance and synaptic plasticity in the hippocampus. However, the pathogenesis and functional role of CNTN4 in the cortex has not yet been investigated. Our study found a reduction in cortical thickness in the motor cortex of Cntn4 -/- mice, but cortical cell migration and differentiation were unaffected. Significant morphological changes were observed in neurons in the M1 region of the motor cortex, indicating that CNTN4 is also involved in the morphology and spine density of neurons in the motor cortex. Furthermore, mass spectrometry analysis identified an interaction partner for CNTN4, confirming an interaction between CNTN4 and amyloid-precursor protein (APP). Knockout human cells for CNTN4 and/or APP revealed a relationship between CNTN4 and APP. This study demonstrates that CNTN4 contributes to cortical development and that binding and interplay with APP controls neural elongation. This is an important finding for understanding the physiological function of APP, a key protein for Alzheimer's disease. The binding between CNTN4 and APP, which is involved in neurodevelopment, is essential for healthy nerve outgrowth.
Collapse
Affiliation(s)
- Rosemary A. Bamford
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Amila Zuko
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Madeline Eve
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Jan J. Sprengers
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht, Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Netherlands Proteomics Centre, Utrecht, The Netherlands
| | - Renske L. R. E. Taggenbrock
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Dominique Fäβler
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Annika Mehr
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Owen J. R. Jones
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Aurimas Kudzinskas
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Josan Gandawijaya
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
| | - Ulrike C. Müller
- Institute for Pharmacy and Molecular Biotechnology (IPMB), Functional Genomics, University of Heidelberg, Heidelberg69120, Germany
| | - Martien J. H. Kas
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - J. Peter H. Burbach
- Department of Translational Neuroscience, UMC Utrecht Brain Center, UMC Utrecht, Utrecht3508 AB, The Netherlands
| | - Asami Oguro-Ando
- University of Exeter Medical School, University of Exeter, ExeterEX2 5DW, UK
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
- Research Institute for Science and Technology, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
15
|
Zhou H, Bi GQ, Liu G. Intracellular magnesium optimizes transmission efficiency and plasticity of hippocampal synapses by reconfiguring their connectivity. Nat Commun 2024; 15:3406. [PMID: 38649706 PMCID: PMC11035601 DOI: 10.1038/s41467-024-47571-3] [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: 07/24/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Synapses at dendritic branches exhibit specific properties for information processing. However, how the synapses are orchestrated to dynamically modify their properties, thus optimizing information processing, remains elusive. Here, we observed at hippocampal dendritic branches diverse configurations of synaptic connectivity, two extremes of which are characterized by low transmission efficiency, high plasticity and coding capacity, or inversely. The former favors information encoding, pertinent to learning, while the latter prefers information storage, relevant to memory. Presynaptic intracellular Mg2+ crucially mediates the dynamic transition continuously between the two extreme configurations. Consequently, varying intracellular Mg2+ levels endow individual branches with diverse synaptic computations, thus modulating their ability to process information. Notably, elevating brain Mg2+ levels in aging animals restores synaptic configuration resembling that of young animals, coincident with improved learning and memory. These findings establish intracellular Mg2+ as a crucial factor reconfiguring synaptic connectivity at dendrites, thus optimizing their branch-specific properties in information processing.
Collapse
Affiliation(s)
- Hang Zhou
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China.
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Guo-Qiang Bi
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology, Shenzhen, 518107, China
- Interdisciplinary Center for Brain Information, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, 518055, China
- Hefei National Laboratory for Physical Sciences at the Microscale, and School of Life Sciences, University of Science and Technology of China, Hefei, 230031, China
| | - Guosong Liu
- School of Medicine, Tsinghua University, Beijing, 100084, China.
- NeuroCentria Inc., Walnut Creek, CA, 94596, USA.
| |
Collapse
|
16
|
Karbowski J, Urban P. Cooperativity, Information Gain, and Energy Cost During Early LTP in Dendritic Spines. Neural Comput 2024; 36:271-311. [PMID: 38101326 DOI: 10.1162/neco_a_01632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 10/04/2023] [Indexed: 12/17/2023]
Abstract
We investigate a mutual relationship between information and energy during the early phase of LTP induction and maintenance in a large-scale system of mutually coupled dendritic spines, with discrete internal states and probabilistic dynamics, within the framework of nonequilibrium stochastic thermodynamics. In order to analyze this computationally intractable stochastic multidimensional system, we introduce a pair approximation, which allows us to reduce the spine dynamics into a lower-dimensional manageable system of closed equations. We found that the rates of information gain and energy attain their maximal values during an initial period of LTP (i.e., during stimulation), and after that, they recover to their baseline low values, as opposed to a memory trace that lasts much longer. This suggests that the learning phase is much more energy demanding than the memory phase. We show that positive correlations between neighboring spines increase both a duration of memory trace and energy cost during LTP, but the memory time per invested energy increases dramatically for very strong, positive synaptic cooperativity, suggesting a beneficial role of synaptic clustering on memory duration. In contrast, information gain after LTP is the largest for negative correlations, and energy efficiency of that information generally declines with increasing synaptic cooperativity. We also find that dendritic spines can use sparse representations for encoding long-term information, as both energetic and structural efficiencies of retained information and its lifetime exhibit maxima for low fractions of stimulated synapses during LTP. Moreover, we find that such efficiencies drop significantly with increasing the number of spines. In general, our stochastic thermodynamics approach provides a unifying framework for studying, from first principles, information encoding, and its energy cost during learning and memory in stochastic systems of interacting synapses.
Collapse
Affiliation(s)
- Jan Karbowski
- Institute of Applied Mathematics and Mechanics, University of Warsaw, Warsaw 02-097, Poland
| | - Paulina Urban
- College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences and Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw 02-097, Poland
- Laboratory of Databases and Business Analytics, National Information Processing Institute, National Research Institute, Warsaw 00-608, Poland
| |
Collapse
|
17
|
Ito H, Morishita R, Nagata KI. Simple Method for the Preparation of Postsynaptic Density Fraction from Mouse Brain. Methods Mol Biol 2024; 2794:71-78. [PMID: 38630221 DOI: 10.1007/978-1-0716-3810-1_7] [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] [Indexed: 04/19/2024]
Abstract
Postsynaptic density (PSD) is a morphologically and functionally specialized postsynaptic membrane structure of excitatory synapses. It contains hundreds of proteins such as neurotransmitter receptors, adhesion molecules, cytoskeletal proteins, and signaling enzymes. The study of the molecular architecture of the PSD is one of the most intriguing issues in neuroscience research. The isolation of the PSD from the brain of an animal is necessary for subsequent biochemical and morphological analyses. Many laboratories have developed methods to isolate PSD from the animal brain. In this chapter, we present a simple method to isolate PSD from the mouse brain using sucrose density gradient-based purification of synaptosomes followed by detergent extraction.
Collapse
Affiliation(s)
- Hidenori Ito
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Aichi, Japan.
| | - Rika Morishita
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Aichi, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Developmental Disability Center, Aichi, Japan
- Department of Neurochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
18
|
Wang L, Lu Z, Teng Y, Pan W, Li Y, Su S, Chang J, Zhao M. Cognitive impairment is associated with BDNF-TrkB signaling mediating synaptic damage and reduction of amino acid neurotransmitters in heart failure. FASEB J 2024; 38:e23351. [PMID: 38085181 DOI: 10.1096/fj.202301699rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023]
Abstract
Heart failure (HF) is often accompanied by cognitive impairment (CI). Brain-derived neurotrophic factor (BDNF) deficiency is closely associated with CI. However, the role and mechanism of BDNF in HF with CI is still not fully understood. Here, the case-control study was designed including 25 HF without CI patients (HF-NCI) and 50 HF with CI patients (HF-CI) to investigate the predictive value of BDNF in HF-CI while animal and cell experiments were used for mechanism research. Results found that BDNF levels in serum neuronal-derived exosomes were downregulated in HF-CI patients. There was no significant difference in serum BDNF levels among the two groups. HF rats showed obvious impairment in learning and memory; also, they had reduced thickness and length of postsynaptic density (PSD) and increased synaptic cleft width. Expression of BDNF, TrkB, PSD95, and VGLUT1 was significantly decreased in HF rats brain. In addition, compared with sham rats, amino acids were significantly reduced with no changes in the acetylcholine and monoamine neurotransmitters. Further examination showed that the number of synaptic bifurcations and the expression of BDNF, TrkB, PSD95, and VGLUT1 were all decreased in the neurons that interfered with BDNF-siRNA compared with those in the negative control neurons. Together, our results demonstrated that neuronal-derived exosomal BDNF act as effective biomarkers for prediction of HF-CI. The decrease of BDNF in the brain triggers synaptic structural damage and a decline in amino acid neurotransmitters via the BDNF-TrkB-PSD95/VGLUT1 pathway. This discovery unveils a novel pathological mechanism underlying cognitive impairment following heart failure.
Collapse
Affiliation(s)
- Lei Wang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ziwen Lu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Teng
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Weibing Pan
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Sha Su
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jingling Chang
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingjing Zhao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
19
|
Karbowski J, Urban P. Information encoded in volumes and areas of dendritic spines is nearly maximal across mammalian brains. Sci Rep 2023; 13:22207. [PMID: 38097675 PMCID: PMC10721930 DOI: 10.1038/s41598-023-49321-9] [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: 09/05/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
Many experiments suggest that long-term information associated with neuronal memory resides collectively in dendritic spines. However, spines can have a limited size due to metabolic and neuroanatomical constraints, which should effectively limit the amount of encoded information in excitatory synapses. This study investigates how much information can be stored in the population of sizes of dendritic spines, and whether it is optimal in any sense. It is shown here, using empirical data for several mammalian brains across different regions and physiological conditions, that dendritic spines nearly maximize entropy contained in their volumes and surface areas for a given mean size in cortical and hippocampal regions. Although both short- and heavy-tailed fitting distributions approach [Formula: see text] of maximal entropy in the majority of cases, the best maximization is obtained primarily for short-tailed gamma distribution. We find that most empirical ratios of standard deviation to mean for spine volumes and areas are in the range [Formula: see text], which is close to the theoretical optimal ratios coming from entropy maximization for gamma and lognormal distributions. On average, the highest entropy is contained in spine length ([Formula: see text] bits per spine), and the lowest in spine volume and area ([Formula: see text] bits), although the latter two are closer to optimality. In contrast, we find that entropy density (entropy per spine size) is always suboptimal. Our results suggest that spine sizes are almost as random as possible given the constraint on their size, and moreover the general principle of entropy maximization is applicable and potentially useful to information and memory storing in the population of cortical and hippocampal excitatory synapses, and to predicting their morphological properties.
Collapse
Affiliation(s)
- Jan Karbowski
- Institute of Applied Mathematics and Mechanics, University of Warsaw, Warsaw, Poland.
| | - Paulina Urban
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
- College of Inter-Faculty Individual Studies in Mathematics and Natural Sciences, University of Warsaw, Warsaw, Poland
- Laboratory of Databases and Business Analytics, National Information Processing Institute, National Research Institute, Warsaw, Poland
| |
Collapse
|
20
|
Yan T, Chen J, Wang Y, Wang Y, Zhang Y, Zhao Y. Deficiency of aldehyde dehydrogenase 2 aggravates ethanol-induced cytotoxicity in N2a cells via CaMKII/Drp1-mediated mitophagy. Food Chem Toxicol 2023; 182:114129. [PMID: 37967785 DOI: 10.1016/j.fct.2023.114129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 10/13/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
Chronic alcohol abuse causes brain damage and has been associated with an increased risk of Alzheimer's disease. The toxic metabolite of alcohol, acetaldehyde, which is converted to acetate by aldehyde dehydrogenase 2 (ALDH2), has been shown to induce excessive mitochondrial fragmentation and dysfunction leading to neurotoxicity. However, it is still unclear how alcohol affects mitochondrial function in ALDH2-deficient cells. The present study investigated the association between abnormal mitochondrial dynamics, mitophagy and cytotoxicity in ALDH2-deficient N2a cells treated with ethanol. It was found that ethanol induced dynamin-related protein 1 (Drp1)-mediated mitochondrial fragmentation and impaired mitochondrial function, causing excessive mitophagy and cytotoxicity in ALDH2-deficient N2a cells while inducing Ca2+ influx and activating Ca2+/calmodulin-dependent protein kinase II (CaMKII). Inhibition of Ca2+ overload or CaMKII activation prevented Drp1 phosphorylation and ameliorated ethanol-induced mitophagy and cytotoxicity, indicating that Ca2+-dependent CaMKII activation was critical for mediating Drp1-dependent excessive mitochondrial fission and mitophagy in ALDH2-deficient N2a cells. The results of the present study suggested that prevention of intracellular Ca2+ overload might be beneficial for preventing neurotoxicity associated with alcohol abuse in individuals with defective ALDH2.
Collapse
Affiliation(s)
- Tingting Yan
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Jiyang Chen
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Yalin Wang
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Yinuo Wang
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Yuanqingzhi Zhang
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China
| | - Yan Zhao
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, Shandong, China.
| |
Collapse
|
21
|
Clarkson-Paredes C, Karl MT, Popratiloff A, Miller RH. A unique cell population expressing the Epithelial-Mesenchymal Transition-transcription factor Snail moderates microglial and astrocyte injury responses. PNAS NEXUS 2023; 2:pgad334. [PMID: 37901440 PMCID: PMC10612478 DOI: 10.1093/pnasnexus/pgad334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/26/2023] [Indexed: 10/31/2023]
Abstract
Insults to the central nervous system (CNS) elicit common glial responses including microglial activation evidenced by functional, morphological, and phenotypic changes, as well as astrocyte reactions including hypertrophy, altered process orientation, and changes in gene expression and function. However, the cellular and molecular mechanisms that initiate and modulate such glial response are less well-defined. Here we show that an adult cortical lesion generates a population of ultrastructurally unique microglial-like cells that express Epithelial-Mesenchymal Transcription factors including Snail. Knockdown of Snail with antisense oligonucleotides results in a postinjury increase in activated microglial cells, elevation in astrocyte reactivity with increased expression of C3 and phagocytosis, disruption of astrocyte junctions and neurovascular structure, increases in neuronal cell death, and reduction in cortical synapses. These changes were associated with alterations in pro-inflammatory cytokine expression. By contrast, overexpression of Snail through microglia-targeted an adeno-associated virus (AAV) improved many of the injury characteristics. Together, our results suggest that the coordination of glial responses to CNS injury is partly mediated by epithelial-mesenchymal transition-factors (EMT-Fsl).
Collapse
Affiliation(s)
- Cheryl Clarkson-Paredes
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 Eye Street NW, Ross 735, Washington, DC 20052, USA
- Nanofabrication and Imaging Center, The George Washington University, 800 22nd Street NW, Washington, DC 20052, USA
| | - Molly T Karl
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 Eye Street NW, Ross 735, Washington, DC 20052, USA
| | - Anastas Popratiloff
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 Eye Street NW, Ross 735, Washington, DC 20052, USA
- Nanofabrication and Imaging Center, The George Washington University, 800 22nd Street NW, Washington, DC 20052, USA
| | - Robert H Miller
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, George Washington University, 2300 Eye Street NW, Ross 735, Washington, DC 20052, USA
| |
Collapse
|
22
|
Huang C, Wang R, Wang Y, Liu H, Chen XT, Gu X, Wang HL. Sialic Acid Enhanced the Antistress Capability under Challenging Situations by Increasing Synaptic Transmission. J Nutr 2023; 153:2561-2570. [PMID: 37543214 DOI: 10.1016/j.tjnut.2023.08.006] [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: 05/08/2023] [Revised: 07/24/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND In early life, sialic acid (SA) plays a crucial role in neurodevelopment and neuronal function. However, it remains unclear whether and how SA supplementation in early life promotes behavioral response to stress in adolescence. OBJECTIVES This study aimed to examine the effects and mechanisms of SA on the antistress capability under challenging situations. METHODS In this study, C57BL/6 mice were daily supplemented with 1 μL SA solution/g body weight at the dose of 10 mg/kg/d from postnatal day (PND) 5-45. The antistress behaviors, including open field, elevated plus maze, forced swimming test, and tail suspension test, were performed at PND 46, PND 48, PND 50, and PND 52 to detect the antistress ability of SA, respectively. RESULTS Our results showed that SA-treated mice were more active in facing challenging situations. The fiber photometry experiment showed that SA promoted the excitatory neuronal response in the medial prefrontal cortex (mPFC), which was extensively interconnected to stress. Besides, electrophysiological results revealed SA enhanced synaptic transmission rather than neuronal excitability of mPFC excitatory neurons. It was also supported by the increasing spine density of mPFC excitatory neurons. At the molecular amount, the SA elevated the transmitter release-related proteins of mPFC, including Synapsin 1 and vesicular glutamate transporter 1 (VGlut 1). Furthermore, SA supplementation enhanced synaptic transmission mainly by altering the kinetics of synaptic transmission. CONCLUSIONS The SA supplementation enhanced the response capability to stress under challenging situations, and the enhanced synaptic transmission of mPFC excitatory neurons may be the neurological basis of active response under challenging situations. In general, our findings suggested that SA supplementation in early life can promote stress resistance in adolescence.
Collapse
Affiliation(s)
- Chengqing Huang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, People's Republic of China
| | - Rongrong Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, People's Republic of China
| | - Yi Wang
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Haoyu Liu
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Xiang-Tao Chen
- School of Pharmacy, Anhui Medical University, Hefei, Anhui, People's Republic of China
| | - Xiaozhen Gu
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, People's Republic of China.
| | - Hui-Li Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei, Anhui, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, People's Republic of China.
| |
Collapse
|
23
|
Ray NR, Kunkle BW, Hamilton-Nelson K, Kurup JT, Rajabli F, Cosacak MI, Kizil C, Jean-Francois M, Cuccaro M, Reyes-Dumeyer D, Cantwell L, Kuzma A, Vance JM, Gao S, Hendrie HC, Baiyewu O, Ogunniyi A, Akinyemi RO, Lee WP, Martin ER, Wang LS, Beecham GW, Bush WS, Farrer LA, Haines JL, Byrd GS, Schellenberg GD, Mayeux R, Pericak-Vance MA, Reitz C. Extended genome-wide association study employing the African Genome Resources Panel identifies novel susceptibility loci for Alzheimer's Disease in individuals of African ancestry. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.08.29.23294774. [PMID: 37693582 PMCID: PMC10491365 DOI: 10.1101/2023.08.29.23294774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
INTRODUCTION Despite a two-fold increased risk, individuals of African ancestry have been significantly underrepresented in Alzheimer's Disease (AD) genomics efforts. METHODS GWAS of 2,903 AD cases and 6,265 cognitive controls of African ancestry. Within-dataset results were meta-analyzed, followed by gene-based and pathway analyses, and analysis of RNAseq and whole-genome sequencing data. RESULTS A novel AD risk locus was identified in MPDZ on chromosome 9p23 (rs141610415, MAF=.002, P =3.68×10 -9 ). Two additional novel common and nine novel rare loci approached genome-wide significance at P <9×10 -7 . Comparison of association and LD patterns between datasets with higher and lower degrees of African ancestry showed differential association patterns at chr12q23.2 ( ASCL1 ), suggesting that the association is modulated by regional origin of local African ancestry. DISCUSSION Increased sample sizes and sample sets from Africa covering as much African genetic diversity as possible will be critical to identify additional disease-associated loci and improve deconvolution of local genetic ancestry effects.
Collapse
|
24
|
Le W, Wang P, Al-Nusaif M, Zhang J, Yang H, Yang Y, Kim K, Li S, Liu C, Cai H. Pathological characteristics of axons and proteome patterns in midbrain dopaminergic neurodegeneration induced by WDR45-deficiency. RESEARCH SQUARE 2023:rs.3.rs-2901370. [PMID: 37292937 PMCID: PMC10246098 DOI: 10.21203/rs.3.rs-2901370/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Although WD repeats domain 45 (WDR45) mutations have been linked to β-propeller protein-associated neurodegeneration (BPAN), the precise molecular and cellular mechanisms behind this disease remain elusive. This study aims to shed light on the effects of WDR45-deficiency on neurodegeneration, specifically axonal degeneration, within the midbrain dopaminergic (DAergic) system. By examining pathological and molecular alterations, we hope to better understand the disease process. Methods To investigate the effects of WDR45 dysfunction on mouse behaviors and DAergic neurons, we developed a mouse model in which WDR45 was conditionally knocked out in midbrain DAergic neurons (WDR45cKO). Through a longitudinal study, we assessed alterations in mouse behavior using open field, rotarod, Y-maze, and 3-chamber social approach tests. To examine the pathological changes in DAergic neuron soma and axons, we utilized a combination of immunofluorescence staining and transmission electron microscopy. Additionally, we performed proteomic analyses of the striatum to identify the molecules and processes involved in striatal pathology. Results Our study of WDR45cKO mice revealed a range of deficits, including impaired motor function, emotional instability, and memory loss, coinciding with the profound loss of midbrain DAergic neurons. Prior to neuronal loss, we observed massive axonal enlargements in both the dorsal and ventral striatum. These enlargements were characterized by the accumulation of extensively fragmented tubular endoplasmic reticulum (ER), a hallmark of axonal degeneration. Additionally, we found that WDR45cKO mice exhibited disrupted autophagic flux. Proteomic analysis of the striatum in these mice showed that many differentially expressed proteins (DEPs) were enriched in amino acid, lipid, and tricarboxylic acid metabolisms. Of note, we observed significant alterations in the expression of genes encoding DEPs that regulate phospholipids catabolic and biosynthetic processes, such as lysophosphatidylcholine acyltransferase 1, ethanolamine-phosphate phospho-lyase, and abhydrolase domain containing 4, N-acyl phospholipase B. These findings suggest a possible link between phospholipid metabolism and striatal axon degeneration. Conclusions In this study, we have uncovered the molecular mechanisms underlying the contribution of WDR45-deficiency to axonal degeneration, revealing intricate relationships between tubular ER dysfunction, phospholipid metabolism, BPAN and other neurodegenerative diseases. These findings significantly advance our understanding of the fundamental molecular mechanisms driving neurodegeneration and may provide a foundation for developing novel, mechanistically-based therapeutic interventions.
Collapse
Affiliation(s)
- Weidong Le
- The First Affiliated Hospital Of Dalian Medical University
| | - Panpan Wang
- First Affiliated Hospital of Dalian Medical University
| | | | - Jun Zhang
- First Affiliated Hospital of Dalian Medical University
| | - Huijia Yang
- First Affiliated Hospital of Dalian Medical University
| | - Yuting Yang
- First Affiliated Hospital of Dalian Medical University
| | - Kunhyok Kim
- First Affiliated Hospital of Dalian Medical University
| | - Song Li
- First Affiliated Hospital of Dalian Medical University
| | - Cong Liu
- Shanghai Institute of Organic Chemistry
| | | |
Collapse
|
25
|
McCarthy CI, Mustafá ER, Cornejo MP, Yaneff A, Rodríguez SS, Perello M, Raingo J. Chlorpromazine, an Inverse Agonist of D1R-Like, Differentially Targets Voltage-Gated Calcium Channel (Ca V) Subtypes in mPFC Neurons. Mol Neurobiol 2023; 60:2644-2660. [PMID: 36694048 DOI: 10.1007/s12035-023-03221-1] [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: 07/15/2022] [Accepted: 01/04/2023] [Indexed: 01/26/2023]
Abstract
The dopamine receptor type 1 (D1R) and the dopamine receptor type 5 (D5R), which are often grouped as D1R-like due to their sequence and signaling similarities, exhibit high levels of constitutive activity. The molecular basis for this agonist-independent activation has been well characterized through biochemical and mutagenesis in vitro studies. In this regard, it was reported that many antipsychotic drugs act as inverse agonists of D1R-like constitutive activity. On the other hand, D1R is highly expressed in the medial prefrontal cortex (mPFC), a brain area with important functions such as working memory. Here, we studied the impact of D1R-like constitutive activity and chlorpromazine (CPZ), an antipsychotic drug and D1R-like inverse agonist, on various neuronal CaV conductances, and we explored its effect on calcium-dependent neuronal functions in the mouse medial mPFC. Using ex vivo brain slices containing the mPFC and transfected HEK293T cells, we found that CPZ reduces CaV2.2 currents by occluding D1R-like constitutive activity, in agreement with a mechanism previously reported by our lab, whereas CPZ directly inhibits CaV1 currents in a D1R-like activity independent manner. In contrast, CPZ and D1R constitutive activity did not affect CaV2.1, CaV2.3, or CaV3 currents. Finally, we found that CPZ reduces excitatory postsynaptic responses in mPFC neurons. Our results contribute to understanding CPZ molecular targets in neurons and describe a novel physiological consequence of CPZ non-canonical action as a D1R-like inverse agonist in the mouse brain.
Collapse
Affiliation(s)
- Clara Inés McCarthy
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology (Argentine Research Council CONICET, Scientific Research Commission of the Buenos Aires Province and National University of La Plata), La Plata, Buenos Aires, Argentina
| | - Emilio Román Mustafá
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology (Argentine Research Council CONICET, Scientific Research Commission of the Buenos Aires Province and National University of La Plata), La Plata, Buenos Aires, Argentina
| | - María Paula Cornejo
- Neurophysiology Laboratory of the Multidisciplinary Institute of Cell Biology (Argentine Research Council CONICET, Scientific Research Commission of the Buenos Aires Province and National University of La Plata), La Plata, Buenos Aires, Argentina
| | - Agustín Yaneff
- Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvia Susana Rodríguez
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology (Argentine Research Council CONICET, Scientific Research Commission of the Buenos Aires Province and National University of La Plata), La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Neurophysiology Laboratory of the Multidisciplinary Institute of Cell Biology (Argentine Research Council CONICET, Scientific Research Commission of the Buenos Aires Province and National University of La Plata), La Plata, Buenos Aires, Argentina
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, University of Uppsala, Uppsala, Sweden
| | - Jesica Raingo
- Electrophysiology Laboratory of the Multidisciplinary Institute of Cell Biology (Argentine Research Council CONICET, Scientific Research Commission of the Buenos Aires Province and National University of La Plata), La Plata, Buenos Aires, Argentina.
| |
Collapse
|
26
|
Wang Y, Lin J, Li J, Yan L, Li W, He X, Ma H. Chronic Neuronal Inactivity Utilizes the mTOR-TFEB Pathway to Drive Transcription-Dependent Autophagy for Homeostatic Up-Scaling. J Neurosci 2023; 43:2631-2652. [PMID: 36868861 PMCID: PMC10089247 DOI: 10.1523/jneurosci.0146-23.2023] [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: 01/24/2023] [Revised: 02/16/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023] Open
Abstract
Activity-dependent changes in protein expression are critical for neuronal plasticity, a fundamental process for the processing and storage of information in the brain. Among the various forms of plasticity, homeostatic synaptic up-scaling is unique in that it is induced primarily by neuronal inactivity. However, precisely how the turnover of synaptic proteins occurs in this homeostatic process remains unclear. Here, we report that chronically inhibiting neuronal activity in primary cortical neurons prepared from embryonic day (E)18 Sprague Dawley rats (both sexes) induces autophagy, thereby regulating key synaptic proteins for up-scaling. Mechanistically, chronic neuronal inactivity causes dephosphorylation of ERK and mTOR, which induces transcription factor EB (TFEB)-mediated cytonuclear signaling and drives transcription-dependent autophagy to regulate αCaMKII and PSD95 during synaptic up-scaling. Together, these findings suggest that mTOR-dependent autophagy, which is often triggered by metabolic stressors such as starvation, is recruited and sustained during neuronal inactivity to maintain synaptic homeostasis, a process that ensures proper brain function and if impaired can cause neuropsychiatric disorders such as autism.SIGNIFICANCE STATEMENT In the mammalian brain, protein turnover is tightly controlled by neuronal activation to ensure key neuronal functions during long-lasting synaptic plasticity. However, a long-standing question is how this process occurs during synaptic up-scaling, a process that requires protein turnover but is induced by neuronal inactivation. Here, we report that mTOR-dependent signaling, which is often triggered by metabolic stressors such as starvation, is "hijacked" by chronic neuronal inactivation, which then serves as a nucleation point for transcription factor EB (TFEB) cytonuclear signaling that drives transcription-dependent autophagy for up-scaling. These results provide the first evidence of a physiological role of mTOR-dependent autophagy in enduing neuronal plasticity, thereby connecting major themes in cell biology and neuroscience via a servo loop that mediates autoregulation in the brain.
Collapse
Affiliation(s)
- Yang Wang
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Jingran Lin
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Jiarui Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Lu Yan
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Wenwen Li
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
| | - Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China
- National Health Commission of the PRC (NHC) and Chinese Academy of Medical Sciences (CAMS) Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 310058, China
- Research Units for Emotion and Emotion disorders, Chinese Academy of Medical Sciences, Beijing 100050, China
| |
Collapse
|
27
|
Jin M, Liu Y, Hu G, Li X, Jia N, Cui X, Li Z, Ai L, Xie M, Xue F, Yang Y, Li W, Zhang M, Yu Q. Establishment of a schizophrenia classifier based on peripheral blood signatures and investigation of pathogenic miRNA-mRNA regulation. J Psychiatr Res 2023; 159:172-184. [PMID: 36738648 DOI: 10.1016/j.jpsychires.2023.01.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/04/2023] [Accepted: 01/26/2023] [Indexed: 01/30/2023]
Abstract
To date, the diagnosis of schizophrenia (SCZ) mainly relies on patients' or guardians' self-reports and clinical observation, and the pathogenesis of SCZ remains elusive. In this study, we sought to develop a reliable classifier for diagnosing SCZ patients and provide clues to the etiology and pathogenesis of SCZ. Based on the high throughput sequencing analysis of peripheral blood miRNA expression profile and weighted gene co-expression network analysis (WGCNA) in our previous study, we selected eleven hub miRNAs for validation by qRT-PCR in 51 SCZ patients and 51 controls. miR-939-5p, miR-4732-3p let-7d-3p, and miR-142-3p were confirmed to be significantly up-regulated, and miR-30e-3p and miR-23a-3p were down-regulated in SCZ patients. miR-30e-3p with the most considerable fold change and statistically significance was selected for targeting validation. We first performed bioinformatics prediction followed by qRT-PCR and verified the up-regulation of potential target mRNAs (ABI1, NMT1, HMGB1) expression. Next, we found that the expression level of ABI1 was significantly up-regulated in SH-SY5Y cells transfected with miR-30e-3p mimics. Lastly, we conducted a luciferase assay in 293T cells confirming that miR-30e-3p could directly bind with the 3'untranslated region (3'-UTR) of ABI1, revealing that miR-30e-3p might play a role in the polymerization of neuronal actin and the reconstruction of the cytoskeleton via the downstream regulation of ABI1. In addition, we constructed a classifier by a series of bioinformatics algorithms and evaluated its diagnostic performance. It appears that the classifier consists of miRNAs and mRNAs possess a better discrimination performance than individual miRNA or mRNA in SCZ.
Collapse
Affiliation(s)
- Mengdi Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yane Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Guoyan Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Xinwei Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Ningning Jia
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Xingyao Cui
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Zhijun Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Lizhe Ai
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Mengtong Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Fengyu Xue
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yuqing Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Weizhen Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Min Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China
| | - Qiong Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
28
|
Dai W, Zhao M, Chen C, Zhou C, Wang P, Yang Z, Gao S, Lu Y, Zhang J, Liu X. Nano C60 Promotes Synaptic Distribution of Phosphorylated CaMKIIα and Improves Cognitive Function in APP/PS1 Transgenic Mice. ACS Chem Neurosci 2022; 13:3534-3543. [PMID: 36441865 DOI: 10.1021/acschemneuro.2c00569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The wide disparity in outcomes of Alzheimer's disease (AD) treatment from preclinical to clinical studies suggests an urgent need for more effective therapeutic targets and approaches to treat AD. CaMKII is a potential target for AD therapy; however, conflicting reports on the relationship between CaMKII and AD suggest a lack of deeper understanding of the interaction between CaMKII and AD. In addition to the lack of effective therapeutic targets, pharmacokinetic limitations of neuroprotective drugs, such as low lipophilicity to cross blood brain barrier, need to be urgently addressed in the practice of AD therapy. In this study, we prepared a carbon-based nanoparticle, Nano C60, and demonstrated that Nano C60 treatment promoted the translocation of phosphorylated CaMKIIα from the cytoplasm to the synapse in Aβ42 oligomers-treated cells and APP/PS1 mice. As a result, Nano C60 administration significantly improved spatial learning and memory in APP/PS1 mice. Our study suggests that synaptic-activated CaMKII may be more important than total CaMKII in AD treatment and provides a new strategy for AD therapy.
Collapse
Affiliation(s)
- Wei Dai
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| | - Mingxu Zhao
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| | - Caiyun Chen
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| | - Chang Zhou
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| | - Peng Wang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| | - Zhilai Yang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| | - Shan Gao
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei 230000, China
| | - Yao Lu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| | - Jiqian Zhang
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| | - Xuesheng Liu
- Department of Anesthesiology, the First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, Hefei 230000, China
| |
Collapse
|
29
|
Zhang L, Padilla‐Flores T, Hernández VS, Zetter MA, Campos‐Lira E, Escobar LI, Millar RP, Eiden LE. Vasopressin acts as a synapse organizer in limbic regions by boosting PSD95 and GluA1 expression. J Neuroendocrinol 2022; 34:e13164. [PMID: 35666232 PMCID: PMC9787762 DOI: 10.1111/jne.13164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 05/01/2022] [Accepted: 05/10/2022] [Indexed: 12/31/2022]
Abstract
Hypothalamic arginine vasopressin (AVP)-containing magnocellular neurosecretory neurons (AVPMNN) emit collaterals to synaptically innervate limbic regions influencing learning, motivational behaviour, and fear responses. Here, we characterize the dynamics of expression changes of two key determinants for synaptic strength, the postsynaptic density (PSD) proteins AMPAR subunit GluA1 and PSD scaffolding protein 95 (PSD95), in response to in vivo manipulations of AVPMNN neuronal activation state, or exposure to exogenous AVP ex vivo. Both long-term water deprivation in vivo, which powerfully upregulates AVPMNN metabolic activity, and exogenous AVP application ex vivo, in brain slices, significantly increased GluA1 and PSD95 expression as measured by western blotting, in brain regions reportedly receiving direct ascending innervations from AVPMNN (i.e., ventral hippocampus, amygdala and lateral habenula). By contrast, the visual cortex, a region not observed to receive AVPMNN projections, showed no such changes. Ex vivo application of V1a and V1b antagonists to ventral hippocampal slices ablated the AVP stimulated increase in postsynaptic protein expression measured by western blotting. Using a modified expansion microscopy technique, we were able to quantitatively assess the significant augmentation of PSD95 and GLUA1 densities in subcellular compartments in locus coeruleus tyrosine hydroxylase immunopositive fibres, adjacent to AVP axon terminals. Our data strongly suggest that the AVPMNN ascending system plays a role in the regulation of the excitability of targeted neuronal circuits through upregulation of key postsynaptic density proteins corresponding to excitatory synapses.
Collapse
Affiliation(s)
- Limei Zhang
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
| | - Teresa Padilla‐Flores
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
| | - Vito S. Hernández
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
| | - Mario A. Zetter
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
| | - Elba Campos‐Lira
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
| | - Laura I. Escobar
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
| | - Robert P. Millar
- Department of Physiology, School of MedicineNational Autonomous University of MexicoMexico CityMexico
- Centre for Neuroendocrinology, Department of ImmunologyUniversity of PretoriaPretoriaSouth Africa
- Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular MedicineUniversity of Cape TownCape TownSouth Africa
| | - Lee E. Eiden
- Section on Molecular NeuroscienceNIMH‐IRP, NIHBethesdaMarylandUSA
| |
Collapse
|
30
|
Liu Z, Jin Q, Yan T, Wo Y, Liu H, Wang Y. Exosome-mediated transduction of mechanical force regulates prostate cancer migration via microRNA. Biochem Biophys Rep 2022; 31:101299. [PMID: 35812347 PMCID: PMC9257336 DOI: 10.1016/j.bbrep.2022.101299] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/29/2022] Open
Abstract
Physical cues in the extracellular microenvironment regulate cancer cell metastasis. Functional microRNA (miRNA) carried by cancer derived exosomes play a critical role in extracellular communication between cells and the extracellular microenvironment. However, little is known about the role of exosomes loaded miRNAs in the mechanical force transmission between cancer cells and extracellular microenvironment. Herein, our results suggest that stiff extracellular matrix (ECM) induced exosomes promote cancer cell migration. The ECM mechanical force regulated the exosome miRNA cargo of prostate cancer cells. Exosome miRNAs regulated by the ECM mechanical force modulated cancer cell metastasis by regulating cell motility, ECM remodeling and the interaction between cancer cells and nerves. Focal adhesion kinase mediated-ECM mechanical force regulated the intracellular miRNA expression, and F-actin mediate-ECM mechanical force regulated miRNA packaging into exosomes. The above results demonstrated that the exosome miRNA cargo promoted cancer metastasis by transmitting the ECM mechanical force. The ECM mechanical force may play multiple roles in maintaining the microenvironment of cancer metastasis through the exosome miRNA cargo. ECM mechanical force-induced exosomes regulate cancer cell migration. ECM mechanical forces regulate the cancer cell exosomes miRNA cargo. ECM mechanical forces regulated exosomes miRNAs modulate cancer metastasis by remodeling extracellular microenvironment.
Collapse
|
31
|
Isolation, cryo-laser scanning confocal microscope imaging and cryo-FIB milling of mouse glutamatergic synaptosomes. PLoS One 2022; 17:e0271799. [PMID: 35960737 PMCID: PMC9374259 DOI: 10.1371/journal.pone.0271799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 07/07/2022] [Indexed: 11/19/2022] Open
Abstract
Ionotropic glutamate receptors (iGluRs) at postsynaptic terminals mediate the majority of fast excitatory neurotransmission in response to release of glutamate from the presynaptic terminal. Obtaining structural information on the molecular organization of iGluRs in their native environment, along with other signaling and scaffolding proteins in the postsynaptic density (PSD), and associated proteins on the presynaptic terminal, would enhance understanding of the molecular basis for excitatory synaptic transmission in normal and in disease states. Cryo-electron tomography (ET) studies of synaptosomes is one attractive vehicle by which to study iGluR-containing excitatory synapses. Here we describe a workflow for the preparation of glutamatergic synaptosomes for cryo-ET studies. We describe the utilization of fluorescent markers for the facile detection of the pre and postsynaptic terminals of glutamatergic synaptosomes using cryo-laser scanning confocal microscope (cryo-LSM). We further provide the details for preparation of lamellae, between ~100 to 200 nm thick, of glutamatergic synaptosomes using cryo-focused ion-beam (FIB) milling. We monitor the lamella preparation using a scanning electron microscope (SEM) and following lamella production, we identify regions for subsequent cryo-ET studies by confocal fluorescent imaging, exploiting the pre and postsynaptic fluorophores.
Collapse
|
32
|
Post-Synapses in the Brain: Role of Dendritic and Spine Structures. Biomedicines 2022; 10:biomedicines10081859. [PMID: 36009405 PMCID: PMC9405724 DOI: 10.3390/biomedicines10081859] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/26/2022] [Accepted: 07/22/2022] [Indexed: 02/07/2023] Open
Abstract
Brain synapses are neuronal structures of the greatest interest. For a long time, however, the knowledge about them was variable, and interest was mostly focused on their pre-synaptic portions, especially neurotransmitter release from axon terminals. In the present review interest is focused on post-synapses, the structures receiving and converting pre-synaptic messages. Upon further modulation, such messages are transferred to dendritic fibers. Dendrites are profoundly different from axons; they are shorter and of variable thickness. Their post-synapses are of two types. Those called flat/intended/aspines, integrated into dendritic fibers, are very frequent in inhibitory neurons. The spines, small and stemming protrusions, connected to dendritic fibers by their necks, are present in almost all excitatory neurons. Several structures and functions including the post-synaptic densities and associated proteins, the nanoscale mechanisms of compartmentalization, the cytoskeletons of actin and microtubules, are analogous in the two post-synaptic forms. However other properties, such as plasticity and its functions of learning and memory, are largely distinct. Several properties of spines, including emersion from dendritic fibers, growth, change in shape and decreases in size up to disappearance, are specific. Spinal heads correspond to largely independent signaling compartments. They are motile, their local signaling is fast, however transport through their thin necks is slow. When single spines are activated separately, their dendritic effects are often lacking; when multiple spines are activated concomitantly, their effects take place. Defects of post-synaptic responses, especially those of spines, take place in various brain diseases. Here alterations affecting symptoms and future therapy are shown to occur in neurodegenerative diseases and autism spectrum disorders.
Collapse
|
33
|
Synaptic Secretion and Beyond: Targeting Synapse and Neurotransmitters to Treat Neurodegenerative Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9176923. [PMID: 35923862 PMCID: PMC9343216 DOI: 10.1155/2022/9176923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/16/2022] [Accepted: 06/04/2022] [Indexed: 11/17/2022]
Abstract
The nervous system is important, because it regulates the physiological function of the body. Neurons are the most basic structural and functional unit of the nervous system. The synapse is an asymmetric structure that is important for neuronal function. The chemical transmission mode of the synapse is realized through neurotransmitters and electrical processes. Based on vesicle transport, the abnormal information transmission process in the synapse can lead to a series of neurorelated diseases. Numerous proteins and complexes that regulate the process of vesicle transport, such as SNARE proteins, Munc18-1, and Synaptotagmin-1, have been identified. Their regulation of synaptic vesicle secretion is complicated and delicate, and their defects can lead to a series of neurodegenerative diseases. This review will discuss the structure and functions of vesicle-based synapses and their roles in neurons. Furthermore, we will analyze neurotransmitter and synaptic functions in neurodegenerative diseases and discuss the potential of using related drugs in their treatment.
Collapse
|
34
|
SAPAP3 regulates epileptic seizures involving GluN2A in post-synaptic densities. Cell Death Dis 2022; 13:437. [PMID: 35513389 PMCID: PMC9072407 DOI: 10.1038/s41419-022-04876-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/15/2022] [Accepted: 04/21/2022] [Indexed: 12/14/2022]
Abstract
Aberrantly synchronized neuronal discharges in the brain lead to epilepsy, a devastating neurological disease whose pathogenesis and mechanism are unclear. SAPAP3, a cytoskeletal protein expressed at high levels in the postsynaptic density (PSD) of excitatory synapses, has been well studied in the striatum, but the role of SAPAP3 in epilepsy remains elusive. In this study, we sought to investigate the molecular, cellular, electrophysiological and behavioral consequences of SAPAP3 perturbations in the mouse hippocampus. We identified a significant increase in the SAPAP3 levels in patients with temporal lobe epilepsy (TLE) and in mouse models of epilepsy. In addition, behavioral studies showed that the downregulation of SAPAP3 by shRNA decreased the seizure severity and that the overexpression of SAPAP3 by recombinant SAPAP3 yielded the opposite effect. Moreover, SAPAP3 affected action potentials (APs), miniature excitatory postsynaptic currents (mEPSCs) and N-methyl-D-aspartate receptor (NMDAR)-mediated currents in the CA1 region, which indicated that SAPAP3 plays an important role in excitatory synaptic transmission. Additionally, the levels of the GluN2A protein, which is involved in synaptic function, were perturbed in the hippocampal PSD, and this perturbation was accompanied by ultrastructural morphological changes. These results revealed a previously unknown function of SAPAP3 in epileptogenesis and showed that SAPAP3 may represent a novel target for the treatment of epilepsy.
Collapse
|
35
|
Autoregulation of switching behavior by cellular compartment size. Proc Natl Acad Sci U S A 2022; 119:e2116054119. [PMID: 35349334 PMCID: PMC9169097 DOI: 10.1073/pnas.2116054119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Biochemical reactions often occur in small volumes within a cell, restricting the number of molecules to the hundreds or even tens. At this scale, reactions are discrete and stochastic, making reliable signaling difficult. This paper shows that the transition between discrete, stochastic reactions and macroscopic reactions can be exploited to make a self-regulating switch. This constitutes a previously unidentified kind of reaction network that may be present in small structures, such as synapses. Many kinds of cellular compartments comprise decision-making mechanisms that control growth and shrinkage of the compartment in response to external signals. Key examples include synaptic plasticity mechanisms that regulate the size and strength of synapses in the nervous system. However, when synaptic compartments and postsynaptic densities are small, such mechanisms operate in a regime where chemical reactions are discrete and stochastic due to low copy numbers of the species involved. In this regime, fluctuations are large relative to mean concentrations, and inherent discreteness leads to breakdown of mass-action kinetics. Understanding how synapses and other small compartments achieve reliable switching in the low–copy number limit thus remains a key open problem. We propose a self-regulating signaling motif that exploits the breakdown of mass-action kinetics to generate a reliable size-regulated switch. We demonstrate this in simple two- and three-species chemical reaction systems and uncover a key role for inhibitory loops among species in generating switching behavior. This provides an elementary motif that could allow size-dependent regulation in more complex reaction pathways and may explain discrepant experimental results on well-studied biochemical pathways.
Collapse
|
36
|
Grassi D, Marraudino M, Garcia-Segura LM, Panzica GC. The hypothalamic paraventricular nucleus as a central hub for the estrogenic modulation of neuroendocrine function and behavior. Front Neuroendocrinol 2022; 65:100974. [PMID: 34995643 DOI: 10.1016/j.yfrne.2021.100974] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/17/2022]
Abstract
Estradiol and hypothalamic paraventricular nucleus (PVN) help coordinate reproduction with body physiology, growth and metabolism. PVN integrates hormonal and neural signals originating in the periphery, generating an output mediated both by its long-distance neuronal projections, and by a variety of neurohormones produced by its magnocellular and parvocellular neurosecretory cells. Here we review the cyto-and chemo-architecture, the connectivity and function of PVN and the sex-specific regulation exerted by estradiol on PVN neurons and on the expression of neurotransmitters, neuromodulators, neuropeptides and neurohormones in PVN. Classical and non-classical estrogen receptors (ERs) are expressed in neuronal afferents to PVN and in specific PVN interneurons, projecting neurons, neurosecretory neurons and glial cells that are involved in the input-output integration and coordination of neurohormonal signals. Indeed, PVN ERs are known to modulate body homeostatic processes such as autonomic functions, stress response, reproduction, and metabolic control. Finally, the functional implications of the estrogenic modulation of the PVN for body homeostasis are discussed.
Collapse
Affiliation(s)
- D Grassi
- Department of Anatomy, Histology and Neuroscience, Universidad Autonoma de Madrid, Madrid, Spain
| | - M Marraudino
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy
| | - L M Garcia-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - G C Panzica
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Torino, Italy; Department of Neuroscience Rita Levi Montalcini, University of Torino, Torino, Italy.
| |
Collapse
|
37
|
Sheibani V, Rajizadeh MA, Bejeshk MA, Haghparast E, Nozari M, Esmaeili-Mahani S, Nezhadi A. The effects of neurosteroid allopregnanolone on synaptic dysfunction in the hippocampus in experimental parkinsonism rats: An electrophysiological and molecular study. Neuropeptides 2022; 92:102229. [PMID: 35158223 DOI: 10.1016/j.npep.2022.102229] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/18/2022] [Accepted: 02/03/2022] [Indexed: 02/07/2023]
Abstract
The dopaminergic system is a powerful candidate targeted for changes of synaptic plasticity in the hippocampus. Higher incidence of Parkinson's disease (PD) in men than women indicates the influence of sex hormones on the PD development. Previous studies have shown that neurodegenerative diseases such as PD are related to the decline of Allopregnanolon (Allo), a metabolite of progesterone; it is also well known that learning and memory are influenced by oscillations in steroidal hormones. Although abnormalities in hippocampal plasticity have been observed in the toxic models of PD, effects of Allo on hippocampal LTP and hippocampal synaptic protein levels, which play an important role in maintaining the integrity of neural connections, have never been analyzed thus far. Experimental groups subjected to the long-term potentiation (LTP) were studied in the CA1 area of the hippocampus. In addition, the levels of hippocampal postsynaptic density protein 95 (PSD-95), neurexin-1 (Nrxn1) and neuroligin (Nlgn) as synaptic molecular components were determined by immunoblotting. Although dopamine denervation did not alter basal synaptic transmission and pair-pulse facilitation of field excitatory postsynaptic potentials (fEPSPs), the induction and maintenance of LTP were impaired in the CA1 region. In addition, the levels of PSD-95, Nrxn1 and Nlgn were significantly decreased in the hippocampus of 6-OHDA-treated animals. Such abnormalities in synaptic electrophysiological aspects and protein levels were abolished by the treatment with Allo. These findings showed that partial dopamine depletion led to unusual synaptic plasticity in the CA1 as well as the decrease in synaptic proteins in the hippocampus. Our results demonstrated that Allo ameliorated these deficits and preserved pre- and post-synaptic proteins. Therefore, Allo may be an effective factor in maintaining synaptic integrity in the mesolimbic pathway.
Collapse
Affiliation(s)
- Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Rajizadeh
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Abbas Bejeshk
- Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Haghparast
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Masoumeh Nozari
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Physiology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Saeed Esmaeili-Mahani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran; Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Akram Nezhadi
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
38
|
Sando R, Ho ML, Liu X, Südhof TC. Engineered synaptic tools reveal localized cAMP signaling in synapse assembly. J Cell Biol 2022; 221:e202109111. [PMID: 34913963 PMCID: PMC8685283 DOI: 10.1083/jcb.202109111] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/15/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
The physiological mechanisms driving synapse formation are elusive. Although numerous signals are known to regulate synapses, it remains unclear which signaling mechanisms organize initial synapse assembly. Here, we describe new tools, referred to as "SynTAMs" for synaptic targeting molecules, that enable localized perturbations of cAMP signaling in developing postsynaptic specializations. We show that locally restricted suppression of postsynaptic cAMP levels or of cAMP-dependent protein-kinase activity severely impairs excitatory synapse formation without affecting neuronal maturation, dendritic arborization, or inhibitory synapse formation. In vivo, suppression of postsynaptic cAMP signaling in CA1 neurons prevented formation of both Schaffer-collateral and entorhinal-CA1/temporoammonic-path synapses, suggesting a general principle. Retrograde trans-synaptic rabies virus tracing revealed that postsynaptic cAMP signaling is required for continuous replacement of synapses throughout life. Given that postsynaptic latrophilin adhesion-GPCRs drive synapse formation and produce cAMP, we suggest that spatially restricted postsynaptic cAMP signals organize assembly of postsynaptic specializations during synapse formation.
Collapse
Affiliation(s)
- Richard Sando
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA
- Department of Pharmacology, Vanderbilt University, Nashville, TN
| | - Milan Lyndie Ho
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Thomas C. Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
39
|
Structure-Based Analysis of Protein Cluster Size for Super-Resolution Microscopy in the Nervous System. Biomedicines 2022; 10:biomedicines10020295. [PMID: 35203503 PMCID: PMC8869149 DOI: 10.3390/biomedicines10020295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 02/01/2023] Open
Abstract
To overcome the diffraction limit and resolve target structures in greater detail, far-field super-resolution techniques such as stochastic optical reconstruction microscopy (STORM) have been developed, and different STORM algorithms have been developed to deal with the various problems that arise. In particular, the effect of the local structure is an important issue. For objects with closely correlated distributions, simple Gaussian-based localization algorithms often used in STORM imaging misinterpret overlapping point spread functions (PSFs) as one, which limits the ability of super-resolution imaging to resolve nanoscale local structures and leads to inaccurate length measurements. The STORM super-resolution images of biological specimens from the cluster-forming proteins in the nervous system were reconstructed for localization-based analysis. Generally, the localization of each fluorophore was determined by two-dimensional Gaussian function fitting. Further, the physical shape of the cluster structure information was incorporated into the size parameter of the localization structure analysis in order to generate structure-based fitting algorithms. In the present study, we proposed a novel, structure-based, super-resolution image analysis method: structure-based analysis (SBA), which combines a structural function and a super-resolution localization algorithm. Using SBA, we estimated the size of fluorescent beads, inclusion proteins, and subtle synaptic structures in both wide-field and STORM images. The results show that SBA has a comparable and often superior performance to the commonly used full width at half maximum (FWHM) parameter. We demonstrated that SBA is able to estimate molecular cluster sizes in far-field super-resolution STORM images, and that SBA was comparable and often superior to FWHM. We also certified that SBA provides size estimations that corroborate previously published electron microscopy data.
Collapse
|
40
|
Yan Q, Wu X, Zhou P, Zhou Y, Li X, Liu Z, Tan H, Yao W, Xia Y, Zhu F. HERV-W Envelope Triggers Abnormal Dopaminergic Neuron Process through DRD2/PP2A/AKT1/GSK3 for Schizophrenia Risk. Viruses 2022; 14:145. [PMID: 35062349 PMCID: PMC8777930 DOI: 10.3390/v14010145] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/08/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
An increasing number of studies have begun considering human endogenous retroviruses (HERVs) as potential pathogenic phenomena. Our previous research suggests that HERV-W Envelope (HERV-W ENV), a HERV-W family envelope protein, is elevated in schizophrenia patients and contributes to the pathophysiology of schizophrenia. The dopamine (DA) hypothesis is the cornerstone in research and clinical practice related to schizophrenia. Here, we found that the concentration of DA and the expression of DA receptor D2 (DRD2) were significantly higher in schizophrenia patients than in healthy individuals. Intriguingly, there was a positive correlation between HERV-W ENV and DA concentration. Depth analyses showed that there was a marked consistency between HERV-W ENV and DRD2 in schizophrenia. Studies in vitro indicated that HERV-W ENV could increase the DA concentration by regulating DA metabolism and induce the expression of DRD2. Co-IP assays and laser confocal scanning microscopy indicated cellular colocalization and a direct interaction between DRD2 and HERV-W ENV. Additionally, HERV-W ENV caused structural and functional abnormalities of DA neurons. Further studies showed that HERV-W ENV could trigger the PP2A/AKT1/GSK3 pathway via DRD2. A whole-cell patch-clamp analysis suggested that HERV-W ENV enhanced sodium influx through DRD2. In conclusion, we uncovered a relationship between HERV-W ENV and the dopaminergic system in the DA neurons. Considering that GNbAC1, a selective monoclonal antibody to the MSRV-specific epitope, has been promised as a therapy for treating type 1 diabetes and multiple sclerosis (MS) in clinical trials, understanding the precise function of HERV-W ENV in the dopaminergic system may provide new insights into the treatment of schizophrenia.
Collapse
Affiliation(s)
- Qiujin Yan
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; (Q.Y.); (X.W.); (P.Z.); (Y.Z.); (X.L.); (W.Y.); (Y.X.)
| | - Xiulin Wu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; (Q.Y.); (X.W.); (P.Z.); (Y.Z.); (X.L.); (W.Y.); (Y.X.)
| | - Ping Zhou
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; (Q.Y.); (X.W.); (P.Z.); (Y.Z.); (X.L.); (W.Y.); (Y.X.)
| | - Yan Zhou
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; (Q.Y.); (X.W.); (P.Z.); (Y.Z.); (X.L.); (W.Y.); (Y.X.)
| | - Xuhang Li
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; (Q.Y.); (X.W.); (P.Z.); (Y.Z.); (X.L.); (W.Y.); (Y.X.)
| | - Zhongchun Liu
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Z.L.); (H.T.)
| | - Huawei Tan
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; (Z.L.); (H.T.)
| | - Wei Yao
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; (Q.Y.); (X.W.); (P.Z.); (Y.Z.); (X.L.); (W.Y.); (Y.X.)
| | - Yaru Xia
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; (Q.Y.); (X.W.); (P.Z.); (Y.Z.); (X.L.); (W.Y.); (Y.X.)
| | - Fan Zhu
- State Key Laboratory of Virology, Department of Medical Microbiology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China; (Q.Y.); (X.W.); (P.Z.); (Y.Z.); (X.L.); (W.Y.); (Y.X.)
- Hubei Province Key Laboratory of Allergy & Immunology, Wuhan University, Wuhan 430071, China
| |
Collapse
|
41
|
Lu X, Zhou M, Liu N, Zhang C, Zhao Z, Cai D. Synaptic Protein Phosphorylation Networks Are Associated With Electroacupuncture-Induced Circadian Control in the Suprachiasmatic Nucleus. Front Genet 2021; 12:762557. [PMID: 34976011 PMCID: PMC8717940 DOI: 10.3389/fgene.2021.762557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 11/29/2021] [Indexed: 11/23/2022] Open
Abstract
Phosphorylation is one of the most important posttranslational modifications and regulates the physiological process. While recent studies highlight a major role of phosphorylation in the regulation of sleep–wake cycles to a lesser extent, the phosphoproteome in the suprachiasmatic nucleus (SCN) is not well-understood. Herein, we reported that the EA treatment elicits partial reparation of circadian rhythmicity when mice were exposure to constant darkness for long time. We investigated the effects of EA on circadian rhythms in constant darkness between EA stimulation and free-running control. Next, mass spectrometry–based phosphoproteome was utilized to explore the molecular characteristics of EA-induced phosphorylation modification in the SCN. A total of 6,192 distinct phosphosites on 2,488 proteins were quantified. Functional annotation analysis and protein–protein interaction networks demonstrated the most significant enriched phosphor-proteins and phosphosites involved in postsynapse and glutamatergic synapse. The current data indicated that most of the altered molecules are structural proteins. The target proteins, NMDAR and CAMK2, were selected for verification, consistent with the results of LC–MS/MS. These findings revealed a complete profile of phosphorylation modification in response to EA.
Collapse
Affiliation(s)
| | | | | | | | | | - Dingjun Cai
- *Correspondence: Zhengyu Zhao, ; Dingjun Cai,
| |
Collapse
|
42
|
Su X, Li W, Li Z, Liu K, Song M, Shao M, Lv L, Chang X. Chronic developmental exposure to low-dose ([C8mim][PF6]) induces neurotoxicity and behavioural abnormalities in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 225:112806. [PMID: 34555716 DOI: 10.1016/j.ecoenv.2021.112806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 08/31/2021] [Accepted: 09/16/2021] [Indexed: 06/13/2023]
Abstract
Ionic liquids (ILs) are widely used for their physical and chemical properties. Toxicological assessments of ILs could help to avoid their threat to human health, but these are rarely reported, and no assessments of IL neurotoxicity in mammals have been performed. Here, we aimed to evaluate the neurotoxicity of chronic 1-octyl-3-methylimidazolium hexafluorophosphate ([C8mim][PF6]) (0, 1 mg/kg) exposure during development on rats. Our results indicated that chronic exposure to low-dose ([C8mim][PF6]) induces behavioural abnormalities, including cognitive deficits, social communication disorders, and sensory gating function impairment. Moreover, rats subjected to chronic ([C8mim][PF6]) exposure showed hypofunction of glutamatergic excitatory synapses, including increased expression of NMDA receptor subunits, increased density and immaturity of dendritic spines, and increased expression of PSD95. Additionally, ([C8mim][PF6]) exposure resulted in hippocampal-specific inflammatory activation, indicated by increased levels of proinflammatory factors, elevated nuclear localisation of NF-κB, and activation of microglia and astrocytes. In conclusion, chronic exposure to low-dose ([C8mim][PF6]) induced neurotoxicity, including damage to glutamatergic excitatory synapses and inflammatory activation, which may illuminate the associated behavioural abnormalities. The results presented here may be helpful for the safe use of ILs in the future.
Collapse
Affiliation(s)
- Xi Su
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453007, PR China; Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, PR China
| | - Wenqiang Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453007, PR China; Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, PR China
| | - Zhen Li
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453007, PR China; Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, PR China
| | - Kang Liu
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453007, PR China; Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, PR China
| | - Meng Song
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453007, PR China; Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, PR China
| | - Minglong Shao
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453007, PR China; Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, PR China
| | - Luxian Lv
- Henan Mental Hospital, The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang 453007, PR China; Henan Key Lab of Biological Psychiatry of Xinxiang Medical University, Xinxiang, PR China
| | - Xulu Chang
- College of Fisheries, Henan Normal University, Xinxiang 453007, PR China.
| |
Collapse
|
43
|
Vallés AS, Barrantes FJ. Dendritic spine membrane proteome and its alterations in autistic spectrum disorder. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 128:435-474. [PMID: 35034726 DOI: 10.1016/bs.apcsb.2021.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dendritic spines are small protrusions stemming from the dendritic shaft that constitute the primary specialization for receiving and processing excitatory neurotransmission in brain synapses. The disruption of dendritic spine function in several neurological and neuropsychiatric diseases leads to severe information-processing deficits with impairments in neuronal connectivity and plasticity. Spine dysregulation is usually accompanied by morphological alterations to spine shape, size and/or number that may occur at early pathophysiological stages and not necessarily be reflected in clinical manifestations. Autism spectrum disorder (ASD) is one such group of diseases involving changes in neuronal connectivity and abnormal morphology of dendritic spines on postsynaptic neurons. These alterations at the subcellular level correlate with molecular changes in the spine proteome, with alterations in the copy number, topography, or in severe cases in the phenotype of the molecular components, predominantly of those proteins involved in spine recognition and adhesion, reflected in abnormally short lifetimes of the synapse and compensatory increases in synaptic connections. Since cholinergic neurotransmission participates in the regulation of cognitive function (attention, memory, learning processes, cognitive flexibility, social interactions) brain acetylcholine receptors are likely to play an important role in the dysfunctional synapses in ASD, either directly or indirectly via the modulatory functions exerted on other neurotransmitter receptor proteins and spine-resident proteins.
Collapse
Affiliation(s)
- Ana Sofía Vallés
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (UNS-CONICET), Bahía Blanca, Argentina
| | - Francisco J Barrantes
- Instituto de Investigaciones Biomédicas (BIOMED), UCA-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
44
|
Imaging intracellular protein interactions/activity in neurons using 2-photon fluorescence lifetime imaging microscopy. Neurosci Res 2021; 179:31-38. [PMID: 34666101 DOI: 10.1016/j.neures.2021.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/23/2022]
Abstract
Through the decades, 2-photon fluorescence microscopy has allowed visualization of microstructures, such as synapses, with high spatial resolution in deep brain tissue. However, signal transduction, such as protein activity and protein-protein interaction in neurons in tissues and in vivo, has remained elusive because of the technical difficulty of observing biochemical reactions at the level of subcellular resolution in light-scattering tissues. Recently, 2-photon fluorescence microscopy combined with fluorescence lifetime imaging microscopy (2pFLIM) has enabled visualization of various protein activities and protein-protein interactions at submicrometer resolution in tissue with a reasonable temporal resolution. Thus far, 2pFLIM has been extensively applied for imaging kinase and small GTPase activation in dendritic spines of hippocampal neurons in slice cultures. However, it has been recently applied to various subcellular structures, such as axon terminals and nuclei, and has increased our understanding of spatially organized molecular dynamics. One of the future directions of 2pFLIM utilization is to combine various optogenetic tools for manipulating protein activity. This combination allows the activation of specific proteins with light and visualization of its readout as the activation of downstream molecules. Here, we have introduced the recent application of 2pFLIM for neurons and present the utilization of a new optogenetic tool in combination with 2pFLIM.
Collapse
|
45
|
Zhang H, Ben Zablah Y, Zhang H, Jia Z. Rho Signaling in Synaptic Plasticity, Memory, and Brain Disorders. Front Cell Dev Biol 2021; 9:729076. [PMID: 34671600 PMCID: PMC8520953 DOI: 10.3389/fcell.2021.729076] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/03/2021] [Indexed: 12/12/2022] Open
Abstract
Memory impairments are associated with many brain disorders such as autism, Alzheimer's disease, and depression. Forming memories involves modifications of synaptic transmission and spine morphology. The Rho family small GTPases are key regulators of synaptic plasticity by affecting various downstream molecules to remodel the actin cytoskeleton. In this paper, we will review recent studies on the roles of Rho proteins in the regulation of hippocampal long-term potentiation (LTP) and long-term depression (LTD), the most extensively studied forms of synaptic plasticity widely regarded as cellular mechanisms for learning and memory. We will also discuss the involvement of Rho signaling in spine morphology, the structural basis of synaptic plasticity and memory formation. Finally, we will review the association between brain disorders and abnormalities of Rho function. It is expected that studying Rho signaling at the synapse will contribute to the understanding of how memory is formed and disrupted in diseases.
Collapse
Affiliation(s)
- Haorui Zhang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Youssif Ben Zablah
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Haiwang Zhang
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Zhengping Jia
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
46
|
Wood BM, Baena V, Huang H, Jorgens DM, Terasaki M, Kornberg TB. Cytonemes with complex geometries and composition extend into invaginations of target cells. J Cell Biol 2021; 220:211896. [PMID: 33734293 PMCID: PMC7980254 DOI: 10.1083/jcb.202101116] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/13/2021] [Accepted: 02/23/2021] [Indexed: 12/16/2022] Open
Abstract
Cytonemes are specialized filopodia that mediate paracrine signaling in Drosophila and other animals. Studies using fluorescence confocal microscopy (CM) established their general paths, cell targets, and essential roles in signaling. To investigate details unresolvable by CM, we used high-pressure freezing and EM to visualize cytoneme structures, paths, contents, and contacts. We observed cytonemes previously seen by CM in the Drosophila wing imaginal disc system, including disc, tracheal air sac primordium (ASP), and myoblast cytonemes, and identified cytonemes extending into invaginations of target cells, and cytonemes connecting ASP cells and connecting myoblasts. Diameters of cytoneme shafts vary between repeating wide (206 ± 51.8 nm) and thin (55.9 ± 16.2 nm) segments. Actin, ribosomes, and membranous compartments are present throughout; rough ER and mitochondria are in wider proximal sections. These results reveal novel structural features of filopodia and provide a basis for understanding cytoneme cell biology and function.
Collapse
Affiliation(s)
- Brent M Wood
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Valentina Baena
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT
| | - Hai Huang
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Danielle M Jorgens
- Electron Microscope Laboratory, University of California, Berkeley, Berkeley, CA
| | - Mark Terasaki
- Department of Cell Biology, University of Connecticut Health Center, Farmington, CT
| | - Thomas B Kornberg
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
47
|
Watkins JC, Evans RH, Bayés À, Booker SA, Gibb A, Mabb AM, Mayer M, Mellor JR, Molnár E, Niu L, Ortega A, Pankratov Y, Ramos-Vicente D, Rodríguez-Campuzano A, Rodríguez-Moreno A, Wang LY, Wang YT, Wollmuth L, Wyllie DJA, Zhuo M, Frenguelli BG. 21st century excitatory amino acid research: A Q & A with Jeff Watkins and Dick Evans. Neuropharmacology 2021; 198:108743. [PMID: 34363811 DOI: 10.1016/j.neuropharm.2021.108743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In 1981 Jeff Watkins and Dick Evans wrote what was to become a seminal review on excitatory amino acids (EAAs) and their receptors (Watkins and Evans, 1981). Bringing together various lines of evidence dating back over several decades on: the distribution in the nervous system of putative amino acid neurotransmitters; enzymes involved in their production and metabolism; the uptake and release of amino acids; binding of EAAs to membranes; the pharmacological action of endogenous excitatory amino acids and their synthetic analogues, and notably the actions of antagonists for the excitations caused by both nerve stimulation and exogenous agonists, often using pharmacological tools developed by Jeff and his colleagues, they provided a compelling account for EAAs, especially l-glutamate, as a bona fide neurotransmitter in the nervous system. The rest, as they say, is history, but far from being consigned to history, EAA research is in rude health well into the 21st Century as this series of Special Issues of Neuropharmacology exemplifies. With EAAs and their receptors flourishing across a wide range of disciplines and clinical conditions, we enter into a dialogue with two of the most prominent and influential figures in the early days of EAA research: Jeff Watkins and Dick Evans.
Collapse
Affiliation(s)
| | | | - Àlex Bayés
- Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau, Barcelona, Spain and Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sam A Booker
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Alasdair Gibb
- Research Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Angela M Mabb
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| | - Mark Mayer
- Bldg 35A, Room 3D-904, 35A Convent Drive, NINDS, NIH, Bethesda, MD, 20892, USA
| | - Jack R Mellor
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Elek Molnár
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Li Niu
- Chemistry Department, University at Albany, SUNY, 1400 Washington Ave, Albany, NY, 12222, USA
| | - Arturo Ortega
- Department of Toxicology, Cinvestav, Mexico City, Mexico
| | - Yuriy Pankratov
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - David Ramos-Vicente
- Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau, Barcelona, Spain and Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | - Lu-Yang Wang
- Program in Neurosciences & Mental Health, SickKids Research Institute and Department of Physiology, University of Toronto, 555 University Ave, Toronto, Ontario, M5G 1X8, Canada
| | - Yu Tian Wang
- Department of Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| | - Lonnie Wollmuth
- Depts. of Neurobiology & Behavior and Biochemistry & Cell Biology, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - David J A Wyllie
- Simons Initiative for the Developing Brain, Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Min Zhuo
- Institute of Brain Research, Qingdao International Academician Park, Qingdao, 266000, China
| | | |
Collapse
|
48
|
Alpha-Synuclein as a Prominent Actor in the Inflammatory Synaptopathy of Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22126517. [PMID: 34204581 PMCID: PMC8234932 DOI: 10.3390/ijms22126517] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/04/2021] [Accepted: 06/15/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson’s disease (PD) is considered the most common disorder of synucleinopathy, which is characterised by intracellular inclusions of aggregated and misfolded α-synuclein (α-syn) protein in various brain regions, and the loss of dopaminergic neurons. During the early prodromal phase of PD, synaptic alterations happen before cell death, which is linked to the synaptic accumulation of toxic α-syn specifically in the presynaptic terminals, affecting neurotransmitter release. The oligomers and protofibrils of α-syn are the most toxic species, and their overexpression impairs the distribution and activation of synaptic proteins, such as the SNARE complex, preventing neurotransmitter exocytosis and neuronal synaptic communication. In the last few years, the role of the immune system in PD has been increasingly considered. Microglial and astrocyte activation, the gene expression of proinflammatory factors, and the infiltration of immune cells from the periphery to the central nervous system (CNS) represent the main features of the inflammatory response. One of the actors of these processes is α-syn accumulation. In light of this, here, we provide a systematic review of PD-related α-syn and inflammation inter-players.
Collapse
|
49
|
Feng Z, Wu X, Zhang M. Presynaptic bouton compartmentalization and postsynaptic density-mediated glutamate receptor clustering via phase separation. Neuropharmacology 2021; 193:108622. [PMID: 34051266 DOI: 10.1016/j.neuropharm.2021.108622] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/25/2021] [Accepted: 05/17/2021] [Indexed: 01/21/2023]
Abstract
Neuronal synapses encompass three compartments: presynaptic axon terminal, synaptic cleft, and postsynaptic dendrite. Each compartment contains densely packed molecular machineries that are involved in synaptic transmission. In recent years, emerging evidence indicates that the assembly of these membraneless substructures or assemblies that are not enclosed by membranes are driven by liquid-liquid phase separation. We review here recent studies that suggest the phase separation-mediated organization of these synaptic compartments. We discuss how synaptic function may be linked to its organization as biomolecular condensates. We conclude with a discussion of areas of future interest in the field for better understanding of the structural architecture of neuronal synapses and its contribution to synaptic functions.
Collapse
Affiliation(s)
- Zhe Feng
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xiandeng Wu
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Mingjie Zhang
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
50
|
Serine/Threonine Phosphatases in LTP: Two B or Not to Be the Protein Synthesis Blocker-Induced Impairment of Early Phase. Int J Mol Sci 2021; 22:ijms22094857. [PMID: 34064311 PMCID: PMC8125358 DOI: 10.3390/ijms22094857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 11/25/2022] Open
Abstract
Dephosphorylation of target proteins at serine/threonine residues is one of the most crucial mechanisms regulating their activity and, consequently, the cellular functions. The role of phosphatases in synaptic plasticity, especially in long-term depression or depotentiation, has been reported. We studied serine/threonine phosphatase activity during the protein synthesis blocker (PSB)-induced impairment of long-term potentiation (LTP). Established protein phosphatase 2B (PP2B, calcineurin) inhibitor cyclosporin A prevented the LTP early phase (E-LTP) decline produced by pretreatment of hippocampal slices with cycloheximide or anisomycin. For the first time, we directly measured serine/threonine phosphatase activity during E-LTP, and its significant increase in PSB-treated slices was demonstrated. Nitric oxide (NO) donor SNAP also heightened phosphatase activity in the same manner as PSB, and simultaneous application of anisomycin + SNAP had no synergistic effect. Direct measurement of the NO production in hippocampal slices by the NO-specific fluorescent probe DAF-FM revealed that PSBs strongly stimulate the NO concentration in all studied brain areas: CA1, CA3, and dentate gyrus (DG). Cyclosporin A fully abolished the PSB-induced NO production in the hippocampus, suggesting a close relationship between nNOS and PP2B activity. Surprisingly, cyclosporin A alone impaired short-term plasticity in CA1 by decreasing paired-pulse facilitation, which suggests bi-directionality of the influences of PP2B in the hippocampus. In conclusion, we proposed a minimal model of signaling events that occur during LTP induction in normal conditions and the PSB-treated slices.
Collapse
|