1
|
Berchtold MW, Villalobo A. Ca 2+/calmodulin signaling in organismal aging and cellular senescence: Impact on human diseases. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167583. [PMID: 39579800 DOI: 10.1016/j.bbadis.2024.167583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 11/25/2024]
Abstract
Molecular mechanisms of aging processes at the level of organisms and cells are in the focus of a large number of research laboratories. This research culminated in recent breakthroughs, which contributed to the better understanding of the natural aging process and aging associated malfunctions leading to age-related diseases. Ca2+ in connection with its master intracellular sensor protein calmodulin (CaM) regulates a plethora of crucial cellular processes orchestrating a wide range of signaling processes. This review focuses on the involvement of Ca2+/CaM in cellular mechanisms, which are associated with normal aging, as well as playing a role in the development of diseases connected with signaling processes during aging. We specifically highlight processes that involve inactivation of proteins, which take part in Ca2+/CaM regulatory systems by oxygen or nitrogen free radical species, during organismal aging and cellular senescence. As examples of organs where aging processes have recently been investigated, we chose to review the literature on molecular aging processes with involvement of Ca2+/CaM in heart and neuronal diseases, as well as in cancer and metabolic diseases, all deeply affected by aging. In addition, this article focuses on cellular senescence, a mechanism that may contribute to aging processes and therefore has been proposed as a target to interfere with the progression of age-associated diseases.
Collapse
Affiliation(s)
- Martin W Berchtold
- Department of Biology, University of Copenhagen, 13 Universitetsparken, DK-2100 Copenhagen Ø, Denmark.
| | - Antonio Villalobo
- Cancer and Human Molecular Genetics Area, Oto-Neurosurgery Research Group, University Hospital La Paz Research Institute (IdiPAZ), Paseo de la Castellana 261, E-28046 Madrid, Spain.
| |
Collapse
|
2
|
Zhang L, Liu G, Peng Y, Gao J, Tian M. Role of Neural Circuits in Cognitive Impairment. Neurochem Res 2024; 50:49. [PMID: 39644416 DOI: 10.1007/s11064-024-04309-3] [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: 10/06/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Cognitive impairment refers to abnormalities in learning, memory and cognitive judgment, mainly manifested as symptoms such as decreased memory, impaired orientation and reduced computational ability. As the fundamental unit of information processing in the brain, neural circuits have recently attracted great attention due to their functions in regulating pain, emotion and behavior. Furthermore, a growing number of studies have suggested that neural circuits play an important role in cognitive impairment. Neural circuits can affect perception, attention and decision-making, they can also regulate language skill, thinking and memory. Pathological conditions crucially affecting the integrity and preservation of neural circuits and their connectivity will heavily impact cognitive abilities. Nowadays, technological developments have led to many novel methods for studying neural circuits, such as brain imaging, optogenetic techniques, and chemical genetics approaches. Therefore, neural circuits show great promise as a potential target in mitigating cognitive impairment. In this review we discuss the pathogenesis of cognitive impairment and the regulation and detection of neural circuits, thus highlighting the role of neural circuits in cognitive impairment. Hence, therapeutic agents against cognitive impairment may be developed that target neural circuits important in cognition.
Collapse
Affiliation(s)
- Li Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Guodong Liu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Yaonan Peng
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, PR China
| | - Jinqi Gao
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, PR China
| | - Mi Tian
- Department of Anesthesiology, Surgery and Pain Management, Zhongda Hospital, the School of Medicine, Southeast University, Nanjing, Jiangsu Province, PR China.
| |
Collapse
|
3
|
Chen Y, Chen J, Liang L, Dai W, Li N, Dong S, Zhan Y, Chen G, Yu Y. Compound heterozygous mutations of NTNG2 cause intellectual disability via inhibition of the CaMKII signaling. J Genet Genomics 2024; 51:1204-1214. [PMID: 39151821 DOI: 10.1016/j.jgg.2024.08.001] [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: 05/22/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
Netrin-G2 is a membrane-anchored protein known to play critical roles in neuronal circuit development and synaptic organization. In this study, we identify compound heterozygous mutations of c.547delC, p.(Arg183Alafs∗186) and c.605G>A, p.(Trp202X) in NTNG2 causing a syndrome exhibiting developmental delay, intellectual disability, hypotonia, and facial dysmorphism. To elucidate the underlying cellular and molecular mechanisms, CRISPR-Cas9 technology is employed to generate a knock-in mouse model expressing the R183Afs and W202X mutations. We report that the Ntng2R183Afs/W202X mice exhibit hypotonia and impaired learning and memory. We find that the levels of CaMKII and p-GluA1Ser831 are decreased, and excitatory postsynaptic transmission and long-term potentiation are impaired. To increase the activity of CaMKII, the mutant mice receive intraperitoneal injections of DCP-LA, a CaMKII agonist, and show improved cognitive function. Together, our findings reveal molecular mechanisms of how NTNG2 deficiency leads to impairments of cognitive ability and synaptic plasticity.
Collapse
Affiliation(s)
- Yaoting Chen
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Jiang Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210008, China
| | - Lili Liang
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Weiqian Dai
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Nan Li
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Shuangshuang Dong
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Yongkun Zhan
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China.
| | - Guiquan Chen
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210061, China.
| | - Yongguo Yu
- Department of Pediatric Endocrinology and Genetics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai 200092, China.
| |
Collapse
|
4
|
Popescu BO, Batzu L, Ruiz PJG, Tulbă D, Moro E, Santens P. Neuroplasticity in Parkinson's disease. J Neural Transm (Vienna) 2024; 131:1329-1339. [PMID: 39102007 PMCID: PMC11502561 DOI: 10.1007/s00702-024-02813-y] [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: 07/06/2024] [Accepted: 07/22/2024] [Indexed: 08/06/2024]
Abstract
Parkinson's disease (PD) is the second most frequent neurodegenerative disorder, affecting millions of people and rapidly increasing over the last decades. Even though there is no intervention yet to stop the neurodegenerative pathology, many efficient treatment methods are available, including for patients with advanced PD. Neuroplasticity is a fundamental property of the human brain to adapt both to external changes and internal insults and pathological processes. In this paper we examine the current knowledge and concepts concerning changes at network level, cellular level and molecular level as parts of the neuroplastic response to protein aggregation pathology, synapse loss and neuronal loss in PD. We analyse the beneficial, compensatory effects, such as augmentation of nigral neurons efficacy, as well as negative, maladaptive effects, such as levodopa-induced dyskinesia. Effects of physical activity and different treatments on neuroplasticity are considered and the opportunity of biomarkers identification and use is discussed.
Collapse
Affiliation(s)
- Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, 'Carol Davila' University of Medicine and Pharmacy Bucharest, Bucharest, Romania.
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, 'Victor Babeș' National Institute of Pathology, Bucharest, Romania.
| | - Lucia Batzu
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, The Maurice Wohl Clinical Neuroscience Institute, King's College London, London, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital, London, UK
| | | | - Delia Tulbă
- Department of Clinical Neurosciences, 'Carol Davila' University of Medicine and Pharmacy Bucharest, Bucharest, Romania
| | - Elena Moro
- Division of Neurology, Centre Hospitalier Universitaire de Grenoble, Grenoble Alpes University, Grenoble Institute of Neuroscience, INSERM U1216, Grenoble, France
| | - Patrick Santens
- Department of Neurology, University Hospital Ghent, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| |
Collapse
|
5
|
Chen L, Li Y, Liu M, Lan Z, Zhang X, Yang X, Zhao Q, Wang S, Xu L, Zhou Y, Kuang Y, Suzuki T, Tabuchi K, Takahashi E, Zhou M, Chen CD, Xu T, Li W. Desipramine reverses remote memory deficits by activating calmodulin-CaMKII pathway in a UTX knockout mouse model of Kabuki syndrome. Gen Psychiatr 2024; 37:e101430. [PMID: 39493372 PMCID: PMC11529476 DOI: 10.1136/gpsych-2023-101430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 06/12/2024] [Indexed: 11/05/2024] Open
Abstract
Background Kabuki syndrome (KS) is a rare developmental disorder characterised by multiple congenital anomalies and intellectual disability. UTX (ubiquitously transcribed tetratricopeptide repeat, X chromosome), which encodes a histone demethylase, is one of the two major pathogenic risk genes for KS. Although intellectual disability is a key phenotype of KS, the role of UTX in cognitive function remains unclear. Currently, no targeted therapies are available for KS. Aims This study aimed to investigate how UTX regulates cognition, to explore the mechanisms underlying UTX dysfunction and to identify potential molecular targets for treatment. Methods We generated UTX conditional knockout mice and found that UTX deletion downregulated calmodulin transcription by disrupting H3K27me3 (trimethylated histone H3 at lysine 27) demethylation. Results UTX-knockout mice showed decreased phosphorylation of calcium / calmodulin-dependent protein kinase II, impaired long-term potentiation and deficit in remote contextual fear memory. These effects were reversed by an Food and Drug Administration-approved drug desipramine. Conclusions Our results reveal an epigenetic mechanism underlying the important role of UTX in synaptic plasticity and cognitive function, and suggest that desipramine could be a potential treatment for KS.
Collapse
Affiliation(s)
- Lei Chen
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yuting Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Minggang Liu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Mental Health and Drug Discovery, Oujiang Laboratory, Wenzhou, Zhejiang, China
| | - Zhaohui Lan
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Zhang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Xiujuan Yang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Zhao
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Shuai Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Longyong Xu
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ying Zhou
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Yifang Kuang
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Tatsuo Suzuki
- Shinshu University School of Medicine, Nagano, Japan
| | | | - Eiki Takahashi
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
| | - Miou Zhou
- Western University of Health Sciences, Pomona, California, USA
| | - Charlie Degui Chen
- Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tianle Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weidong Li
- Bio-X Institutes, Key Laboratory for the Genetics of Development and Neuropsychiatric Disorders (Ministry of Education), Brain Health and Brain Technology Center at Global Institute of Future Technology, Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China
- WLA Laboratories, World Laureates Association, Shanghai, China
| |
Collapse
|
6
|
Song Q, E S, Zhang Z, Liang Y. Neuroplasticity in the transition from acute to chronic pain. Neurotherapeutics 2024; 21:e00464. [PMID: 39438166 PMCID: PMC11585895 DOI: 10.1016/j.neurot.2024.e00464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/25/2024] Open
Abstract
Acute pain is a transient sensation that typically serves as part of the body's defense mechanism. However, in certain patients, acute pain can evolve into chronic pain, which persists for months or even longer. Neuroplasticity refers to the capacity for variation and adaptive alterations in the morphology and functionality of neurons and synapses, and it plays a significant role in the transmission and modulation of pain. In this paper, we explore the molecular mechanisms and signaling pathways underlying neuroplasticity during the transition of pain. We also examine the effects of neurotransmitters, inflammatory mediators, and central sensitization on neuroplasticity, as well as the potential of neuroplasticity as a therapeutic strategy for preventing chronic pain. The aims of this article is to clarify the role of neuroplasticity in the transformation from acute pain to chronic pain, with the hope of providing a novel theoretical basis for unraveling the pathogenesis of chronic pain and offering more effective strategies and approaches for its diagnosis and treatment.
Collapse
Affiliation(s)
- Qingbiao Song
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China
| | - Sihan E
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China
| | - Zhiyu Zhang
- Department of Orthopedics, Affiliated Hospital of Shandong Second Medical University, Weifang 261035, China
| | - Yingxia Liang
- School of Anesthesiology, Shandong Second Medical University, Weifang 261053, China.
| |
Collapse
|
7
|
Zhang L, Zhou Y, Xie Y, Ying Y, Li Y, Ye S, Wang Z. Adjunctive clozapine with bright light mitigates cognitive deficits by synaptic plasticity and neurogenesis in sub-chronic MK-801 treated mice. Pharmacol Biochem Behav 2024; 243:173821. [PMID: 39002805 DOI: 10.1016/j.pbb.2024.173821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Schizophrenia impacts about 1 % of the global population, with clozapine (CLZ) being a critical treatment for refractory cases despite its limitations in effectiveness and adverse effects. Therefore, the search for more effective treatments remains urgent. Light treatment (LT) recognized for enhancing cognition and mood, presents a promising complementary approach. This study investigated the effects of CLZ and LT on cognitive impairments in a sub-chronic MK-801 induced schizophrenia mouse model. Results showed that both CLZ and CLZ + LT treatment elevate cognitive performance of sub-chronic MK-801 treated mice in serial behavioral tests over two months. Histological analysis revealed increased dendritic spine density and branching, and synaptic repair in the hippocampus with CLZ and CLZ + LT interventions. Furthermore, both treatments increased brain-derived neurotrophic factor (BDNF) expression in the hippocampus, likely contributing to cognitive amelioration in MK-801 treated mice. Additionally, BrdU labeling revealed that CLZ + LT further enhances neurogenesis in the dentate gyrus (DG) and lateral ventricle (LV) of sub-chronic MK-801 treated mice. These findings may have implications for the development of noninvasive and adjunctive treatment strategies aimed at alleviating cognitive impairments and improving functional outcomes in individuals with schizophrenia.
Collapse
Affiliation(s)
- Lizhi Zhang
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo 315010, China
| | - Yiying Zhou
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo 315010, China
| | - Yanhong Xie
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo 315010, China
| | - Yudong Ying
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo 315010, China
| | - Yan Li
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo 315010, China
| | - Sen Ye
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo 315010, China
| | - Zhengchun Wang
- Zhejiang Key Laboratory of Pathophysiology, Basic Medical Sciences, Health Science Center, Ningbo University, 818 Fenghua Rd, Ningbo, Zhejiang 315211, China; The Affiliated People's Hospital of Ningbo University, Ningbo 315100, China; Key Laboratory of Addiction Research of Zhejiang Province, Kang Ning Hospital, Ningbo 315010, China.
| |
Collapse
|
8
|
Logan R, Shelton M, Horan N, Xue X, Maturin L, Eacret D, Michaud J, Singh N, Williams B, Gamble M, Seggio J, Kuppe-Fish M, Phan B, Tseng G, Blendy J, Woods LS, Palmer A, George O, Seney M. Sex-specific Concordance of Striatal Transcriptional Signatures of Opioid Addiction in Human and Rodent Brains. RESEARCH SQUARE 2024:rs.3.rs-5006061. [PMID: 39399686 PMCID: PMC11469374 DOI: 10.21203/rs.3.rs-5006061/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Opioid use disorder (OUD) has emerged as a severe, ongoing public health emergency. Current, frontline addiction treatment strategies fail to produce lasting abstinence in most users. This underscores the lasting effects of chronic opioid exposure and emphasizes the need to understand the molecular mechanisms of drug seeking and taking, but also how those alterations persist through acute and protracted withdrawal. Here, we used RNA sequencing in post-mortem human tissue from males (n=10) and females (n=10) with OUD and age and sex-matched comparison subjects. We compared molecular alterations in the nucleus accumbens (NAc) and dorsolateral prefrontal cortex (DLPFC) between humans with OUD and rodent models across distinct stages of opioid use and withdrawal (acute and prolonged) using differential gene expression and network-based approaches. We found that the molecular signature in the NAc of females with OUD mirrored effects seen in the NAc of female mice at all stages of exposure. Conversely, males with OUD showed strong overlap in expression profile with rats in acute withdrawal. Co-expression networks involved in post-transcriptional modification of RNA and epigenetic modification of chromatin state. This study provides fundamental insight into the converging molecular pathways altered by opioids across species. Further, this work helps to disentangle which alterations observed in humans with OUD are driven by acute drug exposure and which alterations are consequences of chronic exposure.
Collapse
Affiliation(s)
- Ryan Logan
- University of Massachusetts Chan Medical School
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Funahashi Y, Ahammad RU, Zhang X, Hossen E, Kawatani M, Nakamuta S, Yoshimi A, Wu M, Wang H, Wu M, Li X, Faruk MO, Shohag MH, Lin YH, Tsuboi D, Nishioka T, Kuroda K, Amano M, Noda Y, Yamada K, Sakimura K, Nagai T, Yamashita T, Uchino S, Kaibuchi K. Signal flow in the NMDA receptor-dependent phosphoproteome regulates postsynaptic plasticity for aversive learning. Sci Signal 2024; 17:eado9852. [PMID: 39255336 DOI: 10.1126/scisignal.ado9852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/21/2024] [Indexed: 09/12/2024]
Abstract
Structural plasticity of dendritic spines in the nucleus accumbens (NAc) is crucial for learning from aversive experiences. Activation of NMDA receptors (NMDARs) stimulates Ca2+-dependent signaling that leads to changes in the actin cytoskeleton, mediated by the Rho family of GTPases, resulting in postsynaptic remodeling essential for learning. We investigated how phosphorylation events downstream of NMDAR activation drive the changes in synaptic morphology that underlie aversive learning. Large-scale phosphoproteomic analyses of protein kinase targets in mouse striatal/accumbal slices revealed that NMDAR activation resulted in the phosphorylation of 194 proteins, including RhoA regulators such as ARHGEF2 and ARHGAP21. Phosphorylation of ARHGEF2 by the Ca2+-dependent protein kinase CaMKII enhanced its RhoGEF activity, thereby activating RhoA and its downstream effector Rho-associated kinase (ROCK/Rho-kinase). Further phosphoproteomic analysis identified 221 ROCK targets, including the postsynaptic scaffolding protein SHANK3, which is crucial for its interaction with NMDARs and other postsynaptic scaffolding proteins. ROCK-mediated phosphorylation of SHANK3 in the NAc was essential for spine growth and aversive learning. These findings demonstrate that NMDAR activation initiates a phosphorylation cascade crucial for learning and memory.
Collapse
Affiliation(s)
- Yasuhiro Funahashi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Rijwan Uddin Ahammad
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA 92121, USA
| | - Xinjian Zhang
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Emran Hossen
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Masahiro Kawatani
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Akira Yoshimi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Division of Clinical Sciences and Neuropsychopharmacology, Faculty and Graduate School of Pharmacy, Meijo University, Nagoya, Aichi 468-8503, Japan
| | - Minhua Wu
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Huanhuan Wang
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Mengya Wu
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Xu Li
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Md Omar Faruk
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Md Hasanuzzaman Shohag
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - You-Hsin Lin
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Daisuke Tsuboi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Tomoki Nishioka
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Keisuke Kuroda
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Mutsuki Amano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yukihiko Noda
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Division of Clinical Sciences and Neuropsychopharmacology, Faculty and Graduate School of Pharmacy, Meijo University, Nagoya, Aichi 468-8503, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kenji Sakimura
- Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Takayuki Yamashita
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
- Division of Neurophysiology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Shigeo Uchino
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi 320-8551, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
10
|
Ma Y, He C, Lin W, Wang J, Xu C, Pan D, Wang Z, Yao W, Dong R, Jia D, Li K. CAMK2G Promotes Neuronal Differentiation and Inhibits Migration in Neuroblastoma. J Pediatr Surg 2024:161679. [PMID: 39266386 DOI: 10.1016/j.jpedsurg.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024]
Abstract
PURPOSE Neuroblastoma (NB) originates from differentiation arrest of sympathoadrenal progenitors in the neural crest. It is necessary to reveal the differentiation mechanism of NB. Previously, we reported that Purkinje cell protein 4 (PCP4) is a well-differentiated marker of NB tissues. Herein, we explored the underlying mechanism of PCP4 induced differentiation in order to find better treatment options for patients. METHODS We screened the interacting proteins of PCP4 by co-immunoprecipitation (Co-IP) and liquid chromatography-mass spectrometry (LC-MS/MS). Then we investigated the relevance between expression of calmodulin-dependent protein kinase II gamma (CAMK2G) and clinical features using R2 platform. We also explored the function of CAMK2G in NB cells by knockdown and RNA sequencing. RESULTS Here, we verified the binding of PCP4 and calmodulin (CaM) by Co-IP and identified a target kinase of CaM, CAMK2G, by LC-MS/MS. PCP4 overexpression activates the autophosphorylation of CAMK2G. Patients with high CAMK2G expression had better survival while low CAMK2G was associated with unfavorable clinical features including MYCN-amplification, unfavorable histology, progression and high INSS stage. CAMK2G knockdown inhibited neurite outgrowth and down-regulated neuronal differentiation markers (NF-H, MAP2), yet promoted migration, invasion and proliferation. Gene Ontology (GO) analysis showed that knockdown of CAMK2G downregulated the expression of neuronal differentiation-related genes. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that knockdown of CAMK2G upregulated the expression of migration-related genes. CONCLUSION These findings indicate that CAMK2G activated by PCP4/CaM complex promotes differentiation and inhibits migration in NB cells. LEVEL OF EVIDENCE Not applicable.
Collapse
Affiliation(s)
- Yujie Ma
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Cong He
- Laboratory of Cancer Genomics and Biology, Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China; Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai 201321, China
| | - Weihong Lin
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Jing Wang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Chaoliang Xu
- Laboratory of Cancer Genomics and Biology, Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Deshen Pan
- Laboratory of Cancer Genomics and Biology, Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zuopeng Wang
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Wei Yao
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Rui Dong
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China
| | - Deshui Jia
- Laboratory of Cancer Genomics and Biology, Department of Thoracic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Kai Li
- Department of Pediatric Surgery, Children's Hospital of Fudan University, National Children's Medical Center, 399 Wanyuan Road, Shanghai 201102, China.
| |
Collapse
|
11
|
Xie S, Zhu M, Zhu H, Wang W. Effects of β-asarone on spatial learning and memory impairment by exhaustive exercise-induced fatigue: Role of NR-CaMKII-ERK/CREB signal in hippocampus of rats. Behav Brain Res 2024; 471:115076. [PMID: 38825021 DOI: 10.1016/j.bbr.2024.115076] [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: 01/25/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024]
Abstract
OBJECTIVE It is to investigate the effects of β-asarone on learning and memory, hippocampal morphology, synaptophysin (SYP) and postsynaptic density 95(PSD95) protein expression, N-methyl-D-aspartic acid receptor 2B (NR2B)- Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) - Extracellular signal-regulated kinase (ERK) / Cyclic-AMP response element binding protein (CREB) signal in hippocampus of rats with exhaustive exercise-induced fatigue. METHODS Fifty Sprague-Dawley male rats were randomly divided into five groups: normal group, exercise group, exercise and β-asarone (2.5, 10, 40 mg/kg)-treated groups. The learning and memory in rats were tested by Morris water maze experiment. We measured the hippocampal morphology by Nissl staining. The levels of SYP, PSD95, NR2B, CaMKII, ERK1/2, CREB, p-NR2B, p-CaMKII, p-ERK1/2 and p-CREB expression were measured by western blot analysis. RESULTS The results demonstrated that β-asarone (10, 40 mg/kg) treatment significantly decreased the latency to find the platform, increased the time spent in the target quadrant and the number of crossing the platform of rats with exhaustive exercise-induced fatigue. β-asarone (10, 40 mg/kg) treatment increased the cell density in the hippocampus CA1 region, significantly up-regulated NR2B-CaMKII-ERK/CREB signal and improved the protein expression levels of SYP and PSD95 in hippocampus of rats with exhaustive exercise-induced fatigue. CONCLUSIONS It suggests that β-asarone could improve learning and memory of rats with exhaustive exercise-induced fatigue. The mechanism might be related to β-asarone protecting the morphology of hippocampus, increasing the protein expression levels of SYP and PSD95 and up-regulating NR2B-CaMKII-ERK/CREB signal in hippocampus of rats with exhaustive exercise-induced fatigue.
Collapse
Affiliation(s)
- Shifeng Xie
- School of Sports Science, Jinggangshan University, Jian, Jiangxi 343009, China
| | - Meiju Zhu
- School of Sports Science, Jinggangshan University, Jian, Jiangxi 343009, China.
| | - Hongzhu Zhu
- School of Sports Science, Jinggangshan University, Jian, Jiangxi 343009, China
| | - Wei Wang
- School of Sports Science, Jinggangshan University, Jian, Jiangxi 343009, China
| |
Collapse
|
12
|
Tan YQ, Zhang W, Xie ZC, Li J, Chen HW. CaMK II in Cardiovascular Diseases, Especially CaMK II-δ: Friends or Enemies. Drug Des Devel Ther 2024; 18:3461-3476. [PMID: 39132626 PMCID: PMC11314529 DOI: 10.2147/dddt.s473251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/05/2024] [Indexed: 08/13/2024] Open
Abstract
Cardiovascular diseases (CVDs) tend to affect the young population and are associated with a significant economic burden and psychological distress to the society and families. The physiological and pathological processes underlying CVDs are complex. Ca2+/calmodulin-dependent kinase II (CaMK II), a protein kinase, has multiple biological functions. It participates in multiple pathological processes and plays a central role in the development of CVDs. Based on this, this paper analyzes the structural characteristics and distribution of CaMK II, the mechanism of action of CaMK II, and the relationship between CaMK II and CVDs, including ion channels, ischemia-reperfusion injury, arrhythmias, myocardial hypertrophy, cardiotoxicity, hypertension, and dilated cardiomyopathy. Given the different regulatory mechanisms of different isoforms of CaMK II, the clinical use of specific targeted inhibitors or novel compounds should be evaluated in future research to provide new directions.
Collapse
Affiliation(s)
- Yu-Qing Tan
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Wang Zhang
- Department of Pharmacy, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Zi-Cong Xie
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Jun Li
- Department of Cardiology, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| | - Heng-Wen Chen
- New Drug Research and Development Office, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, People’s Republic of China
| |
Collapse
|
13
|
Hagena H, Manahan-Vaughan D. Interplay of hippocampal long-term potentiation and long-term depression in enabling memory representations. Philos Trans R Soc Lond B Biol Sci 2024; 379:20230229. [PMID: 38853558 PMCID: PMC11343234 DOI: 10.1098/rstb.2023.0229] [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: 01/08/2024] [Revised: 04/22/2024] [Accepted: 05/07/2024] [Indexed: 06/11/2024] Open
Abstract
Hippocampal long-term potentiation (LTP) and long-term depression (LTD) are Hebbian forms of synaptic plasticity that are widely believed to comprise the physiological correlates of associative learning. They comprise a persistent, input-specific increase or decrease, respectively, in synaptic efficacy that, in rodents, can be followed for days and weeks in vivo. Persistent (>24 h) LTP and LTD exhibit distinct frequency-dependencies and molecular profiles in the hippocampal subfields. Moreover, causal and genetic studies in behaving rodents indicate that both LTP and LTD fulfil specific and complementary roles in the acquisition and retention of spatial memory. LTP is likely to be responsible for the generation of a record of spatial experience, which may serve as an associative schema that can be re-used to expedite or facilitate subsequent learning. In contrast, LTD may enable modification and dynamic updating of this representation, such that detailed spatial content information is included and the schema is rendered unique and distinguishable from other similar representations. Together, LTP and LTD engage in a dynamic interplay that supports the generation of complex associative memories that are resistant to generalization. This article is part of a discussion meeting issue 'Long-term potentiation: 50 years on'.
Collapse
Affiliation(s)
- Hardy Hagena
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Bochum44780, Germany
| | - Denise Manahan-Vaughan
- Medical Faculty, Department of Neurophysiology, Ruhr University Bochum, Bochum44780, Germany
| |
Collapse
|
14
|
Bartol TM, Ordyan M, Sejnowski TJ, Rangamani P, Kennedy MB. A spatial model of autophosphorylation of Ca 2+ /calmodulin-dependent protein kinase II in a glutamatergic spine reveals dynamics of kinase activation in the first several seconds after a complex synaptic stimulus. 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 in excitatory glutamatergic synapses in the Central Nervous System (CNS). It is initiated by a bout of synaptic activation that is strong enough to contribute to production of an action potential in the axon of the postsynaptic neuron, and it results in an increase in the size of postsynaptic depolarization during subsequent activity. The first step leading to LTP is activation and autophosphorylation of an abundant postsynaptic enzyme, Ca 2+ /calmodulin-dependent protein kinase II (CaMKII). We use simulation of activation of CaMKII holoenzymes in a realistic spatial model of a spine synapse, created in MCell4, to test three hypotheses about how the autophosphorylation response of CaMKII is shaped during a repeated high-frequency stimulus. First, the simulation results indicate that autophosphorylation of CaMKII does not constitute a bistable switch under biologically realistic conditions. Instead, prolonged autophosphorylation of CaMKII may contribute to a biochemical "kinetic proof-reading" mechanism that controls induction of synaptic plasticity. Second, concentration of CaMKII near the postsynaptic membrane increases the local concentration of kinase activity. However, neither localization nor "Ca 2+ -calmodulin-trapping (CaM-trapping)" increase the proportion of autophosphorylated subunits in holoenzymes after a complex stimulus, as previously hypothesized. Finally, we show that, as hypothesized, the amplitude of autophosphorylation in the first 30 seconds after a stimulus is extremely sensitive to the level and location of PP1 activity when PP1 is present in biologically accurate amounts. We further show that prolonged steric hindrance of dephosphorylation of CaMKII, caused by CaM-trapping, can increase the amplitude of autophosphorylation after a complex stimulus. These simulation results sharpen our quantitative understanding of the early events leading to LTP at excitatory synapses. Author Summary Neurons in the brain are interconnected in an organized fashion by synapses that transmit neuronal activity from one neuron to another. Most of the billions of neurons in the brain have about 10,000 synapses spread over the neuronal membrane. Information is stored in the brain when the ability of specific synapses to pass along neuronal activity is strengthened resulting in formation of new networks. The increase in strength of a synapse is tightly controlled by the frequency and amplitude of its activity, and by neurohormonal signals, which, in combination, can cause long-lasting biochemical changes at the synapse that underlie learning and memory. Defects in these biochemical pathways cause mental and neurological diseases. To develop treatments, we need to understand the precise choreography of these critical biochemical changes. However, the tiny size of the synaptic compartment makes precise measurements of the biochemical reactions impossible. We have used computer simulation techniques and information gathered from experiments on purified synaptic proteins to simulate, within a single synapse, the choreography of the first biochemical step in synaptic strengthening: activation of the enzyme Ca 2+ / calmodulin-dependent protein kinase II. Our results provide insights that can be used in future studies to develop treatments for neuronal diseases.
Collapse
|
15
|
Anjum R, Clarke VRJ, Nagasawa Y, Murakoshi H, Paradis S. Rem2 interacts with CaMKII at synapses and restricts long-term potentiation in hippocampus. PLoS One 2024; 19:e0301063. [PMID: 38995900 PMCID: PMC11244776 DOI: 10.1371/journal.pone.0301063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/11/2024] [Indexed: 07/14/2024] Open
Abstract
Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme calcium/calmodulin-dependent protein kinase II (CaMKII) plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.
Collapse
Affiliation(s)
- Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts, United States of America
| | - Vernon R. J. Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Yutaro Nagasawa
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi, Japan
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts, United States of America
| |
Collapse
|
16
|
Caya-Bissonnette L, Béïque JC. Half a century legacy of long-term potentiation. Curr Biol 2024; 34:R640-R662. [PMID: 38981433 DOI: 10.1016/j.cub.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
In 1973, two papers from Bliss and Lømo and from Bliss and Gardner-Medwin reported that high-frequency synaptic stimulation in the dentate gyrus of rabbits resulted in a long-lasting increase in synaptic strength. This form of synaptic plasticity, commonly referred to as long-term potentiation (LTP), was immediately considered as an attractive mechanism accounting for the ability of the brain to store information. In this historical piece looking back over the past 50 years, we discuss how these two landmark contributions directly motivated a colossal research effort and detail some of the resulting milestones that have shaped our evolving understanding of the molecular and cellular underpinnings of LTP. We highlight the main features of LTP, cover key experiments that defined its induction and expression mechanisms, and outline the evidence supporting a potential role of LTP in learning and memory. We also briefly explore some ramifications of LTP on network stability, consider current limitations of LTP as a model of associative memory, and entertain future research orientations.
Collapse
Affiliation(s)
- Léa Caya-Bissonnette
- Graduate Program in Neuroscience, University of Ottawa, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada; Brain and Mind Research Institute's Centre for Neural Dynamics and Artificial Intelligence, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada
| | - Jean-Claude Béïque
- Brain and Mind Research Institute's Centre for Neural Dynamics and Artificial Intelligence, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada; Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, 451 ch. Smyth Road (3501N), Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
17
|
Stockwell I, Watson JF, Greger IH. Tuning synaptic strength by regulation of AMPA glutamate receptor localization. Bioessays 2024; 46:e2400006. [PMID: 38693811 PMCID: PMC7616278 DOI: 10.1002/bies.202400006] [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: 01/10/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
Long-term potentiation (LTP) of excitatory synapses is a leading model to explain the concept of information storage in the brain. Multiple mechanisms contribute to LTP, but central amongst them is an increased sensitivity of the postsynaptic membrane to neurotransmitter release. This sensitivity is predominantly determined by the abundance and localization of AMPA-type glutamate receptors (AMPARs). A combination of AMPAR structural data, super-resolution imaging of excitatory synapses, and an abundance of electrophysiological studies are providing an ever-clearer picture of how AMPARs are recruited and organized at synaptic junctions. Here, we review the latest insights into this process, and discuss how both cytoplasmic and extracellular receptor elements cooperate to tune the AMPAR response at the hippocampal CA1 synapse.
Collapse
Affiliation(s)
- Imogen Stockwell
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Jake F. Watson
- Institute of Science and Technology, Technology (IST) Austria, Klosterneuburg, Austria
| | - Ingo H. Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
18
|
Chen X, Cai Q, Zhou J, Pleasure SJ, Schulman H, Zhang M, Nicoll RA. CaMKII autophosphorylation is the only enzymatic event required for synaptic memory. Proc Natl Acad Sci U S A 2024; 121:e2402783121. [PMID: 38889145 PMCID: PMC11214084 DOI: 10.1073/pnas.2402783121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
Ca2+/calmodulin (CaM)-dependent kinase II (CaMKII) plays a critical role in long-term potentiation (LTP), a well-established model for learning and memory through the enhancement of synaptic transmission. Biochemical studies indicate that CaMKII catalyzes a phosphotransferase (kinase) reaction of both itself (autophosphorylation) and of multiple downstream target proteins. However, whether either type of phosphorylation plays any role in the synaptic enhancing action of CaMKII remains hotly contested. We have designed a series of experiments to define the minimal requirements for the synaptic enhancement by CaMKII. We find that autophosphorylation of T286 and further binding of CaMKII to the GluN2B subunit are required both for initiating LTP and for its maintenance (synaptic memory). Once bound to the NMDA receptor, the synaptic action of CaMKII occurs in the absence of target protein phosphorylation. Thus, autophosphorylation and binding to the GluN2B subunit are the only two requirements for CaMKII in synaptic memory.
Collapse
Affiliation(s)
- Xiumin Chen
- Department of Neurology and Institute of Neuroscience of Soochow University, Second Affiliated Hospital of Soochow University, Suzhou215004, China
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA94158
| | - Qixu Cai
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Department of Laboratory Medicine, State Key Laboratory of Vaccines for Infectious Diseases,School of Public Heath, Xiamen University, Xiamen, Fujian361102, China
| | - Jing Zhou
- Department of Neurology, University of California, San Francisco, CA94158
| | - Samuel J. Pleasure
- Department of Neurology, University of California, San Francisco, CA94158
| | - Howard Schulman
- Department of Pharmacology, Stanford University School of Medicine, Stanford, CA
- Department of Pharmacology, Panorama Research Institute, Sunnyvale, CA
| | - 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
- Department of Laboratory Medicine, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong518055, China
| | - Roger A. Nicoll
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA94158
| |
Collapse
|
19
|
Chien CT, Puhl H, Vogel SS, Molloy JE, Chiu W, Khan S. Hub stability in the calcium calmodulin-dependent protein kinase II. Commun Biol 2024; 7:766. [PMID: 38918547 PMCID: PMC11199487 DOI: 10.1038/s42003-024-06423-y] [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: 06/20/2023] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
The calcium calmodulin protein kinase II (CaMKII) is a multi-subunit ring assembly with a central hub formed by the association domains. There is evidence for hub polymorphism between and within CaMKII isoforms, but the link between polymorphism and subunit exchange has not been resolved. Here, we present near-atomic resolution cryogenic electron microscopy (cryo-EM) structures revealing that hubs from the α and β isoforms, either standalone or within an β holoenzyme, coexist as 12 and 14 subunit assemblies. Single-molecule fluorescence microscopy of Venus-tagged holoenzymes detects intermediate assemblies and progressive dimer loss due to intrinsic holoenzyme lability, and holoenzyme disassembly into dimers upon mutagenesis of a conserved inter-domain contact. Molecular dynamics (MD) simulations show the flexibility of 4-subunit precursors, extracted in-silico from the β hub polymorphs, encompassing the curvature of both polymorphs. The MD explains how an open hub structure also obtained from the β holoenzyme sample could be created by dimer loss and analysis of its cryo-EM dataset reveals how the gap could open further. An assembly model, considering dimer concentration dependence and strain differences between polymorphs, proposes a mechanism for intrinsic hub lability to fine-tune the stoichiometry of αβ heterooligomers for their dynamic localization within synapses in neurons.
Collapse
Affiliation(s)
- Chih-Ta Chien
- Department of Bioengineering, and Department of Microbiology and Immunology, James H. Clark Center, Stanford University, Stanford, CA, 94305, USA
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institute of Health, Bethesda, MD, 20892, USA
| | - Henry Puhl
- Laboratory of Biophotonics and Quantum Biology, National Institutes on Alcohol, Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 208952, USA
| | - Steven S Vogel
- Laboratory of Biophotonics and Quantum Biology, National Institutes on Alcohol, Abuse and Alcoholism, National Institutes of Health, Rockville, MD, 208952, USA
| | - Justin E Molloy
- The Francis Crick Institute, London, UK
- CMCB, Warwick Medical School, Coventry, CV4 7AL, UK
| | - Wah Chiu
- Department of Bioengineering, and Department of Microbiology and Immunology, James H. Clark Center, Stanford University, Stanford, CA, 94305, USA.
- CryoEM and Bioimaging Division, Stanford Synchrotron Radiation Light source, SLAC National Accelerator Laboratory, Stanford University, Menlo Park, CA, 94025, USA.
| | - Shahid Khan
- Molecular Biology Consortium @ Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
20
|
Habecker BA, Bers DM, Birren SJ, Chang R, Herring N, Kay MW, Li D, Mendelowitz D, Mongillo M, Montgomery JM, Ripplinger CM, Tampakakis E, Winbo A, Zaglia T, Zeltner N, Paterson DJ. Molecular and cellular neurocardiology in heart disease. J Physiol 2024:10.1113/JP284739. [PMID: 38778747 PMCID: PMC11582088 DOI: 10.1113/jp284739] [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: 02/23/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024] Open
Abstract
This paper updates and builds on a previous White Paper in this journal that some of us contributed to concerning the molecular and cellular basis of cardiac neurobiology of heart disease. Here we focus on recent findings that underpin cardiac autonomic development, novel intracellular pathways and neuroplasticity. Throughout we highlight unanswered questions and areas of controversy. Whilst some neurochemical pathways are already demonstrating prognostic viability in patients with heart failure, we also discuss the opportunity to better understand sympathetic impairment by using patient specific stem cells that provides pathophysiological contextualization to study 'disease in a dish'. Novel imaging techniques and spatial transcriptomics are also facilitating a road map for target discovery of molecular pathways that may form a therapeutic opportunity to treat cardiac dysautonomia.
Collapse
Affiliation(s)
- Beth A Habecker
- Department of Chemical Physiology & Biochemistry, Department of Medicine Knight Cardiovascular Institute, Oregon Health and Science University, Portland, OR, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | - Susan J Birren
- Department of Biology, Volen Center for Complex Systems, Brandeis University, Waltham, MA, USA
| | - Rui Chang
- Department of Neuroscience, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Neil Herring
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Matthew W Kay
- Department of Biomedical Engineering, George Washington University, Washington, DC, USA
| | - Dan Li
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - David Mendelowitz
- Department of Pharmacology and Physiology, George Washington University, Washington, DC, USA
| | - Marco Mongillo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Johanna M Montgomery
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Crystal M Ripplinger
- Department of Pharmacology, University of California, Davis School of Medicine, Davis, CA, USA
| | | | - Annika Winbo
- Department of Physiology and Manaaki Manawa Centre for Heart Research, University of Auckland, Auckland, New Zealand
| | - Tania Zaglia
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Nadja Zeltner
- Departments of Biochemistry and Molecular Biology, Cell Biology, and Center for Molecular Medicine, University of Georgia, Athens, GA, USA
| | - David J Paterson
- Burdon Sanderson Cardiac Science Centre and BHF Centre of Research Excellence, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
21
|
Chang X, Zhang H, Chen S. Neural circuits regulating visceral pain. Commun Biol 2024; 7:457. [PMID: 38615103 PMCID: PMC11016080 DOI: 10.1038/s42003-024-06148-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/05/2024] [Indexed: 04/15/2024] Open
Abstract
Visceral hypersensitivity, a common clinical manifestation of irritable bowel syndrome, may contribute to the development of chronic visceral pain, which is a major challenge for both patients and health providers. Neural circuits in the brain encode, store, and transfer pain information across brain regions. In this review, we focus on the anterior cingulate cortex and paraventricular nucleus of the hypothalamus to highlight the progress in identifying the neural circuits involved in visceral pain. We also discuss several neural circuit mechanisms and emphasize the importance of cross-species, multiangle approaches and the identification of specific neurons in determining the neural circuits that control visceral pain.
Collapse
Affiliation(s)
- Xiaoli Chang
- College of Acupuncture and Massage, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Haiyan Zhang
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shaozong Chen
- Research Institute of Acupuncture and Moxibustion, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
22
|
Kakizawa S, Arasaki T, Yoshida A, Sato A, Takino Y, Ishigami A, Akaike T, Yanai S, Endo S. Essential role of ROS - 8-Nitro-cGMP signaling in long-term memory of motor learning and cerebellar synaptic plasticity. Redox Biol 2024; 70:103053. [PMID: 38340634 PMCID: PMC10869263 DOI: 10.1016/j.redox.2024.103053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 01/12/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Although reactive oxygen species (ROS) are known to have harmful effects in organisms, recent studies have demonstrated expression of ROS synthases at various parts of the organisms and the controlled ROS generation, suggesting possible involvement of ROS signaling in physiological events of individuals. However, physiological roles of ROS in the CNS, including functional roles in higher brain functions or neuronal activity-dependent ROS production, remain to be elucidated. Here, we demonstrated involvement of ROS - 8-NO2-cGMP signaling in motor learning and synaptic plasticity in the cerebellum. In the presence of inhibitors of ROS signal or ROS synthases, cerebellar motor learning was impaired, and the stimulus inducing long-term depression (LTD), cellular basis for the motor learning, failed to induce LTD but induced long-term potentiation (LTP)-like change at cerebellar synapses. Furthermore, ROS was produced by LTD-inducing stimulus in enzyme-dependent manner, and excess administration of the antioxidant vitamin E impaired cerebellar motor learning, suggesting beneficial roles of endogenous ROS in the learning. As a downstream signal, involvement of 8-NO2-cGMP in motor learning and cerebellar LTD were also revealed. These findings indicate that ROS - 8-NO2-cGMP signal is activated by neuronal activity and is essential for cerebellum-dependent motor learning and synaptic plasticity, demonstrating involvement of the signal in physiological function of brain systems.
Collapse
Affiliation(s)
- Sho Kakizawa
- Department of Biological Chemistry, Graduate School and Faculty of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan.
| | - Tomoko Arasaki
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Ayano Yoshida
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Ayami Sato
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Yuka Takino
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Shuichi Yanai
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Shogo Endo
- Aging Neuroscience Research Team, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan.
| |
Collapse
|
23
|
Anjum R, Clarke VRJ, Nagasawa Y, Murakoshi H, Paradis S. Rem2 interacts with CaMKII at synapses and restricts long-term potentiation in hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584540. [PMID: 38558974 PMCID: PMC10979978 DOI: 10.1101/2024.03.11.584540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Synaptic plasticity, the process whereby neuronal connections are either strengthened or weakened in response to stereotyped forms of stimulation, is widely believed to represent the molecular mechanism that underlies learning and memory. The holoenzyme CaMKII plays a well-established and critical role in the induction of a variety of forms of synaptic plasticity such as long-term potentiation (LTP), long-term depression (LTD) and depotentiation. Previously, we identified the GTPase Rem2 as a potent, endogenous inhibitor of CaMKII. Here, we report that knock out of Rem2 enhances LTP at the Schaffer collateral to CA1 synapse in hippocampus, consistent with an inhibitory action of Rem2 on CaMKII in vivo. Further, re-expression of WT Rem2 rescues the enhanced LTP observed in slices obtained from Rem2 conditional knock out (cKO) mice, while expression of a mutant Rem2 construct that is unable to inhibit CaMKII in vitro fails to rescue increased LTP. In addition, we demonstrate that CaMKII and Rem2 interact in dendritic spines using a 2pFLIM-FRET approach. Taken together, our data lead us to propose that Rem2 serves as a brake on runaway synaptic potentiation via inhibition of CaMKII activity. Further, the enhanced LTP phenotype we observe in Rem2 cKO slices reveals a previously unknown role for Rem2 in the negative regulation of CaMKII function.
Collapse
Affiliation(s)
- Rabia Anjum
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, United States of America
| | - Vernon R J Clarke
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Yutaro Nagasawa
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa 240-0193, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi 444-8585, Japan
| | - Hideji Murakoshi
- Department of Physiological Sciences, The Graduate University for Advanced Studies; Hayama, Kanagawa 240-0193, Japan
- Supportive Center for Brain Research, National Institute for Physiological Sciences; Okazaki, Aichi 444-8585, Japan
| | - Suzanne Paradis
- Department of Biology and Volen Center for Complex Systems, Brandeis University, Waltham, Massachusetts 02454, United States of America
| |
Collapse
|
24
|
Li G, McLaughlin DW, Peskin CS. A biochemical description of postsynaptic plasticity-with timescales ranging from milliseconds to seconds. Proc Natl Acad Sci U S A 2024; 121:e2311709121. [PMID: 38324573 PMCID: PMC10873618 DOI: 10.1073/pnas.2311709121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/29/2023] [Indexed: 02/09/2024] Open
Abstract
Synaptic plasticity [long-term potentiation/depression (LTP/D)], is a cellular mechanism underlying learning. Two distinct types of early LTP/D (E-LTP/D), acting on very different time scales, have been observed experimentally-spike timing dependent plasticity (STDP), on time scales of tens of ms; and behavioral time scale synaptic plasticity (BTSP), on time scales of seconds. BTSP is a candidate for a mechanism underlying rapid learning of spatial location by place cells. Here, a computational model of the induction of E-LTP/D at a spine head of a synapse of a hippocampal pyramidal neuron is developed. The single-compartment model represents two interacting biochemical pathways for the activation (phosphorylation) of the kinase (CaMKII) with a phosphatase, with ion inflow through channels (NMDAR, CaV1,Na). The biochemical reactions are represented by a deterministic system of differential equations, with a detailed description of the activation of CaMKII that includes the opening of the compact state of CaMKII. This single model captures realistic responses (temporal profiles with the differing timescales) of STDP and BTSP and their asymmetries. The simulations distinguish several mechanisms underlying STDP vs. BTSP, including i) the flow of [Formula: see text] through NMDAR vs. CaV1 channels, and ii) the origin of several time scales in the activation of CaMKII. The model also realizes a priming mechanism for E-LTP that is induced by [Formula: see text] flow through CaV1.3 channels. Once in the spine head, this small additional [Formula: see text] opens the compact state of CaMKII, placing CaMKII ready for subsequent induction of LTP.
Collapse
Affiliation(s)
- Guanchun Li
- Courant Institute and Center for Neural Science, Department of Mathematics, New York University, New York, NY10012
| | - David W. McLaughlin
- Courant Institute and Center for Neural Science, Department of Mathematics, New York University, New York, NY10012
- Center for Neural Science, Department of Neural Science, New York University, New York, NY10012
- Institute of Mathematical Science, Mathematics Department, New York University-Shanghai, Shanghai200122, China
- Neuroscience Institute of New York University Langone Health, New York University, New York, NY10016
| | - Charles S. Peskin
- Courant Institute and Center for Neural Science, Department of Mathematics, New York University, New York, NY10012
- Center for Neural Science, Department of Neural Science, New York University, New York, NY10012
| |
Collapse
|
25
|
Wang J, Feng Y, Qi Z, Li J, Chen Z, Zhang J, Zhu D. The role and mechanism of esketamine in preventing and treating remifentanil-induced hyperalgesia based on the NMDA receptor-CaMKII pathway. Open Life Sci 2024; 19:20220816. [PMID: 38314140 PMCID: PMC10836417 DOI: 10.1515/biol-2022-0816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 02/06/2024] Open
Abstract
Remifentanil-induced hyperalgesia (RIH) is a common clinical phenomenon that limits the use of opioids in pain management. Esketamine, a non-competitive N-methyl-d-aspartate (NMDA) receptor antagonist, has been shown to prevent and treat RIH. However, the underlying effect mechanism of esketamine on RIH remains unclear. This study aimed to investigate the role and mechanism of esketamine in preventing and treating RIH based on the NMDA receptor-CaMKIIα pathway. In this study, an experimental animal model was used to determine the therapeutic effect of esketamine on pain elimination. Moreover, the mRNA transcription and protein expression levels of CaMKII and GluN2B were investigated to offer evidence of the protective capability of esketamine in ameliorating RIH. The results demonstrated that esketamine attenuated RIH by inhibiting CaMKII phosphorylation and downstream signaling pathways mediated by the NMDA receptor. Furthermore, ketamine reversed the upregulation of spinal CaMKII induced by remifentanil. These findings suggest that the NMDA receptor-CaMKII pathway plays a critical role in the development of RIH, and ketamine's effect on this pathway may provide a new therapeutic approach for the prevention and treatment of RIH.
Collapse
Affiliation(s)
- Jiafang Wang
- Department of Anesthesiology, Wuhan No. 1 Hospital, No. 215 Zhongshan Avenue, Qiaokou District, Wuhan, Hubei, 430022, China
| | - Yankun Feng
- Department of Anesthesiology, Wuhan No. 1 Hospital, No. 215 Zhongshan Avenue, Qiaokou District, Wuhan, Hubei, 430022, China
| | - Zhong Qi
- Department of Anesthesiology, Wuhan No. 1 Hospital, No. 215 Zhongshan Avenue, Qiaokou District, Wuhan, Hubei, 430022, China
| | - Jin Li
- Department of Anesthesiology, Wuhan No. 1 Hospital, No. 215 Zhongshan Avenue, Qiaokou District, Wuhan, Hubei, 430022, China
| | - Zhijun Chen
- Department of Anesthesiology, Wuhan No. 1 Hospital, No. 215 Zhongshan Avenue, Qiaokou District, Wuhan, Hubei, 430022, China
| | - Jinming Zhang
- Hubei Provincial Key Laboratory for Applied Toxicology, Hubei Center for Disease Control and Prevention, No. 35 Zhuodaoquan North Road, Hongshan District, Wuhan 430079, China
| | - Degang Zhu
- Department of Anesthesiology, Wuhan No. 1 Hospital, No. 215 Zhongshan Avenue, Qiaokou District, Wuhan, Hubei, 430022, China
| |
Collapse
|
26
|
Sobue K. Calmodulin: a highly conserved and ubiquitous Ca 2+ sensor. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2024; 100:368-386. [PMID: 39085063 DOI: 10.2183/pjab.100.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Calcium ions (Ca2+) play critical roles in various biological phenomena. The free Ca2+ concentration in the cytoplasm of a resting cell is at the 10-7 M level, whereas that outside the cell is 10-3 M, creating a 10,000-fold gradient of Ca2+ concentrations across the cell membrane, separating the intracellular and extracellular solutions.1),2) When a cell is activated by external stimuli, the intracellular Ca2+ concentration increases to levels of 10-6-10-5 M through Ca2+ entry from the extracellular solution via plasma membrane Ca2+ channels and/or Ca2+ release from intracellular stores. This transient increase in Ca2+ functions as an important signal mediated by Ca2+ sensors. Thus, Ca2+ signals are transmitted to intracellular loci such as distinct, localized targets of Ca2+ sensors. Among numerous Ca2+ sensors present in cells, calmodulin is a highly conserved and ubiquitous Ca2+ sensor.3).
Collapse
Affiliation(s)
- Kenji Sobue
- Iwate Medical University, Yahaba, Shiwa-gun, Iwate, Japan
| |
Collapse
|
27
|
Zheng Z, Qiu Z, Xiong X, Nie A, Zhou W, Qiu H, Zhao H, Wu H, Guo J. Co-activation of NMDAR and mGluRs controls protein nanoparticle-induced osmotic pressure in neurotoxic edema. Biomed Pharmacother 2023; 169:115917. [PMID: 38006617 DOI: 10.1016/j.biopha.2023.115917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Glutamate stimuli and hyperactivation of its receptor are predominant determinants of ischemia-induced cytotoxic cerebral edema, which is closely associated with protein nanoparticle (PN)-induced increases in osmotic pressure. Herein, we investigated the electrochemical and mechanical mechanisms underlying the neuron swelling induced by PNs via the co-activation of N-methyl-D-aspartate receptor subunit (NMDAR) and excitatory metabotropic glutamate receptors (mGluRs). RESULTS We observed that co-activation of ionic glutamate receptor NMDAR and Group I metabotropic mGluRs promoted alteration of PN-induced membrane potential and increased intracellular osmosis, which was closely associated with calcium and voltage-dependent ion channels. In addition, activation of NMDAR-induced calmodulin (CaM) and mGluR downstream diacylglycerol (DAG)/protein kinase C α (PKCα) were observed to play crucial roles in cytotoxic hyperosmosis. The crosstalk between CaM and PKCα could upregulate the sensitivity and sustained opening of sulfonylurea receptor 1 (SUR1)-transient receptor potential cation channel subfamily M member 4 (TRPM4) and transmembrane protein 16 A (TMEM16A) channels, respectively, maintaining the massive Na+/Cl- influx, and the resultant neuron hyperosmosis and swelling. Intracellular PNs and Na+/Cl- influx were found to be as potential targets for cerebral edema treatment, using the neurocyte osmosis system and a cerebral ischemic rat model. CONCLUSIONS This study highlights PNs as a key factor in "electrochemistry-tension" signal transduction controlling Na+/Cl- ion channels and increased osmotic pressure in ischemia-induced cytotoxic edema. Moreover, enhanced sensitivity in both Na+ and Cl- ion channels also has a crucial role in cerebral edema.
Collapse
Affiliation(s)
- Zihui Zheng
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Zhaoshun Qiu
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Xiyu Xiong
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Aobo Nie
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Wenzhao Zhou
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Huimin Qiu
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Huanhuan Zhao
- Basic Medical Experiment Center, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Huiwen Wu
- Laboratory Center for Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China.
| | - Jun Guo
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China.
| |
Collapse
|
28
|
Ma H, Khaled HG, Wang X, Mandelberg NJ, Cohen SM, He X, Tsien RW. Excitation-transcription coupling, neuronal gene expression and synaptic plasticity. Nat Rev Neurosci 2023; 24:672-692. [PMID: 37773070 DOI: 10.1038/s41583-023-00742-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/30/2023]
Abstract
Excitation-transcription coupling (E-TC) links synaptic and cellular activity to nuclear gene transcription. It is generally accepted that E-TC makes a crucial contribution to learning and memory through its role in underpinning long-lasting synaptic enhancement in late-phase long-term potentiation and has more recently been linked to late-phase long-term depression: both processes require de novo gene transcription, mRNA translation and protein synthesis. E-TC begins with the activation of glutamate-gated N-methyl-D-aspartate-type receptors and voltage-gated L-type Ca2+ channels at the membrane and culminates in the activation of transcription factors in the nucleus. These receptors and ion channels mediate E-TC through mechanisms that include long-range signalling from the synapse to the nucleus and local interactions within dendritic spines, among other possibilities. Growing experimental evidence links these E-TC mechanisms to late-phase long-term potentiation and learning and memory. These advances in our understanding of the molecular mechanisms of E-TC mean that future efforts can focus on understanding its mesoscale functions and how it regulates neuronal network activity and behaviour in physiological and pathological conditions.
Collapse
Affiliation(s)
- Huan Ma
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China.
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China.
- Research Units for Emotion and Emotional Disorders, Chinese Academy of Medical Sciences, Beijing, China.
| | - Houda G Khaled
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
- Center for Neural Science, New York University, New York, NY, USA
| | - Xiaohan Wang
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Nataniel J Mandelberg
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Samuel M Cohen
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA
| | - Xingzhi He
- Department of Neurobiology, Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-Machine Integration, State Key Laboratory of Brain-Machine Intelligence, Zhejiang University, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou, China
- Research Units for Emotion and Emotional Disorders, Chinese Academy of Medical Sciences, Beijing, China
| | - Richard W Tsien
- NYU Neuroscience Institute and Department of Neuroscience and Physiology, NYU Langone Medical Center, New York, NY, USA.
- Center for Neural Science, New York University, New York, NY, USA.
| |
Collapse
|
29
|
Zhou L, Chen W, Jiang S, Xu R. In Vitro Models of Amyotrophic Lateral Sclerosis. Cell Mol Neurobiol 2023; 43:3783-3799. [PMID: 37870685 DOI: 10.1007/s10571-023-01423-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/04/2023] [Indexed: 10/24/2023]
Abstract
Amyotrophic Lateral Sclerosis (ALS) is one of the commonest neurodegenerative diseases of adult-onset, which is characterized by the progressive death of motor neurons in the cerebral cortex, brain stem and spinal cord. The dysfunction and death of motor neurons lead to the progressive muscle weakness, atrophy, fasciculations, spasticity and ultimately the whole paralysis of body. Despite the identification of several genetic mutations associated with the pathogenesis of ALS, including mutations in chromosome 9 open reading frame 72 leading to the abnormal expansion of GGGGCC repeat sequence, TAR DNA-binding protein 43, fused in sarcoma/translocated in liposarcoma, copper/zinc superoxide dismutase 1 (SOD1) and TANK-binding kinase 1, the exact mechanisms underlying the specific degeneration of motor neurons that causes ALS remain incompletely understood. At present, since the transgenic model expressed SOD1 mutants was established, multiple in vitro models of ALS have been developed for studying the pathology, pathophysiology and pathogenesis of ALS as well as searching the effective neurotherapeutics. This review reviewed the details of present established in vitro models used in studying the pathology, pathophysiology and pathogenesis of ALS. Meanwhile, we also discussed the advantages, disadvantages, cost and availability of each models.
Collapse
Affiliation(s)
- Lijun Zhou
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, National Regional Medical Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, 330008, Jiangxi, China
- Medical College of Nanchang University, Nanchang, 330006, China
| | - Wenzhi Chen
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, National Regional Medical Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, 330008, Jiangxi, China
| | - Shishi Jiang
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, National Regional Medical Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, 330008, Jiangxi, China
- Medical College of Nanchang University, Nanchang, 330006, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People's Hospital, Clinical College of Nanchang Medical College, First Affiliated Hospital of Nanchang Medical College, Xiangya Hospital of Central South University Jiangxi Hospital, National Regional Medical Center for Neurological Diseases, No. 266 Fenghe North Avenue, Honggutan District, Nanchang, 330008, Jiangxi, China.
- Medical College of Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
30
|
Chen X, Cai Q, Zhou J, Pleasure SJ, Schulman H, Zhang M, Nicoll RA. CaMKII autophosphorylation but not downstream kinase activity is required for synaptic memory. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.25.554912. [PMID: 37662326 PMCID: PMC10473743 DOI: 10.1101/2023.08.25.554912] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
CaMKII plays a critical role in long-term potentiation (LTP), a well-established model for learning and memory through the enhancement of synaptic transmission. Biochemical studies indicate that CaMKII catalyzes a phosphotransferase (kinase) reaction of both itself (autophosphorylation) and of multiple downstream target proteins. However, whether either type of phosphorylation plays any role in the synaptic enhancing action of CaMKII remains hotly contested. We have designed a series of experiments to define the minimal requirements for the synaptic enhancement by CaMKII. We find that autophosphorylation of T286 and further binding of CaMKII to the GluN2B subunit are required both for initiating LTP and for its maintenance (synaptic memory). Once bound to the NMDA receptor, the synaptic action of CaMKII occurs in the absence of kinase activity. Thus, autophosphorylation, together with binding to the GluN2B subunit, are the only two requirements for CaMKII in synaptic memory.
Collapse
|