1
|
Chagraoui A, Thibaut F, De Deurwaerdère P. 5-HT6 receptors: Contemporary views on their neurobiological and pharmacological relevance in neuropsychiatric disorders. DIALOGUES IN CLINICAL NEUROSCIENCE 2025; 27:112-128. [PMID: 40347153 PMCID: PMC12068339 DOI: 10.1080/19585969.2025.2502028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 04/02/2025] [Accepted: 04/30/2025] [Indexed: 05/12/2025]
Abstract
Despite the relatively limited number of serotonergic neurons in humans, serotonin plays a key role in neurophysiological functions, including sleep, pain perception, learning, memory, cognition, emotion, reward, and mood regulation. Altered serotonergic neurotransmission is linked to conditions such as anxiety, depression, anorexia, migraine, insomnia, schizophrenia, Alzheimer's disease (AD), and cognitive impairments. The 5-HT6 receptor (5-HT6R), mainly found in brain regions involved in cognition, is a promising therapeutic target for cognitive deficits in neuropsychiatric disorders, particularly AD and schizophrenia. Preclinical studies have shown that 5-HT6R antagonists improve cognitive function. 5-HT6R interacts dynamically with an extensive intracellular protein network, regulating the localisation, trafficking, and signalling of these proteins. Proteomic and genetic studies have revealed interactions with mTOR kinase and neurofibromin, both of which are crucial for synaptic plasticity, learning, and memory. Fyn kinase is also associated with 5-HT6Rs, reinforcing receptor expression and G-protein coupling. Notably, the G protein-regulated inducer of neurite outgrowth 1 (GPRIN1) interacts with 5-HT6Rs independently of agonists, enhancing receptor activity. This review highlights the clinical testing of 5-HT6R ligands as regulators of these complex signalling properties, underscoring their therapeutic potential in addressing cognitive impairments associated with neuropsychiatric disorders.
Collapse
Affiliation(s)
- Abdeslam Chagraoui
- Department of Medical Biochemistry, Rouen University Hospital, CHU de Rouen, France
- University of Rouen, Faculty of Medicine and Pharmacy, Inserm U1239, Neuroendocrine, Endocrine, and Germinal Differentiation and Communication (NorDiC), Mont-Saint-Aignan, France
| | - Florence Thibaut
- Department of Psychiatry and Addictive Disorders, University Hospital Cochin (site Tarnier) AP-HP, Paris, France
- INSERM U1266, Institute of Psychiatry and Neurosciences, University of Paris, Cité, Paris, France
| | - Philippe De Deurwaerdère
- Centre National de la Recherche Scientifique, Institut des Neurosciences Intégratives et Cognitives d’Aquitaine, UMR 5287, Bordeaux, France
| |
Collapse
|
2
|
Gerlai R. The importance of understanding the ethology and ecology of the zebrafish, and of other fish species, in experimental research. Biol Futur 2025:10.1007/s42977-025-00257-3. [PMID: 40316876 DOI: 10.1007/s42977-025-00257-3] [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: 01/04/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025]
Abstract
This short review appears in a special issue assembled to celebrate the 90th birthday of a Hungarian ethologist, Professor Vilmos Csányi. As such, it includes some autobiographical details specific to that scientist and the author of this review. However, these details also serve an important general message. They exemplify how science, i.e., specifically the use of fish in the analysis of behaviour and brain function progressed from the mid-1970s to the current day. They illuminate how scientists choose their study species, and how this choice influences the research questions one may be able to pose. The review discusses why the zebrafish has become a popular research subject of biology, including behavioural neuroscience. It argues that behavioural analysis should be an integral part of research into the analysis of brain function. It considers the dichotomy between the historical effect of North American behaviourism vs. the legacy of European Nobel laureate ethologists. It demonstrates, through a theoretical example, why merging these two "schools" of thoughts is the appropriate way to conduct behavioural research. It provides a few examples for how combining knowledge of ethology and ecology of the species with systematic laboratory studies may be beneficial. And it presents a brief outlook for the future of fish in biology research.
Collapse
Affiliation(s)
- Robert Gerlai
- Department of Psychology, Rm CCT4004, University of Toronto Mississauga, 3359 Mississauga Road, Mississauga, ON, L5L 1C6, Canada.
| |
Collapse
|
3
|
Zelek-Molik A, Gądek-Michalska A, Wilczkowski M, Bielawski A, Maziarz K, Kreiner G, Nalepa I. Restraint stress effects on glutamate signaling protein levels in the rats' frontal cortex: Does β1 adrenoceptor activity matter? Front Pharmacol 2025; 15:1451895. [PMID: 39834820 PMCID: PMC11743458 DOI: 10.3389/fphar.2024.1451895] [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: 06/20/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction Stress-evoked dysfunctions of the frontal cortex (FC) are correlated with changes in the functioning of the glutamatergic system, and evidence demonstrates that noradrenergic transmission is an important regulator of this process. In the current study, we adopted a restraint stress (RS) model in male Wistar rats to investigate whether the blockade of β1 adrenergic receptors (β1AR) with betaxolol (BET) in stressed animals influences the body's stress response and the expression of selected signaling proteins in the medial prefrontal cortex (mPFC). Methods The study was divided into two parts. In the first part, rats were exposed to RS for 3, 7, or 14 days, and the expression of glutamate signaling proteins (p(S845)/t GluA1, p(Y1472)/t GluN2B, VGLUT1, and VGLUT2) in the FC was analyzed to determine the optimal RS duration for studying the mechanisms of hypofrontality. In the second part, rats were exposed to RS for 14 days, and BET (5 mg/kg, p. o.) was administered during the last 8 days immediately after RS. The body's stress reaction was assessed by analyzing body weight and blood levels of adrenocorticotropic hormone (ACTH) and corticosterone (CORT). Behavioral responses were evaluated using the novel object recognition (NOR) and elevated plus maze (EPM) tests. The impact of RS and BET on the expression of p(Y530)/t Fyn and p (S133)/t CREB in the mPFC was measured via Western blotting. Results and Discussion The first part of the study demonstrated a decreased level of glutamate receptors in rats exposed to 14 days of RS, following an initial increase observed after 7 days of RS. Results from the second part revealed that chronic RS reduced body weight, impaired recognition memory in the NOR test, augmented blood levels of ACTH, and increased the expression of p(Y530) Fyn in the mPFC. However, β1AR blockade did not alter the effects of RS on weight gain, cognitive function, or the expression of p(Y530) Fyn. β1AR blockade normalized only the blood concentration of ACTH. These results suggest that decreased Fyn kinase activity, indicated by phosphorylation at Y530, underlies the stress-evoked downregulation of GluN2B in the FC in a manner independent of β1AR activity.
Collapse
Affiliation(s)
- Agnieszka Zelek-Molik
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Anna Gądek-Michalska
- Department of Physiology, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Michał Wilczkowski
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Adam Bielawski
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Katarzyna Maziarz
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Grzegorz Kreiner
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| | - Irena Nalepa
- Department of Brain Biochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Krakow, Poland
| |
Collapse
|
4
|
Xu C, Tang Y, Lu X, Chen R. Fyn, an important molecule in the brain, is a potential therapeutic target for brain tumours. Front Pharmacol 2024; 15:1485919. [PMID: 39697541 PMCID: PMC11652172 DOI: 10.3389/fphar.2024.1485919] [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: 08/25/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
Under normal physiological conditions, Fyn, a nonreceptor tyrosine kinase, is involved in signal transduction pathways in the nervous system and in the formation and activation of T lymphocytes. Fyn is a member of the Src family of kinases (SFKs) and plays a role in cell morphogenic transformation, motility, proliferation, and death, which in turn influences the development and progression of various cancer types. SFKs are overexpressed or hyperactive in tumours, and they are engaged in several signalling pathways that lead to tumour development. Inhibition of Fyn can enhance patient outcomes and prolong survival. Thus, Fyn is a desirable therapeutic target in a variety of tumour types. To lay the groundwork for further investigation and targeted therapy in tumours, in this article, we review the most recent findings on the function of Fyn in tumours, with an emphasis on its role in gliomas. Understanding the function of Fyn during tumourigenesis and development and in resistance to anticancer therapeutic agents can aid in the development and application of innovative medicines that specifically target this kinase, thus improving the management of cancers.
Collapse
Affiliation(s)
- Chongxi Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Tang
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xing Lu
- Department of Gynecological Nursing, West China Second Hospital, Sichuan University, Chengdu, China
| | - Ruiqi Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Zhou J, Lu X, Wang H. The underlying molecular mechanisms of Fyn in neonatal hypoxic-ischaemic encephalopathy. Front Cell Neurosci 2024; 18:1476856. [PMID: 39664999 PMCID: PMC11631624 DOI: 10.3389/fncel.2024.1476856] [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: 08/12/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Fyn is a cytoplasmic tyrosine kinase (TK) that is a nonreceptor and a member of the Src family of kinases (SFKs). It is involved in several transduction pathways in the central nervous system (CNS), such as oligodendrocyte development, myelination, axon guidance, and synaptic transmission. Owing to its wide range of activities in the molecular signaling pathways that underpin both neuropathologic and neurodevelopmental events, Fyn has remained of great interest for more than a century. Accumulating preclinical data have highlighted the potential role of Fyn in the pathophysiology of neonatal hypoxic-ischaemic encephalopathy (HIE). By mediating important signaling pathways, Fyn may control glutamate excitotoxicity, promote neuroinflammation and facilitate the death of neurons caused by oxidative stress. In this review, we address new evidence regarding the role of Fyn in the pathogenesis of this condition, with the aim of providing a reference for the development of new strategies to improve the prognosis of neonatal HIE. In addition, we also offer insights into additional Fyn-related molecular mechanisms involved in HIE pathology.
Collapse
Affiliation(s)
- Jiao Zhou
- Department of Reproductive Medicine Nursing, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Xiang Lu
- Department of Cardiology, The First People’s Hospital of Yuexi County, Yuexi, China
| | - Haichuan Wang
- Department of Paediatrics, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
6
|
Putra M, Rao NS, Gardner C, Liu G, Trommater J, Bunney M, Gage M, Bassuk AG, Hefti M, Lee G, Thippeswamy T. Enhanced Fyn-tau and NR2B-PSD95 interactions in epileptic foci in experimental models and human epilepsy. Brain Commun 2024; 6:fcae327. [PMID: 39355003 PMCID: PMC11444080 DOI: 10.1093/braincomms/fcae327] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 10/03/2024] Open
Abstract
Epilepsy and Alzheimer's disease share some common pathologies such as neurodegeneration, seizures and impaired cognition. However, the molecular mechanisms of these changes are still largely unknown. Fyn, a Src-family non-receptor tyrosine kinase (SFK), and its interaction with tau in mediating brain pathology in epilepsy and Alzheimer's disease can be a potential therapeutic target for disease modification. Although Fyn and tau pathology occurs in both Alzheimer's disease and epilepsy, the dynamics of Fyn-tau and PSD95-NR2B interactions affected by seizures and their impact on brain pathology in epilepsy have not been investigated. In this study, we demonstrate a significant increase of Fyn-tau interactions following seizure induction by kainate in both acute and chronic rodent models and in human epilepsy. In the early phase of epileptogenesis, we show increased Fyn/tau/NR2B/PSD95/neuronal nitric oxide synthase complexes after status epilepticus and a postsynaptic increase of phosphorylated tau (pY18 and AT8), Fyn (pSFK-Y416), NMDAR (pNR2B-Y1472) and neuronal nitric oxide synthase. Hippocampal proximity ligation assay and co-immunoprecipitation revealed a sustained increase of Fyn-tau and NR2B-PSD95 complexes/binding in rat chronic epilepsy at 3 months post-status epilepticus. Enhanced Fyn-tau complexes strongly correlated with the frequency of spontaneously recurring convulsive seizures and epileptiform spikes in the chronic epilepsy model. In human epileptic brains, we also identified increased Fyn-tau and NR2B-PSD95 complexes, tau phosphorylation (pY18 and AT8) and Fyn activation (pSFK-Y416), implying the translational and therapeutic potential of these molecular interactions. In tau knockout mice and in rats treated with a Fyn/SFK inhibitor saracatinib, we found a significant reduction of phosphorylated Fyn, tau (AT8 in saracatinib-treated), NR2B and neuronal nitric oxide synthase and their interactions (Fyn-tau and NR2B-PSD95 in saracatinib-treated group; NR2B-PSD95 in tau knockout group). The reduction of Fyn-tau and NR2B-PSD95 interactions in the saracatinib-treated group, in contrast to the vehicle-treated group, correlated with the modification in seizure progression in the rat chronic epilepsy model. These findings from animal models and human epilepsy provide evidence for the role of Fyn-tau and NR2B-PSD95 interactions in seizure-induced brain pathology and suggest that blocking such interactions could modify the progression of epilepsy.
Collapse
Affiliation(s)
- Marson Putra
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Nikhil S Rao
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Cara Gardner
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Guanghao Liu
- Department of Internal Medicine, Carver College of Medicine, Carver College of Medicine University of Iowa, Iowa City, IA 52242, USA
| | - Jordan Trommater
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Michael Bunney
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Meghan Gage
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| | - Alexander G Bassuk
- Department of Pediatrics, The University of Iowa Stead Family, Iowa City, IA 52242, USA
- Department of Neurology, The University of Iowa Stead Family, Iowa City, IA 52242, USA
- Iowa Neuroscience Institute (INI), College of Medicine, University of Iowa Carver, Iowa City, IA 52242, USA
| | - Marco Hefti
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52240, USA
| | - Gloria Lee
- Department of Internal Medicine, Carver College of Medicine, Carver College of Medicine University of Iowa, Iowa City, IA 52242, USA
| | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50010, USA
| |
Collapse
|
7
|
Bartošík V, Plucarová J, Laníková A, Janáčková Z, Padrta P, Jansen S, Vařečka V, Gruber T, Feller SM, Žídek L. Structural basis of binding the unique N-terminal domain of microtubule-associated protein 2c to proteins regulating kinases of signaling pathways. J Biol Chem 2024; 300:107551. [PMID: 39002671 PMCID: PMC11367651 DOI: 10.1016/j.jbc.2024.107551] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/15/2024] Open
Abstract
Isoforms of microtubule-associated protein 2 (MAP2) differ from their homolog Tau in the sequence and interactions of the N-terminal region. Binding of the N-terminal region of MAP2c (N-MAP2c) to the dimerization/docking domains of the regulatory subunit RIIα of cAMP-dependent protein kinase (RIIDD2) and to the Src-homology domain 2 (SH2) of growth factor receptor-bound protein 2 (Grb2) have been described long time ago. However, the structural features of the complexes remained unknown due to the disordered nature of MAP2. Here, we provide structural description of the complexes. We have solved solution structure of N-MAP2c in complex with RIIDD2, confirming formation of an amphiphilic α-helix of MAP2c upon binding, defining orientation of the α-helix in the complex and showing that its binding register differs from previous predictions. Using chemical shift mapping, we characterized the binding interface of SH2-Grb2 and rat MAP2c phosphorylated by the tyrosine kinase Fyn in their complex and proposed a model explaining differences between SH2-Grb2 complexes with rat MAP2c and phosphopeptides with a Grb2-specific sequence. The results provide the structural basis of a potential role of MAP2 in regulating cAMP-dependent phosphorylation cascade via interactions with RIIDD2 and Ras signaling pathway via interactions with SH2-Grb2.
Collapse
Affiliation(s)
- Viktor Bartošík
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Jitka Plucarová
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic; Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Alice Laníková
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic; Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Zuzana Janáčková
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petr Padrta
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic; Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Séverine Jansen
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic; Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Vojtěch Vařečka
- Institute of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Tobias Gruber
- Institute of Molecular Medicine, Tumor Biology, Martin-Luther-University of Halle-Wittenberg, Germany; Institute of Physics, Biophysics, Martin-Luther-University of Halle-Wittenberg, Germany
| | - Stephan M Feller
- Institute of Molecular Medicine, Tumor Biology, Martin-Luther-University of Halle-Wittenberg, Germany
| | - Lukáš Žídek
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno, Czech Republic; Central European Institute of Technology, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
8
|
Yu K, Yao KR, Aguinaga MA, Choquette JM, Liu C, Wang Y, Liao D. G272V and P301L Mutations Induce Isoform Specific Tau Mislocalization to Dendritic Spines and Synaptic Dysfunctions in Cellular Models of 3R and 4R Tau Frontotemporal Dementia. J Neurosci 2024; 44:e1215232024. [PMID: 38858079 PMCID: PMC11236579 DOI: 10.1523/jneurosci.1215-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 06/12/2024] Open
Abstract
Tau pathologies are detected in the brains of some of the most common neurodegenerative diseases including Alzheimer's disease (AD), Lewy body dementia (LBD), chronic traumatic encephalopathy (CTE), and frontotemporal dementia (FTD). Tau proteins are expressed in six isoforms with either three or four microtubule-binding repeats (3R tau or 4R tau) due to alternative RNA splicing. AD, LBD, and CTE brains contain pathological deposits of both 3R and 4R tau. FTD patients can exhibit either 4R tau pathologies in most cases or 3R tau pathologies less commonly in Pick's disease, which is a subfamily of FTD. Here, we report the isoform-specific roles of tau in FTD. The P301L mutation, linked to familial 4R tau FTD, induces mislocalization of 4R tau to dendritic spines in primary hippocampal cultures that were prepared from neonatal rat pups of both sexes. Contrastingly, the G272V mutation, linked to familial Pick's disease, induces phosphorylation-dependent mislocalization of 3R tau but not 4R tau proteins to dendritic spines. The overexpression of G272V 3R tau but not 4R tau proteins leads to the reduction of dendritic spine density and suppression of mEPSCs in 5-week-old primary rat hippocampal cultures. The decrease in mEPSC amplitude caused by G272V 3R tau is dynamin-dependent whereas that caused by P301L 4R tau is dynamin-independent, indicating that the two tau isoforms activate different signaling pathways responsible for excitatory synaptic dysfunction. Our 3R and 4R tau studies here will shed new light on diverse mechanisms underlying FTD, AD, LBD, and CTE.
Collapse
Affiliation(s)
- Ke Yu
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- Department of General Practice, The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Katherine R Yao
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- College of Biological Sciences, University of Minnesota, St Paul, Minnesota 55108
| | - Miguel A Aguinaga
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
- College of Biological Sciences, University of Minnesota, St Paul, Minnesota 55108
| | - Jessica M Choquette
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Chengliang Liu
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Yuxin Wang
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| | - Dezhi Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455
| |
Collapse
|
9
|
Stamenkovic V, Lautz JD, Harsh FM, Smith SEP. SRC family kinase inhibition rescues molecular and behavioral phenotypes, but not protein interaction network dynamics, in a mouse model of Fragile X syndrome. Mol Psychiatry 2024; 29:1392-1405. [PMID: 38297084 PMCID: PMC11524049 DOI: 10.1038/s41380-024-02418-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/02/2024]
Abstract
Glutamatergic synapses encode information from extracellular inputs using dynamic protein interaction networks (PINs) that undergo widespread reorganization following synaptic activity, allowing cells to distinguish between signaling inputs and generate coordinated cellular responses. Here, we investigate how Fragile X Messenger Ribonucleoprotein (FMRP) deficiency disrupts signal transduction through a glutamatergic synapse PIN downstream of NMDA receptor or metabotropic glutamate receptor (mGluR) stimulation. In cultured cortical neurons or acute cortical slices from P7, P17 and P60 FMR1-/y mice, the unstimulated protein interaction network state resembled that of wildtype littermates stimulated with mGluR agonists, demonstrating resting state pre-activation of mGluR signaling networks. In contrast, interactions downstream of NMDAR stimulation were similar to WT. We identified the Src family kinase (SFK) Fyn as a network hub, because many interactions involving Fyn were pre-activated in FMR1-/y animals. We tested whether targeting SFKs in FMR1-/y mice could modify disease phenotypes, and found that Saracatinib (SCB), an SFK inhibitor, normalized elevated basal protein synthesis, novel object recognition memory and social behavior in FMR1-/y mice. However, SCB treatment did not normalize the PIN to a wild-type-like state in vitro or in vivo, but rather induced extensive changes to protein complexes containing Shank3, NMDARs and Fyn. We conclude that targeting abnormal nodes of a PIN can identify potential disease-modifying drugs, but behavioral rescue does not correlate with PIN normalization.
Collapse
Affiliation(s)
- Vera Stamenkovic
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Jonathan D Lautz
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Felicia M Harsh
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Stephen E P Smith
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
| |
Collapse
|
10
|
Vorhees CV, Williams MT. Tests for learning and memory in rodent regulatory studies. Curr Res Toxicol 2024; 6:100151. [PMID: 38304257 PMCID: PMC10832385 DOI: 10.1016/j.crtox.2024.100151] [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] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 01/11/2024] [Accepted: 01/16/2024] [Indexed: 02/03/2024] Open
Abstract
For decades, regulatory guidelines for safety assessment in rodents for drugs, chemicals, pesticides, and food additives with developmental neurotoxic potential have recommended a single test of learning and memory (L&M). In recent years some agencies have requested two such tests. Given the importance of higher cognitive function to health, and the fact that different types of L&M are mediated by different brain regions assessing higher functions represents a step forward in providing better evidence-based protection against adverse brain effects. Given the myriad of tests available for assessing L&M in rodents this leads to the question of which tests best fit regulatory guidelines. To address this question, we begin by describing the central role of two types of L&M essential to all mammalian species and the regions/networks that mediate them. We suggest that the tests recommended possess characteristics that make them well suited to the needs in regulatory safety studies. By brain region, these are (1) the hippocampus and entorhinal cortex for spatial navigation, which assesses explicit L&M for reference and episodic memory and (2) the striatum and related structures for egocentric navigation, which assesses implicit or procedural memory and path integration. Of the tests available, we suggest that in this context, the evidence supports the use of water mazes, specifically, the Morris water maze (MWM) for spatial L&M and the Cincinnati water maze (CWM) for egocentric/procedural L&M. We review the evidentiary basis for these tests, describe their use, and explain procedures that optimize their sensitivity.
Collapse
Affiliation(s)
- Charles V. Vorhees
- Corresponding author at: Div. of Neurology, Cincinnati Children’s Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229, USA.
| | | |
Collapse
|
11
|
Kvergelidze E, Barbakadze T, Bátor J, Kalandadze I, Mikeladze D. Thyroid hormone T3 induces Fyn modification and modulates palmitoyltransferase gene expression through αvβ3 integrin receptor in PC12 cells during hypoxia. Transl Neurosci 2024; 15:20220347. [PMID: 39118829 PMCID: PMC11306964 DOI: 10.1515/tnsci-2022-0347] [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: 03/08/2024] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Thyroid hormones (THs) are essential in neuronal and glial cell development and differentiation, synaptogenesis, and myelin sheath formation. In addition to nuclear receptors, TH acts through αvβ3-integrin on the plasma membrane, influencing transcriptional regulation of signaling proteins that, in turn, affect adhesion and survival of nerve cells in various neurologic disorders. TH exhibits protective properties during brain hypoxia; however, precise intracellular mechanisms responsible for the preventive effects of TH remain unclear. In this study, we investigated the impact of TH on integrin αvβ3-dependent downstream systems in normoxic and hypoxic conditions of pheochromocytoma PC12 cells. Our findings reveal that triiodothyronine (T3), acting through αvβ3-integrin, induces activation of the JAK2/STAT5 pathway and suppression of the SHP2 in hypoxic PC12 cells. This activation correlates with the downregulation of the expression palmitoyltransferase-ZDHHC2 and ZDHHC9 genes, leading to a subsequent decrease in palmitoylation and phosphorylation of Fyn tyrosine kinase. We propose that these changes may occur due to STAT5-dependent epigenetic silencing of the palmitoyltransferase gene, which in turn reduces palmitoylation/phosphorylation of Fyn with a subsequent increase in the survival of cells. In summary, our study provides the first evidence demonstrating the involvement of integrin-dependent JAK/STAT pathway, SHP2 suppression, and altered post-translational modification of Fyn in protective effects of T3 during hypoxia.
Collapse
Affiliation(s)
- Elisabed Kvergelidze
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, 0162, Georgia
| | - Tamar Barbakadze
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, 0162, Georgia
- Laboratory of Biochemistry, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, 0160, Georgia
| | - Judit Bátor
- Department of Medical Biology and Central Electron Microscopic Laboratory, Medical School, University of Pécs, Pécs, 7624, Hungary
- Janos Szentagothai Research Centre, University of Pécs, Pécs, 7624, Hungary
| | - Irine Kalandadze
- Laboratory of Biochemistry, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, 0160, Georgia
| | - David Mikeladze
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, 0162, Georgia
- Laboratory of Biochemistry, Ivane Beritashvili Center of Experimental Biomedicine, Tbilisi, 0160, Georgia
| |
Collapse
|
12
|
Nachtigall EG, de C Myskiw J, Izquierdo I, Furini CRG. Cellular mechanisms of contextual fear memory reconsolidation: Role of hippocampal SFKs, TrkB receptors and GluN2B-containing NMDA receptors. Psychopharmacology (Berl) 2024; 241:61-73. [PMID: 37700085 DOI: 10.1007/s00213-023-06463-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023]
Abstract
Memories are stored into long-term representations through a process that depends on protein synthesis. However, a consolidated memory is not static and inflexible and can be reactivated under certain circumstances, the retrieval is able to reactivate memories and destabilize them engaging a process of restabilization known as reconsolidation. Although the molecular mechanisms that mediate fear memory reconsolidation are not entirely known, so here we investigated the molecular mechanisms in the hippocampus involved in contextual fear conditioning memory (CFC) reconsolidation in male Wistar rats. We demonstrated that the blockade of Src family kinases (SFKs), GluN2B-containing NMDA receptors and TrkB receptors (TrkBR) in the CA1 region of the hippocampus immediately after the reactivation session impaired contextual fear memory reconsolidation. These impairments were blocked by the neurotrophin BDNF and the NMDAR agonist, D-Serine. Considering that the study of the link between synaptic proteins is crucial for understanding memory processes, targeting the reconsolidation process may provide new ways of disrupting maladaptive memories, such as those seen in post-traumatic stress disorder. Here we provide new insights into the cellular mechanisms involved in contextual fear memory reconsolidation, demonstrating that SFKs, GluN2B-containing NMDAR, and TrkBR are necessary for the reconsolidation process. Our findings suggest a link between BDNF and SFKs and GluN2B-containing NMDAR as well as a link between NMDAR and SFKs and TrkBR in fear memory reconsolidation. These preliminary pharmacological findings provide new evidence of the mechanisms involved in the reconsolidation of fear memory and have the potential to contribute to the development of treatments for psychiatric disorders involving maladaptive memories.
Collapse
Affiliation(s)
- Eduarda G Nachtigall
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, Porto Alegre, RS, 90610-000, Brazil
- Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 2nd floor - HSL, Porto Alegre, RS, 90610-000, Brazil
| | - Jociane de C Myskiw
- Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 2nd floor - HSL, Porto Alegre, RS, 90610-000, Brazil
| | - Ivan Izquierdo
- Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 2nd floor - HSL, Porto Alegre, RS, 90610-000, Brazil
| | - Cristiane R G Furini
- Laboratory of Cognition and Memory Neurobiology, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 3rd floor, Porto Alegre, RS, 90610-000, Brazil.
- Memory Center, Brain Institute, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Av. Ipiranga, 6690 - 2nd floor - HSL, Porto Alegre, RS, 90610-000, Brazil.
| |
Collapse
|
13
|
Eyford BA, Lazarczyk MJ, Choi KB, Varghese M, Arora H, Kari S, Munro L, Pfeifer CG, Sowa A, Dickstein DR, Dickstein DL, Jefferies WA. Outside-in signaling through the major histocompatibility complex class-I cytoplasmic tail modulates glutamate receptor expression in neurons. Sci Rep 2023; 13:13079. [PMID: 37567897 PMCID: PMC10421907 DOI: 10.1038/s41598-023-38663-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/12/2023] [Indexed: 08/13/2023] Open
Abstract
The interplay between AMPA-type glutamate receptors (AMPARs) and major histocompatibility complex class I (MHC-I) proteins in regulating synaptic signaling is a crucial aspect of central nervous system (CNS) function. In this study, we investigate the significance of the cytoplasmic tail of MHC-I in synaptic signaling within the CNS and its impact on the modulation of synaptic glutamate receptor expression. Specifically, we focus on the Y321 to F substitution (Y321F) within the conserved cytoplasmic tyrosine YXXΦ motif, known for its dual role in endocytosis and cellular signaling of MHC-I. Our findings reveal that the Y321F substitution influences the expression of AMPAR subunits GluA2/3 and leads to alterations in the phosphorylation of key kinases, including Fyn, Lyn, p38, ERK1/2, JNK1/2/3, and p70 S6 kinase. These data illuminate the crucial role of MHC-I in AMPAR function and present a novel mechanism by which MHC-I integrates extracellular cues to modulate synaptic plasticity in neurons, which ultimately underpins learning and memory.
Collapse
Affiliation(s)
- Brett A Eyford
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
| | - Maciej J Lazarczyk
- Division of Institutional Measures, Department of Medical Direction and Quality, University Hospitals of Geneva, Geneva, Switzerland
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Kyung Bok Choi
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Merina Varghese
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Hitesh Arora
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Suresh Kari
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lonna Munro
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Cheryl G Pfeifer
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Allison Sowa
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Daniel R Dickstein
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Dara L Dickstein
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA.
- Department of Pathology, Uniformed Services University of Health Sciences, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine (HJF), 6720A Rockledge Drive, Bethesda, MD, 20817, USA.
| | - Wilfred A Jefferies
- Michael Smith Laboratories, The University of British Columbia, 2185 East Mall, Vancouver, BC, V6T 1Z4, Canada.
- The Vancouver Prostate Centre, Jack Bell Research Centre, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Centre for Blood Research, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 2215 Wesbrook Mall, Vancouver, BC, V6T 2B5, Canada.
- Department of Microbiology and Immunology, University of British Columbia, 1365-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- Department of Zoology, University of British Columbia, 2370-6270 University Blvd., Vancouver, BC, V6T 1Z4, Canada.
- Department of Medical Genetics, University of British Columbia, 1364-2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
14
|
Magnuson J, Ozdemir MA, Mathieson E, Kirkman S, Passera B, Rampersad S, Dufour AB, Brooks D, Pascual-Leone A, Fried PJ, Shafi MM, Ozdemir RA. Neuromodulatory effects and reproducibility of the most widely used repetitive transcranial magnetic stimulation protocols. PLoS One 2023; 18:e0286465. [PMID: 37352290 PMCID: PMC10289434 DOI: 10.1371/journal.pone.0286465] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/16/2023] [Indexed: 06/25/2023] Open
Abstract
BACKGROUND Repetitive transcranial magnetic stimulation (rTMS) is widely used in both research and clinical settings to modulate human brain function and behavior through the engagement of the mechanisms of plasticity. Based upon experiments using single-pulse TMS as a probe, the physiologic mechanism of these effects is often assumed to be via changes in cortical excitability, with 10 Hz rTMS increasing and 1 Hz rTMS decreasing the excitability of the stimulated region. However, the reliability and reproducibility of these rTMS protocols on cortical excitability across and within individual subjects, particularly in comparison to robust sham stimulation, have not been systematically examined. OBJECTIVES In a cohort of 28 subjects (39 ± 16 years), we report the first comprehensive study to (1) assess the neuromodulatory effects of traditional 1 Hz and 10 Hz rTMS on corticospinal excitability against both a robust sham control, and two other widely used patterned rTMS protocols (intermittent theta burst stimulation, iTBS; and continuous theta burst stimulation, cTBS), and (2) determine the reproducibility of all rTMS protocols across identical repeat sessions. RESULTS At the group level, neither 1 Hz nor 10 Hz rTMS significantly modulated corticospinal excitability. 1 Hz and 10 Hz rTMS were also not significantly different from sham and both TBS protocols. Reproducibility was poor for all rTMS protocols except for sham. Importantly, none of the real rTMS and TBS protocols demonstrated greater neuromodulatory effects or reproducibility after controlling for potential experimental factors including baseline corticospinal excitability, TMS coil deviation and the number of individual MEP trials. CONCLUSIONS These results call into question the effectiveness and reproducibility of widely used rTMS techniques for modulating corticospinal excitability, and suggest the need for a fundamental rethinking regarding the potential mechanisms by which rTMS affects brain function and behavior in humans.
Collapse
Affiliation(s)
- Justine Magnuson
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Department of Neurology, Harvard Medical School, Boston, MA, United States of America
- Health and Exercise Sciences, University of British Columbia Okanagan, Kelowna, BC, CA
| | - Mehmet A. Ozdemir
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Department of Neurology, Harvard Medical School, Boston, MA, United States of America
- Department of Biomedical Engineering, Izmir Katip Celebi University, Izmir, Turkey
| | - Elon Mathieson
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Sofia Kirkman
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Brice Passera
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Department of Neurology, Harvard Medical School, Boston, MA, United States of America
| | - Sumientra Rampersad
- Department of Physics, University of Massachusetts, Boston, MA, United States of America
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, United States of America
| | - Alyssa B. Dufour
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
- Hinda and Arthur Marcus Institute for Aging Research, Hebrew Senior Life, Boston, MA, United States of America
| | - Dana Brooks
- Department of Electrical and Computer Engineering, Northeastern University, Boston, MA, United States of America
| | - Alvaro Pascual-Leone
- Department of Neurology, Harvard Medical School, Boston, MA, United States of America
- Hinda and Arthur Marcus Institute for Aging Research and Deanne and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, United States of America
- Guttmann Brain Health Institute, Institut Guttmann de Neurorehabilitació, Universitat Autonoma de Barcelona, Badalona, Spain
| | - Peter J. Fried
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Department of Neurology, Harvard Medical School, Boston, MA, United States of America
| | - Mouhsin M. Shafi
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Department of Neurology, Harvard Medical School, Boston, MA, United States of America
| | - Recep A. Ozdemir
- Berenson-Allen Center for Noninvasive Brain Stimulation, Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
- Department of Neurology, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
15
|
Chemogenetic emulation of intraneuronal oxidative stress affects synaptic plasticity. Redox Biol 2023; 60:102604. [PMID: 36640726 PMCID: PMC9852792 DOI: 10.1016/j.redox.2023.102604] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/11/2022] [Accepted: 01/07/2023] [Indexed: 01/11/2023] Open
Abstract
Oxidative stress, a state of disrupted redox signaling, reactive oxygen species (ROS) overproduction, and oxidative cell damage, accompanies numerous brain pathologies, including aging-related dementia and Alzheimer's disease, the most common neurodegenerative disorder of the elderly population. However, a causative role of neuronal oxidative stress in the development of aging-related cognitive decline and neurodegeneration remains elusive because of the lack of approaches for modeling isolated oxidative injury in the brain. Here, we present a chemogenetic approach based on the yeast flavoprotein d-amino acid oxidase (DAAO) for the generation of intraneuronal hydrogen peroxide (H2O2). To validate this chemogenetic tool, DAAO and HyPer7, an ultrasensitive genetically encoded H2O2 biosensor, were targeted to neurons. Changes in the fluorescence of HyPer7 upon treatment of neurons expressing DAAO with d-norvaline (D-Nva), a DAAO substrate, confirmed chemogenetically induced production of intraneuornal H2O2. Then, using the verified chemogenetic tool, we emulated isolated intraneuronal oxidative stress in acute brain slices and, using electrophysiological recordings, revealed that it does not alter basal synaptic transmission and the probability of neurotransmitter release from presynaptic terminals but reduces long-term potentiation (LTP). Moreover, treating neurons expressing DAAO with D-Nva via the patch pipette also decreases LTP. This observation indicates that isolated oxidative stress affects synaptic plasticity at single cell level. Our results broaden the toolset for studying normal redox regulation in the brain and elucidating the role of oxidative stress to the pathogenesis of cognitive aging and the early stages of aging-related neurodegenerative diseases. The proposed approach is useful for identification of early markers of neuronal oxidative stress and may be used in screens of potential antioxidants effective against neuronal oxidative injury.
Collapse
|
16
|
Fyn nanoclustering requires switching to an open conformation and is enhanced by FTLD-Tau biomolecular condensates. Mol Psychiatry 2023; 28:946-962. [PMID: 36258016 PMCID: PMC9908554 DOI: 10.1038/s41380-022-01825-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/08/2022]
Abstract
Fyn is a Src kinase that controls critical signalling cascades and has been implicated in learning and memory. Postsynaptic enrichment of Fyn underpins synaptotoxicity in dementias such as Alzheimer's disease and frontotemporal lobar degeneration with Tau pathology (FTLD-Tau). The FLTD P301L mutant Tau is associated with a higher propensity to undergo liquid-liquid phase separation (LLPS) and form biomolecular condensates. Expression of P301L mutant Tau promotes aberrant trapping of Fyn in nanoclusters within hippocampal dendrites by an unknown mechanism. Here, we used single-particle tracking photoactivated localisation microscopy to demonstrate that the opening of Fyn into its primed conformation promotes its nanoclustering in dendrites leading to increased Fyn/ERK/S6 downstream signalling. Preventing the auto-inhibitory closed conformation of Fyn through phospho-inhibition or through perturbation of its SH3 domain increased Fyn's nanoscale trapping, whereas inhibition of the catalytic domain had no impact. By combining pharmacological and genetic approaches, we demonstrate that P301L Tau enhanced both Fyn nanoclustering and Fyn/ERK/S6 signalling via its ability to form biomolecular condensates. Together, our findings demonstrate that Fyn alternates between a closed and an open conformation, the latter being enzymatically active and clustered. Furthermore, pathogenic immobilisation of Fyn relies on the ability of P301L Tau to form biomolecular condensates, thus highlighting the critical importance of LLPS in controlling nanoclustering and downstream intracellular signalling events.
Collapse
|
17
|
Fortner A, Chera A, Tanca A, Bucur O. Apoptosis regulation by the tyrosine-protein kinase CSK. Front Cell Dev Biol 2022; 10:1078180. [PMID: 36578781 PMCID: PMC9792154 DOI: 10.3389/fcell.2022.1078180] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
C-terminal Src kinase (CSK) is a cytosolic tyrosine-protein kinase with an important role in regulating critical cellular decisions, such as cellular apoptosis, survival, proliferation, cytoskeletal organization and many others. Current knowledge on the CSK mechanisms of action, regulation and functions is still at an early stage, most of CSK's known actions and functions being mediated by the negative regulation of the SRC family of tyrosine kinases (SFKs) through phosphorylation. As SFKs play a vital role in apoptosis, cell proliferation and survival regulation, SFK inhibition by CSK has a pro-apoptotic effect, which is mediated by the inhibition of cellular signaling cascades controlled by SFKs, such as the MAPK/ERK, STAT3 and PI3K/AKT signaling pathways. Abnormal functioning of CSK and SFK activation can lead to diseases such as cancer, cardiovascular and neurological manifestations. This review describes apoptosis regulation by CSK, CSK inhibition of the SFKs and further explores the clinical relevance of CSK in important pathologies, such as cancer, autoimmune, autoinflammatory, neurologic diseases, hypertension and HIV/AIDS.
Collapse
Affiliation(s)
- Andra Fortner
- Victor Babes National Institute of Pathology, Bucharest, Romania,Medical School, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - Alexandra Chera
- Victor Babes National Institute of Pathology, Bucharest, Romania,Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Antoanela Tanca
- Victor Babes National Institute of Pathology, Bucharest, Romania,Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania,*Correspondence: Octavian Bucur, ; Antoanela Tanca,
| | - Octavian Bucur
- Victor Babes National Institute of Pathology, Bucharest, Romania,Viron Molecular Medicine Institute, Boston, MA, United States,*Correspondence: Octavian Bucur, ; Antoanela Tanca,
| |
Collapse
|
18
|
Dumrongprechachan V, Salisbury RB, Butler L, MacDonald ML, Kozorovitskiy Y. Dynamic proteomic and phosphoproteomic atlas of corticostriatal axons in neurodevelopment. eLife 2022; 11:e78847. [PMID: 36239373 PMCID: PMC9629834 DOI: 10.7554/elife.78847] [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: 03/22/2022] [Accepted: 10/12/2022] [Indexed: 11/17/2022] Open
Abstract
Mammalian axonal development begins in embryonic stages and continues postnatally. After birth, axonal proteomic landscape changes rapidly, coordinated by transcription, protein turnover, and post-translational modifications. Comprehensive profiling of axonal proteomes across neurodevelopment is limited, with most studies lacking cell-type and neural circuit specificity, resulting in substantial information loss. We create a Cre-dependent APEX2 reporter mouse line and map cell-type-specific proteome of corticostriatal projections across postnatal development. We synthesize analysis frameworks to define temporal patterns of axonal proteome and phosphoproteome, identifying co-regulated proteins and phosphorylations associated with genetic risk for human brain disorders. We discover proline-directed kinases as major developmental regulators. APEX2 transgenic reporter proximity labeling offers flexible strategies for subcellular proteomics with cell type specificity in early neurodevelopment, a critical period for neuropsychiatric disease.
Collapse
Affiliation(s)
- Vasin Dumrongprechachan
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
- The Chemistry of Life Processes Institute, Northwestern UniversityEvanstonUnited States
| | - Ryan B Salisbury
- Department of Psychiatry, University of PittsburghPittsburghUnited States
| | - Lindsey Butler
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | | | - Yevgenia Kozorovitskiy
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
- The Chemistry of Life Processes Institute, Northwestern UniversityEvanstonUnited States
| |
Collapse
|
19
|
An opinion on the debatable function of brain resident immune protein, T-cell receptor beta subunit in the central nervous system. IBRO Neurosci Rep 2022; 13:235-242. [PMID: 36590097 PMCID: PMC9795316 DOI: 10.1016/j.ibneur.2022.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 09/02/2022] [Indexed: 01/04/2023] Open
Abstract
In recent years scientific research has established that the nervous and immune systems have shared molecular signaling components. Proteins native to immune cells, which are also found in the brain, have neuronal functions in the nervous system where they affect synaptic plasticity, axonal regeneration, neurogenesis, and neurotransmission. Certain native immune molecules like major histocompatibility complex I (MHC-I), paired immunoglobulin receptor B (PirB), toll-like receptor (TLR), cluster of differentiation-3 zeta (CD3ζ), CD4 co-receptor, and T-cell receptor beta (TCR-β) expression in neurons have been extensively documented. In this review, we provide our opinion and discussed the possible roles of T-cell receptor beta subunits in modulating the function of neurons in the central nervous system. Based on the previous findings of Syken and Shatz., 2003; Nishiyori et al., 2004; Rodriguez et., 1993 and Komal et., 2014; we discuss whether restrictive expression of TCR-β subunits in selected brain regions could be involved in the pathology of neurological disorders and whether their aberrant enhancement in expression may be considered as a suitable biomarker for aging or neurodegenerative diseases like Huntington's disease (HD).
Collapse
|
20
|
A missense mutation in Kcnc3 causes hippocampal learning deficits in mice. Proc Natl Acad Sci U S A 2022; 119:e2204901119. [PMID: 35881790 PMCID: PMC9351536 DOI: 10.1073/pnas.2204901119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Although a wide variety of genetic tools has been developed to study learning and memory, the molecular basis of memory encoding remains incompletely understood. Here, we undertook an unbiased approach to identify novel genes critical for memory encoding. From a large-scale, in vivo mutagenesis screen using contextual fear conditioning, we isolated in mice a mutant, named Clueless, with spatial learning deficits. A causative missense mutation (G434V) was found in the voltage-gated potassium channel, subfamily C member 3 (Kcnc3) gene in a region that encodes a transmembrane voltage sensor. Generation of a Kcnc3G434V CRISPR mutant mouse confirmed this mutation as the cause of the learning defects. While G434V had no effect on transcription, translation, or trafficking of the channel, electrophysiological analysis of the G434V mutant channel revealed a complete loss of voltage-gated conductance, a broadening of the action potential, and decreased neuronal firing. Together, our findings have revealed a role for Kcnc3 in learning and memory.
Collapse
|
21
|
Muthukumar S, Mehrotra K, Fouda M, Hamimi S, Jantzie LL, Robinson S. Prenatal and postnatal insults differentially contribute to executive function and cognition: Utilizing touchscreen technology for perinatal brain injury research. Exp Neurol 2022; 354:114104. [PMID: 35525306 PMCID: PMC10085749 DOI: 10.1016/j.expneurol.2022.114104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 04/27/2022] [Accepted: 04/30/2022] [Indexed: 12/19/2022]
Abstract
The use of touchscreen technology to evaluate cognitive deficits in animal models has grown tremendously over the past 20 years. The touchscreen apparatus encompasses many advantages, namely a high level of standardization and translational capability. Improvements in technology in recent years have expanded the versatility of the touchscreen platform, as it is able to test distinct cognitive modalities including working memory, attention, discrimination, and association. Importantly, touchscreen technology has allowed researchers to explore deficits in multiple pillars of cognition in a wide variety of perinatal disorders with neurological sequelae across critical developmental windows. The touchscreen platform has been used to dissect deficits in antenatal CNS injury including fetal alcohol syndrome, prenatal opioid exposure, and chorioamnionitis, to peripartum insults such as term hypoxic-ischemic encephalopathy, to early postnatal insults including infantile traumatic brain injury. Most importantly, touchscreen technology offers the sensitivity necessary to detect subtle injury and treatment-induced changes in cognition and executive function beyond those offered by more rudimentary tests of rodent cognition. Understanding the pathophysiology of these disorders in rodents is paramount to addressing these deficits in human infants and dissecting the neural circuitry essential to perinatal brain injury pathophysiology and responsiveness to novel therapeutics. Touchscreen testing provides an effective, facile, sophisticated technique to accelerate the goal of improving cognitive and behavioral outcomes of children who suffer perinatal brain injury.
Collapse
Affiliation(s)
- Sankar Muthukumar
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Karnika Mehrotra
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mohammed Fouda
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah Hamimi
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lauren L Jantzie
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Kennedy Krieger Institute, Baltimore, MD, USA
| | - Shenandoah Robinson
- Department of Neurosurgery, Division of Pediatric Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Division of Neonatology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Li H, Zhou X, Chen R, Xiao Y, Zhou T. The Src-Kinase Fyn is Required for Cocaine-Associated Memory Through Regulation of Tau. Front Pharmacol 2022; 13:769827. [PMID: 35185557 PMCID: PMC8850722 DOI: 10.3389/fphar.2022.769827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 01/10/2022] [Indexed: 11/24/2022] Open
Abstract
Drug-associated context-induced relapse of cocaine-seeking behaviour requires the retrieval of drug-associated memory. Studies exploring the underlying neurobiological mechanism of drug memory formation will likely contribute to the development of treatments for drug addiction and the prevention of relapse. In our study, we applied a cocaine-conditioned place preference (CPP) paradigm and a self-administration paradigm (two drug-associated memory formation model) to confirm the hypothesis that the Src kinase Fyn critically regulates cocaine-associated memory formation in the hippocampus. For this experiment, we administered the Src kinase inhibitor PP2 into the bilateral hippocampus before cocaine-CPP and self-administration training, and the results showed that pharmacological manipulation of the Src kinase Fyn activity significantly attenuated the response to cocaine-paired cues in the cocaine-CPP and self-administration paradigms, indicating that hippocampal Fyn activity contributes to cocaine-associated memory formation. In addition, the regulation of cocaine-associated memory formation by Fyn depends on Tau expression, as restoring Tau to normal levels disrupted cocaine memory formation. Together, these results indicate that hippocampal Fyn activity plays a key role in the formation of cocaine-associated memory, which underlies cocaine-associated contextual stimulus-mediated regulation of cocaine-seeking behaviour, suggesting that Fyn represents a promising therapeutic target for weakening cocaine-related memory and treating cocaine addiction.
Collapse
Affiliation(s)
- Hongchun Li
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Hongchun Li,
| | - Xinglong Zhou
- Department of Respiratory and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Chen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuzhou Xiao
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhou
- Department of Drug and Equipment, China Rongtong Bayi Orthopaedic Hospital, Chengdu, China
| |
Collapse
|
23
|
Kimura Y, Hayashi Y, Hitomi S, Ikutame D, Urata K, Shibuta I, Sakai A, Ni J, Iwata K, Tonogi M, Shinoda M. IL-33 induces orofacial neuropathic pain through Fyn-dependent phosphorylation of GluN2B in the trigeminal spinal subnucleus caudalis. Brain Behav Immun 2022; 99:266-280. [PMID: 34715301 DOI: 10.1016/j.bbi.2021.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 10/14/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Orofacial neuropathic pain can cause considerable disruptions in patients' daily lives, especially because of a lack of effective medications as its underlying causative mechanisms are not fully understood. Here, we found neuron-specific expression of the interleukin (IL)-33 receptor in the trigeminal spinal subnucleus caudalis (Vc), distinct from the spinal dorsal horn. Reduction in head withdrawal threshold in response to von Frey filament stimulation of the whisker pad skin was inversely correlated with the upregulation of IL-33 in the Vc after infraorbital nerve injury (IONI). Neutralization of IL-33 in the Vc alleviated mechanical allodynia in the whisker pad skin after IONI; conversely, intracisternal administration of IL-33 elicited mechanical allodynia in the whisker pad skin, which was relieved by GluN2B antagonism. Moreover, IL-33 triggered the potentiation of GluN2B-containing N-methyl-D-aspartate receptor-mediated synaptic currents and phosphorylation of synaptosomal GluN2B in the Vc, whereas IONI-induced GluN2B phosphorylation was inhibited by neutralization of IL-33 in the Vc. IL-33-induced GluN2B phosphorylation was mediated by phosphorylation of Fyn kinase, and inhibition of the Fyn kinase pathway prevented the development of IL-33-induced mechanical allodynia. Our findings provide insights into a new mechanism by which IL-33 directly regulates synaptic transmission and suggest that IL-33 signaling could be a candidate target for therapeutic interventions for orofacial neuropathic pain.
Collapse
Affiliation(s)
- Yuki Kimura
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry, Tokyo, Japan
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan.
| | - Suzuro Hitomi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Daisuke Ikutame
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan
| | - Kentaro Urata
- Department of Complete Denture Prosthodontics, Nihon University School of Dentistry, Tokyo, Japan
| | - Ikuko Shibuta
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Atsushi Sakai
- Department of Pharmacology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy in the Ministry of Industry and Information Technology, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Koichi Iwata
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| | - Morio Tonogi
- Department of Oral and Maxillofacial Surgery, Nihon University School of Dentistry, Tokyo, Japan
| | - Masamichi Shinoda
- Department of Physiology, Nihon University School of Dentistry, Tokyo, Japan
| |
Collapse
|
24
|
Gage M, Putra M, Wachter L, Dishman K, Gard M, Gomez-Estrada C, Thippeswamy T. Saracatinib, a Src Tyrosine Kinase Inhibitor, as a Disease Modifier in the Rat DFP Model: Sex Differences, Neurobehavior, Gliosis, Neurodegeneration, and Nitro-Oxidative Stress. Antioxidants (Basel) 2021; 11:61. [PMID: 35052568 PMCID: PMC8773289 DOI: 10.3390/antiox11010061] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/17/2021] [Accepted: 12/23/2021] [Indexed: 12/13/2022] Open
Abstract
Diisopropylfluorophosphate (DFP), an organophosphate nerve agent (OPNA), exposure causes status epilepticus (SE) and epileptogenesis. In this study, we tested the protective effects of saracatinib (AZD0530), a Src kinase inhibitor, in mixed-sex or male-only Sprague Dawley rats exposed to 4-5 mg/kg DFP followed by 2 mg/kg atropine and 25 mg/kg 2-pralidoxime. Midazolam (3 mg/kg) was given to the mixed-sex cohort (1 h post-DFP) and male-only cohort (~30 min post-DFP). Saracatinib (20 mg/kg, oral, daily for 7 days) or vehicle was given two hours later and euthanized eight days or ten weeks post-DFP. Brain immunohistochemistry (IHC) showed increased microgliosis, astrogliosis, and neurodegeneration in DFP-treated animals. In the 10-week post-DFP male-only group, there were no significant differences between groups in the novel object recognition, Morris water maze, rotarod, or forced swim test. Brain IHC revealed significant mitigation by saracatinib in contrast to vehicle-treated DFP animals in microgliosis, astrogliosis, neurodegeneration, and nitro-oxidative stressors, such as inducible nitric oxide synthase, GP91phox, and 3-Nitrotyrosine. These findings suggest the protective effects of saracatinib on brain pathology seem to depend on the initial SE severity. Further studies on dose optimization, including extended treatment regimen depending on the SE severity, are required to determine its disease-modifying potential in OPNA models.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Thimmasettappa Thippeswamy
- Department of Biomedical Sciences and Interdepartmental Neuroscience Program, Iowa State University, Ames, IA 50011, USA; (M.G.); (M.P.); (L.W.); (K.D.); (M.G.); (C.G.-E.)
| |
Collapse
|
25
|
Proskura AL, Islamova MY, Vechkapova SO. Cross-Talk of the Glutamate and Leptin Receptor Pathways. Mol Biol 2021. [DOI: 10.1134/s0026893321020291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
Boyer O, Mollet G, Dorval G. Neurological involvement in monogenic podocytopathies. Pediatr Nephrol 2021; 36:3571-3583. [PMID: 33791874 DOI: 10.1007/s00467-020-04903-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/27/2020] [Accepted: 12/11/2020] [Indexed: 01/22/2023]
Abstract
Genetic studies of hereditary nephrotic syndrome (NS) have identified more than 50 genes that, if mutated, are responsible for monogenic forms of steroid-resistant NS (SRNS), either isolated or syndromic. Most of these genes encode proteins expressed in the podocyte with various functions such as transcription factors, mitochondrial proteins, or enzymes, but mainly structural proteins of the slit diaphragm (SD) as well as cytoskeletal binding and regulator proteins. Syndromic NS is sometimes associated with neurological features. Over recent decades, various studies have established links between the physiology of podocytes and neurons, both morphologically (slit diaphragm and synapse) and functionally (signaling platforms). Variants in genes expressed in different compartments of the podocyte and neurons are responsible for phenotypes associating kidney lesions with proteinuria (mainly Focal and Segmental Glomerulosclerosis (FSGS) or Diffuse Mesangial Sclerosis (DMS)) and central and/or peripheral neurological disorders. The Galloway-Mowat syndrome (GAMOS, OMIM#251300) associates neurological defects, microcephaly, and proteinuria and is caused by variants in genes encoding proteins of various functions (microtubule cytoskeleton regulation (WDR73), regulation of protein synthesis via transfer RNAs (KEOPS and WDR4 complexes)). Pierson syndrome (OMIM#609049) associating congenital nephrotic syndrome and central neurological and ophthalmological anomalies is secondary to variants in LAMB2, involved in glomerular and ocular basement membranes. Finally, Charcot-Marie-Tooth-FSGS (OMIM#614455) combines peripheral sensory-motor neuropathy and proteinuria and arises from INF2 variants, resulting in cytoskeletal polymerization defects. This review focuses on genetic syndromes associating nephrotic range proteinuria and neurological involvement and provides the latest advances in the description of these neuro-renal disorders.
Collapse
Affiliation(s)
- Olivia Boyer
- Service de Néphrologie Pédiatrique, AP-HP, Centre de Référence de maladies rénales rares de l'enfant et de l'adulte (MARHEA), Hôpital Necker - Enfants Malades, 149 Rue de Sèvres, 75015, Paris, France.
- Institut Imagine, Laboratoire des maladies rénales héréditaires, INSERM UMR 1163, Université de Paris, Paris, France.
| | - Géraldine Mollet
- Institut Imagine, Laboratoire des maladies rénales héréditaires, INSERM UMR 1163, Université de Paris, Paris, France
| | - Guillaume Dorval
- Institut Imagine, Laboratoire des maladies rénales héréditaires, INSERM UMR 1163, Université de Paris, Paris, France
- Service de Génétique Moléculaire, AP-HP, Hôpital Necker-Enfants Malades, Paris, France
| |
Collapse
|
27
|
de Pins B, Mendes T, Giralt A, Girault JA. The Non-receptor Tyrosine Kinase Pyk2 in Brain Function and Neurological and Psychiatric Diseases. Front Synaptic Neurosci 2021; 13:749001. [PMID: 34690733 PMCID: PMC8527176 DOI: 10.3389/fnsyn.2021.749001] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/14/2021] [Indexed: 12/28/2022] Open
Abstract
Pyk2 is a non-receptor tyrosine kinase highly enriched in forebrain neurons. Pyk2 is closely related to focal adhesion kinase (FAK), which plays an important role in sensing cell contacts with extracellular matrix and other extracellular signals controlling adhesion and survival. Pyk2 shares some of FAK’s characteristics including recruitment of Src-family kinases after autophosphorylation, scaffolding by interacting with multiple partners, and activation of downstream signaling pathways. Pyk2, however, has the unique property to respond to increases in intracellular free Ca2+, which triggers its autophosphorylation following stimulation of various receptors including glutamate NMDA receptors. Pyk2 is dephosphorylated by the striatal-enriched phosphatase (STEP) that is highly expressed in the same neuronal populations. Pyk2 localization in neurons is dynamic, and altered following stimulation, with post-synaptic and nuclear enrichment. As a signaling protein Pyk2 is involved in multiple pathways resulting in sometimes opposing functions depending on experimental models. Thus Pyk2 has a dual role on neurites and dendritic spines. With Src family kinases Pyk2 participates in postsynaptic regulations including of NMDA receptors and is necessary for specific types of synaptic plasticity and spatial memory tasks. The diverse functions of Pyk2 are also illustrated by its role in pathology. Pyk2 is activated following epileptic seizures or ischemia-reperfusion and may contribute to the consequences of these insults whereas Pyk2 deficit may contribute to the hippocampal phenotype of Huntington’s disease. Pyk2 gene, PTK2B, is associated with the risk for late-onset Alzheimer’s disease. Studies of underlying mechanisms indicate a complex contribution with involvement in amyloid toxicity and tauopathy, combined with possible functional deficits in neurons and contribution in microglia. A role of Pyk2 has also been proposed in stress-induced depression and cocaine addiction. Pyk2 is also important for the mobility of astrocytes and glioblastoma cells. The implication of Pyk2 in various pathological conditions supports its potential interest for therapeutic interventions. This is possible through molecules inhibiting its activity or increasing it through inhibition of STEP or other means, depending on a precise evaluation of the balance between positive and negative consequences of Pyk2 actions.
Collapse
Affiliation(s)
- Benoit de Pins
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Tiago Mendes
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| | - Albert Giralt
- Departament de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain.,Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
| | - Jean-Antoine Girault
- Institut du Fer à Moulin, Paris, France.,Inserm UMR-S 1270, Paris, France.,Faculté des Sciences et Ingénierie, Sorbonne Université, Paris, France
| |
Collapse
|
28
|
Yasuda G, Moriuchi E, Hamanaka R, Fujishita A, Yoshimi T, Yamamoto K, Hayashida K, Koga Y, Yoshida N. Visualization of mandibular movement relative to the maxilla during mastication in mice: integration of kinematic analysis and reconstruction of a three-dimensional model of the maxillofacial structure. BMC Oral Health 2021; 21:527. [PMID: 34649558 PMCID: PMC8515672 DOI: 10.1186/s12903-021-01879-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/28/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mastication is one of the most fundamental functions for the conservation of human life. To clarify the pathogenetic mechanism of various oral dysfunctions, the demand for devices for evaluating stomatognathic function has been increasing. The aim of the present study was to develop a system to reconstruct and visualize 3-dimensional (3D) mandibular movements relative to the maxilla, including dynamic transition of occlusal contacts between the upper and lower dentitions during mastication in mice. METHODS First, mandibular movements with six degrees of freedom were measured using a motion capture system comprising two high-speed cameras and four reflective markers. Second, 3D models of maxillofacial structure were reconstructed from micro-computed tomography images. Movement trajectories of anatomical landmark points on the mandible were then reproduced by integrating the kinematic data of mandibular movements with the anatomical data of maxillofacial structures. Lastly, 3D surface images of the upper dentition with the surrounding maxillofacial structures were transferred to each of the motion capture images to reproduce mandibular movements relative to the maxilla. We also performed electromyography (EMG) of masticatory muscles associated with mandibular movements. RESULTS The developed system could reproduce the 3D movement trajectories of arbitrary points on the mandible, such as incisor, molars and condylar points with high accuracy and could visualize dynamic transitions of occlusal contacts between upper and lower teeth associated with mandibular movements. CONCLUSIONS The proposed system has potential to elucidate the mechanisms underlying motor coordination of masticatory muscles and to clarify their roles during mastication by taking advantage of the capability to record EMG data synchronously with mandibular movements. Such insights will enhance our understanding of the pathogenesis and diagnosis of oral motor disorders by allowing comparisons between normal mice and genetically modified mice with oral behavioral dysfunctions.
Collapse
Affiliation(s)
- Go Yasuda
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Emi Moriuchi
- Department of Orthodontics, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Ryo Hamanaka
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Ayumi Fujishita
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Tomoko Yoshimi
- Department of Orthodontics, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Kana Yamamoto
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Kaori Hayashida
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Yoshiyuki Koga
- Department of Orthodontics, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan
| | - Noriaki Yoshida
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, 1-7-1 Sakamoto, Nagasaki, 852-8588, Japan.
| |
Collapse
|
29
|
Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: A pathological or physiological phenomenon? Acta Neuropathol Commun 2021; 9:149. [PMID: 34503576 PMCID: PMC8428049 DOI: 10.1186/s40478-021-01246-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss the synaptic aspects of Tau pathology occurring during Alzheimer's disease (AD) and how this may relate to memory impairment, a major hallmark of AD. Whilst the clinical diagnosis of AD patients is a loss of working memory and long-term declarative memory, the histological diagnosis is the presence of neurofibrillary tangles of hyperphosphorylated Tau and Amyloid-beta plaques. Tau pathology spreads through synaptically connected neurons to impair synaptic function preceding the formation of neurofibrillary tangles, synaptic loss, axonal retraction and cell death. Alongside synaptic pathology, recent data suggest that Tau has physiological roles in the pre- or post- synaptic compartments. Thus, we have seen a shift in the research focus from Tau as a microtubule-stabilising protein in axons, to Tau as a synaptic protein with roles in accelerating spine formation, dendritic elongation, and in synaptic plasticity coordinating memory pathways. We collate here the myriad of emerging interactions and physiological roles of synaptic Tau, and discuss the current evidence that synaptic Tau contributes to pathology in AD.
Collapse
|
30
|
Demuro S, Di Martino RMC, Ortega JA, Cavalli A. GSK-3β, FYN, and DYRK1A: Master Regulators in Neurodegenerative Pathways. Int J Mol Sci 2021; 22:9098. [PMID: 34445804 PMCID: PMC8396491 DOI: 10.3390/ijms22169098] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/14/2022] Open
Abstract
Protein kinases (PKs) have been recognized as central nervous system (CNS)-disease-relevant targets due to their master regulatory role in different signal transduction cascades in the neuroscience space. Among them, GSK-3β, FYN, and DYRK1A play a crucial role in the neurodegeneration context, and the deregulation of all three PKs has been linked to different CNS disorders with unmet medical needs, including Alzheimer's disease (AD), Parkinson's disease (PD), frontotemporal lobar degeneration (FTLD), and several neuromuscular disorders. The multifactorial nature of these diseases, along with the failure of many advanced CNS clinical trials, and the lengthy approval process of a novel CNS drug have strongly limited the CNS drug discovery. However, in the near-decade from 2010 to 2020, several computer-assisted drug design strategies have been combined with synthetic efforts to develop potent and selective GSK-3β, FYN, and DYRK1A inhibitors as disease-modifying agents. In this review, we described both structural and functional aspects of GSK-3β, FYN, and DYRK1A and their involvement and crosstalk in different CNS pathological signaling pathways. Moreover, we outlined attractive medicinal chemistry approaches including multi-target drug design strategies applied to overcome some limitations of known PKs inhibitors and discover improved modulators with suitable blood-brain barrier (BBB) permeability and drug-like properties.
Collapse
Affiliation(s)
- Stefania Demuro
- Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy; (S.D.); (R.M.C.D.M.); (J.A.O.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Rita M. C. Di Martino
- Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy; (S.D.); (R.M.C.D.M.); (J.A.O.)
| | - Jose A. Ortega
- Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy; (S.D.); (R.M.C.D.M.); (J.A.O.)
| | - Andrea Cavalli
- Computational and Chemical Biology, Italian Institute of Technology, 16163 Genoa, Italy; (S.D.); (R.M.C.D.M.); (J.A.O.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
31
|
Pyk2 in dorsal hippocampus plays a selective role in spatial memory and synaptic plasticity. Sci Rep 2021; 11:16357. [PMID: 34381140 PMCID: PMC8358019 DOI: 10.1038/s41598-021-95813-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
Pyk2 is a Ca2+-activated non-receptor tyrosine kinase enriched in the forebrain, especially in pyramidal neurons of the hippocampus. Previous reports suggested its role in hippocampal synaptic plasticity and spatial memory but with contradictory findings possibly due to experimental conditions. Here we address this issue and show that novel object location, a simple test of spatial memory induced by a single training session, is altered in Pyk2 KO mice and that re-expression of Pyk2 in the dorsal hippocampus corrects this deficit. Bilateral targeted deletion of Pyk2 in dorsal hippocampus CA1 region also alters novel object location. Long term potentiation (LTP) in CA1 is impaired in Pyk2 KO mice using a high frequency stimulation induction protocol but not with a theta burst protocol, explaining differences between previous reports. The same selective LTP alteration is observed in mice with Pyk2 deletion in dorsal hippocampus CA1 region. Thus, our results establish the role of Pyk2 in specific aspects of spatial memory and synaptic plasticity and show the dependence of the phenotype on the type of experiments used to reveal it. In combination with other studies, we provide evidence for a selective role of non-receptor tyrosine kinases in specific aspects of hippocampal neurons synaptic plasticity.
Collapse
|
32
|
Lemieux M, Thiry L, Laflamme OD, Bretzner F. Role of DSCAM in the Development of Neural Control of Movement and Locomotion. Int J Mol Sci 2021; 22:ijms22168511. [PMID: 34445216 PMCID: PMC8395195 DOI: 10.3390/ijms22168511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/02/2021] [Accepted: 08/04/2021] [Indexed: 11/30/2022] Open
Abstract
Locomotion results in an alternance of flexor and extensor muscles between left and right limbs generated by motoneurons that are controlled by the spinal interneuronal circuit. This spinal locomotor circuit is modulated by sensory afferents, which relay proprioceptive and cutaneous inputs that inform the spatial position of limbs in space and potential contacts with our environment respectively, but also by supraspinal descending commands of the brain that allow us to navigate in complex environments, avoid obstacles, chase prey, or flee predators. Although signaling pathways are important in the establishment and maintenance of motor circuits, the role of DSCAM, a cell adherence molecule associated with Down syndrome, has only recently been investigated in the context of motor control and locomotion in the rodent. DSCAM is known to be involved in lamination and delamination, synaptic targeting, axonal guidance, dendritic and cell tiling, axonal fasciculation and branching, programmed cell death, and synaptogenesis, all of which can impact the establishment of motor circuits during development, but also their maintenance through adulthood. We discuss herein how DSCAM is important for proper motor coordination, especially for breathing and locomotion.
Collapse
Affiliation(s)
- Maxime Lemieux
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, CHUL-Neurosciences P09800, 2705 boul. Laurier, Québec, QC G1V 4G2, Canada; (M.L.); (L.T.); (O.D.L.)
| | - Louise Thiry
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, CHUL-Neurosciences P09800, 2705 boul. Laurier, Québec, QC G1V 4G2, Canada; (M.L.); (L.T.); (O.D.L.)
| | - Olivier D. Laflamme
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, CHUL-Neurosciences P09800, 2705 boul. Laurier, Québec, QC G1V 4G2, Canada; (M.L.); (L.T.); (O.D.L.)
| | - Frédéric Bretzner
- Centre de Recherche du Centre Hospitalier Universitaire de Québec, CHUL-Neurosciences P09800, 2705 boul. Laurier, Québec, QC G1V 4G2, Canada; (M.L.); (L.T.); (O.D.L.)
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Université Laval, Québec, QC G1V 4G2, Canada
- Correspondence:
| |
Collapse
|
33
|
Briner A, Götz J, Polanco JC. Fyn Kinase Controls Tau Aggregation In Vivo. Cell Rep 2021; 32:108045. [PMID: 32814048 DOI: 10.1016/j.celrep.2020.108045] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 06/14/2020] [Accepted: 07/27/2020] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a proteinopathy exhibiting aggregation of β-amyloid (Aβ) as amyloid plaques and tau as neurofibrillary tangles (NFTs), whereas primary tauopathies display only a tau pathology. Aβ toxicity is mediated by Fyn kinase in a tau-dependent process; however, whether Fyn controls tau pathology in diseases that lack Aβ pathology remains unexplored. To address this, we generate the Tg/Fyn-/- mouse, which couples mutant tau overexpression with Fyn knockout. Surprisingly, Tg/Fyn-/- mice exhibit a near-complete ablation of NFTs, alongside reduced tau hyperphosphorylation, altered tau solubility, and diminished synaptic tau accumulation. Furthermore, Tg/Fyn-/- brain lysates elicit less tau seeding in tau biosensor cells. Lastly, the fibrillization of tau is boosted by its pseudophosphorylation at the Fyn epitope Y18. Together, this identifies Fyn as a key regulator of tau pathology independently of Aβ-induced toxicity and thereby represents a potentially valuable therapeutic target for not only AD but also tauopathies more generally.
Collapse
Affiliation(s)
- Adam Briner
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jürgen Götz
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD 4072, Australia
| | - Juan Carlos Polanco
- Clem Jones Centre for Ageing Dementia Research (CJCADR), Queensland Brain Institute (QBI), The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
34
|
Rajani V, Sengar AS, Salter MW. Src and Fyn regulation of NMDA receptors in health and disease. Neuropharmacology 2021; 193:108615. [PMID: 34051267 DOI: 10.1016/j.neuropharm.2021.108615] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 11/28/2022]
Abstract
The Src family kinases (SFKs) are cytoplasmic non-receptor tyrosine kinases involved in multiple signalling pathways. In the central nervous system (CNS), SFKs are key regulators of N-methyl-d-aspartate receptor (NMDAR) function and major points of convergence for neuronal transduction pathways. Physiological upregulation of NMDAR activity by members of the SFKs, namely Src and Fyn, is crucial for induction of plasticity at Schaffer collateral-CA1 synapses of the hippocampus. Aberrant SFK regulation of NMDARs is implicated in several pathological conditions in the CNS including schizophrenia and pain hypersensitivity. Here, evidence is presented to highlight the current understanding of the intermolecular interactions of SFKs within the NMDAR macromolecular complex, the upstream regulators of SFK activity on NMDAR function and the role Src and Fyn have in synaptic plasticity and metaplasticity. The targeting of SFK protein-protein interactions is discussed as a potential therapeutic strategy to restore signalling activity underlying glutamatergic dysregulation in CNS disease pathophysiology.
Collapse
Affiliation(s)
- Vishaal Rajani
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada; Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Ameet S Sengar
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
35
|
Mueller RL, Combs B, Alhadidy MM, Brady ST, Morfini GA, Kanaan NM. Tau: A Signaling Hub Protein. Front Mol Neurosci 2021; 14:647054. [PMID: 33815057 PMCID: PMC8017207 DOI: 10.3389/fnmol.2021.647054] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Abstract
Over four decades ago, in vitro experiments showed that tau protein interacts with and stabilizes microtubules in a phosphorylation-dependent manner. This observation fueled the widespread hypotheses that these properties extend to living neurons and that reduced stability of microtubules represents a major disease-driving event induced by pathological forms of tau in Alzheimer’s disease and other tauopathies. Accordingly, most research efforts to date have addressed this protein as a substrate, focusing on evaluating how specific mutations, phosphorylation, and other post-translational modifications impact its microtubule-binding and stabilizing properties. In contrast, fewer efforts were made to illuminate potential mechanisms linking physiological and disease-related forms of tau to the normal and pathological regulation of kinases and phosphatases. Here, we discuss published work indicating that, through interactions with various kinases and phosphatases, tau may normally act as a scaffolding protein to regulate phosphorylation-based signaling pathways. Expanding on this concept, we also review experimental evidence linking disease-related tau species to the misregulation of these pathways. Collectively, the available evidence supports the participation of tau in multiple cellular processes sustaining neuronal and glial function through various mechanisms involving the scaffolding and regulation of selected kinases and phosphatases at discrete subcellular compartments. The notion that the repertoire of tau functions includes a role as a signaling hub should widen our interpretation of experimental results and increase our understanding of tau biology in normal and disease conditions.
Collapse
Affiliation(s)
- Rebecca L Mueller
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Benjamin Combs
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States
| | - Mohammed M Alhadidy
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States
| | - Scott T Brady
- Department of Anatomy and Cell Biology, The University of Illinois at Chicago, Chicago, IL, United States.,Marine Biological Laboratory, Woods Hole, MA, United States
| | - Gerardo A Morfini
- Department of Anatomy and Cell Biology, The University of Illinois at Chicago, Chicago, IL, United States.,Marine Biological Laboratory, Woods Hole, MA, United States
| | - Nicholas M Kanaan
- Department of Translational Neuroscience, Michigan State University, Grand Rapids, MI, United States.,Neuroscience Program, Michigan State University, East Lansing, MI, United States.,Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, United States
| |
Collapse
|
36
|
Luo XM, Zhao J, Wu WY, Fu J, Li ZY, Zhang M, Lu J. Post-status epilepticus treatment with the Fyn inhibitor, saracatinib, improves cognitive function in mice. BMC Neurosci 2021; 22:2. [PMID: 33451301 PMCID: PMC7811255 DOI: 10.1186/s12868-020-00606-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023] Open
Abstract
Background Status epilepticus (SE) is a life-threatening neurological disorder. The hippocampus, as an important area of the brain that regulates cognitive function, is usually damaged after SE, and cognitive deficits often result from hippocampal neurons lost after SE. Fyn, a non-receptor Src family of tyrosine kinases, is potentially associated with the onset of seizure. Saracatinib, a Fyn inhibitor, suppresses epileptogenesis and reduces epileptiform spikes. However, whether saracatinib inhibits cognitive deficits after SE is still unknown. Methods In the present study, a pilocarpine-induced SE mouse model was used to answer this question by using the Morris water maze and normal object recognition behavioral tests. Results We found that saracatinib inhibited the loss in cognitive function following SE. Furthermore, we found that the number of hippocampal neurons in the saracatinib treatment group was increased, when compared to the SE group. Conclusions These results showed that saracatinib can improve cognitive functions by reducing the loss of hippocampal neurons after SE, suggesting that Fyn dysfunction is involved in cognitive deficits after SE, and that the inhibition of Fyn is a possible treatment to improve cognitive function in SE patients.
Collapse
Affiliation(s)
- Xin-Ming Luo
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China. .,Institute of Neuroscience, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Jing Zhao
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Wen-Yue Wu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Jie Fu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Zheng-Yu Li
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Ming Zhang
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Jie Lu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Nanchang, 330006, Jiangxi, China
| |
Collapse
|
37
|
Gage MC, Thippeswamy T. Inhibitors of Src Family Kinases, Inducible Nitric Oxide Synthase, and NADPH Oxidase as Potential CNS Drug Targets for Neurological Diseases. CNS Drugs 2021; 35:1-20. [PMID: 33515429 PMCID: PMC7893831 DOI: 10.1007/s40263-020-00787-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 12/21/2022]
Abstract
Neurological diseases share common neuroinflammatory and oxidative stress pathways. Both phenotypic and molecular changes in microglia, astrocytes, and neurons contribute to the progression of disease and present potential targets for disease modification. Src family kinases (SFKs) are present in both neurons and glial cells and are upregulated following neurological insults in both human and animal models. In neurons, SFKs interact with post-synaptic protein domains to mediate hyperexcitability and neurotoxicity. SFKs are upstream of signaling cascades that lead to the modulation of neurotransmitter receptors and the transcription of pro-inflammatory cytokines as well as producers of free radicals through the activation of glia. Inducible nitric oxide synthase (iNOS/NOS-II) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2), the major mediators of reactive nitrogen/oxygen species (RNS/ROS) production in the brain, are also upregulated along with the pro-inflammatory cytokines following neurological insult and contribute to disease progression. Persistent neuronal hyperexcitability, RNS/ROS, and cytokines can exacerbate neurodegeneration, a common pathognomonic feature of the most prevalent neurological disorders such as Alzheimer's disease, Parkinson's disease, and epilepsy. Using a wide variety of preclinical disease models, inhibitors of the SFK-iNOS-NOX2 signaling axis have been tested to cure or modify disease progression. In this review, we discuss the SFK-iNOS-NOX2 signaling pathway and their inhibitors as potential CNS targets for major neurological diseases.
Collapse
|
38
|
Ehinger Y, Morisot N, Phamluong K, Sakhai SA, Soneja D, Adrover MF, Alvarez VA, Ron D. cAMP-Fyn signaling in the dorsomedial striatum direct pathway drives excessive alcohol use. Neuropsychopharmacology 2021; 46:334-342. [PMID: 32417851 PMCID: PMC7852539 DOI: 10.1038/s41386-020-0712-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/11/2020] [Indexed: 12/19/2022]
Abstract
Fyn kinase in the dorsomedial striatum (DMS) of rodents plays a central role in mechanisms underlying excessive alcohol intake. The DMS is comprised of medium spiny neurons (MSNs) that project directly (dMSNs) or indirectly (iMSNs) to the substantia nigra. Here, we examined the cell-type specificity of Fyn's actions in alcohol use. First, we knocked down Fyn selectively in DMS dMSNs or iMSNs of mice and measured the level of alcohol consumption. We found that downregulation of Fyn in dMSNs, but not in iMSNs, reduces excessive alcohol but not saccharin intake. D1Rs are coupled to Gαs/olf, which activate cAMP signaling. To examine whether Fyn's actions are mediated through cAMP signaling, DMS dMSNs were infected with GαsDREADD, and the activation of Fyn signaling was measured following CNO treatment. We found that remote stimulation of cAMP signaling in DMS dMSNs activates Fyn and promotes the phosphorylation of the Fyn substrate, GluN2B. In contract, remote activation of GαsDREADD in DLS dMSNs did not alter Fyn signaling. We then tested whether activation of GαsDREADD in DMS dMSNs or iMSNs alters alcohol intake and observed that CNO-dependent activation of GαsDREADD in DMS dMSNs but not iMSNs increases alcohol but not saccharin intake. Finally, we examined the contribution of Fyn to GαsDREADD-dependent increase in alcohol intake, and found that systemic administration of the Fyn inhibitor, AZD0503 blocks GαsDREADD-dependent increase in alcohol consumption. Our results suggest that the cAMP-Fyn axis in the DMS dMSNs is a molecular transducer of mechanisms underlying the development of excessive alcohol consumption.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Nadege Morisot
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Nkarta Therapeutics, San Francisco, CA, USA
| | - Khanhky Phamluong
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Samuel A Sakhai
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
- Sage Therapeutics, San Francisco, CA, USA
| | - Drishti Soneja
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA
| | - Martin F Adrover
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- INGEBI, CONICET, Buenos Aires, Argentina
| | - Veronica A Alvarez
- National Institutes of Alcohol Abuse and Alcoholism, National Institute of Health, Bethesda, MD, 20892, USA
- Center on Compulsive Behaviors, Intramural Research Program, National Institute of Health, Bethesda, MD, 20892, USA
| | - Dorit Ron
- Department of Neurology, University of California San Francisco (UCSF), San Francisco, CA, 94143, USA.
| |
Collapse
|
39
|
Putra M, Puttachary S, Liu G, Lee G, Thippeswamy T. Fyn-tau Ablation Modifies PTZ-Induced Seizures and Post-seizure Hallmarks of Early Epileptogenesis. Front Cell Neurosci 2020; 14:592374. [PMID: 33363455 PMCID: PMC7752812 DOI: 10.3389/fncel.2020.592374] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/16/2020] [Indexed: 12/22/2022] Open
Abstract
Both Fyn and tau have been associated with neuronal hyperexcitability and neurotoxicity in many tauopathies, including Alzheimer's disease (AD). Individual genetic ablation of fyn or tau appears to be protective against aberrant excitatory neuronal activities in AD and epilepsy models. It is, however, still unknown whether ablation of both Fyn and tau can likely elicit more profound anti-seizure and neuroprotective effects. Here, we show the effects of genetic deletion of Fyn and/or tau on seizure severity in response to pentylenetetrazole (PTZ)-induced seizure in mouse models and neurobiological changes 24 h post-seizures. We used Fyn KO (fyn -/-), tau KO (tau -/-), double knockout (DKO) (fyn -/- / tau -/-), and wild-type (WT) mice of the same genetic background. Both tau KO and DKO showed a significant increase in latency to convulsive seizures and significantly decreased the severity of seizures post-PTZ. Although Fyn KO did not differ significantly from WT, in response to PTZ, Fyn KO still had 36 ± 8% seizure reduction and a 30% increase in seizure latency compared to WT. Surprisingly, in contrast to WT, Fyn KO mice showed higher mortality in <20 min of seizure induction; these mice had severe hydrocephalous. None of the tau -/- and DKO died during the study. In response to PTZ, all KO groups showed a significant reduction in neurodegeneration and gliosis, in contrast to WT, which showed increased neurodegeneration [especially, parvalbumin (PV)-GABAergic interneurons] and gliosis. DKO mice had the most reduced gliosis. Immunohistochemically, phospho-tau (AT8, pS199/S202), Fyn expression, as well as Fyn-tau interaction as measured by PLA increased in WT post-PTZ. Moreover, hippocampal Western blots revealed increased levels of AT8, tyrosine phospho-tau (pY18), and phosphorylated Src tyrosine family kinases (pSFK) in PTZ-treated WT, but not in KO, compared to respective controls. Furthermore, PV interneurons were protected from PTZ-induced seizure effects in all KO mice. The levels of inwardly rectifying potassium (Kir 4.1) channels were also downregulated in astrocytes in the WT post-PTZ, while its levels did not change in KO groups. Overall, our results demonstrated the role of Fyn and tau in seizures and their impact on the mediators of early epileptogenesis in PTZ model.
Collapse
Affiliation(s)
- Marson Putra
- Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Sreekanth Puttachary
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Guanghao Liu
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Gloria Lee
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, United States
| | - Thimmasettappa Thippeswamy
- Neuroscience Interdepartmental Program, Iowa State University, Ames, IA, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
40
|
Levetiracetam Reduced the Basal Excitability of the Dentate Gyrus without Restoring Impaired Synaptic Plasticity in Rats with Temporal Lobe Epilepsy. Brain Sci 2020; 10:brainsci10090634. [PMID: 32933015 PMCID: PMC7565946 DOI: 10.3390/brainsci10090634] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 12/13/2022] Open
Abstract
Temporal lobe epilepsy (TLE), the most common type of focal epilepsy, affects learning and memory; these effects are thought to emerge from changes in synaptic plasticity. Levetiracetam (LEV) is a widely used antiepileptic drug that is also associated with the reversal of cognitive dysfunction. The long-lasting effect of LEV treatment and its participation in synaptic plasticity have not been explored in early chronic epilepsy. Therefore, through the measurement of evoked field potentials, this study aimed to comprehensively identify the alterations in the excitability and the short-term (depression/facilitation) and long-term synaptic plasticity (long-term potentiation, LTP) of the dentate gyrus of the hippocampus in a lithium–pilocarpine rat model of TLE, as well as their possible restoration by LEV (1 week; 300 mg/kg/day). TLE increased the population spike (PS) amplitude (input/output curve); interestingly, LEV treatment partially reduced this hyperexcitability. Furthermore, TLE augmented synaptic depression, suppressed paired-pulse facilitation, and reduced PS-LTP; however, LEV did not alleviate such alterations. Conversely, the excitatory postsynaptic potential (EPSP)-LTP of TLE rats was comparable to that of control rats and was decreased by LEV. LEV caused a long-lasting attenuation of basal hyperexcitability but did not restore impaired synaptic plasticity in the early chronic phase of TLE.
Collapse
|
41
|
CRL5-dependent regulation of the small GTPases ARL4C and ARF6 controls hippocampal morphogenesis. Proc Natl Acad Sci U S A 2020; 117:23073-23084. [PMID: 32873638 DOI: 10.1073/pnas.2002749117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The small GTPase ARL4C participates in the regulation of cell migration, cytoskeletal rearrangements, and vesicular trafficking in epithelial cells. The ARL4C signaling cascade starts by the recruitment of the ARF-GEF cytohesins to the plasma membrane, which, in turn, bind and activate the small GTPase ARF6. However, the role of ARL4C-cytohesin-ARF6 signaling during hippocampal development remains elusive. Here, we report that the E3 ubiquitin ligase Cullin 5/RBX2 (CRL5) controls the stability of ARL4C and its signaling effectors to regulate hippocampal morphogenesis. Both RBX2 knockout and Cullin 5 knockdown cause hippocampal pyramidal neuron mislocalization and development of multiple apical dendrites. We used quantitative mass spectrometry to show that ARL4C, Cytohesin-1/3, and ARF6 accumulate in the RBX2 mutant telencephalon. Furthermore, we show that depletion of ARL4C rescues the phenotypes caused by Cullin 5 knockdown, whereas depletion of CYTH1 or ARF6 exacerbates overmigration. Finally, we show that ARL4C, CYTH1, and ARF6 are necessary for the dendritic outgrowth of pyramidal neurons to the superficial strata of the hippocampus. Overall, we identified CRL5 as a key regulator of hippocampal development and uncovered ARL4C, CYTH1, and ARF6 as CRL5-regulated signaling effectors that control pyramidal neuron migration and dendritogenesis.
Collapse
|
42
|
Cizeron M, Qiu Z, Koniaris B, Gokhale R, Komiyama NH, Fransén E, Grant SGN. A brainwide atlas of synapses across the mouse life span. Science 2020; 369:270-275. [PMID: 32527927 PMCID: PMC7115813 DOI: 10.1126/science.aba3163] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 05/19/2020] [Indexed: 01/03/2023]
Abstract
Synapses connect neurons together to form the circuits of the brain, and their molecular composition controls innate and learned behavior. We analyzed the molecular and morphological diversity of 5 billion excitatory synapses at single-synapse resolution across the mouse brain from birth to old age. A continuum of changes alters synapse composition in all brain regions across the life span. Expansion in synapse diversity produces differentiation of brain regions until early adulthood, and compositional changes cause dedifferentiation in old age. The spatiotemporal synaptome architecture of the brain potentially accounts for life-span transitions in intellectual ability, memory, and susceptibility to behavioral disorders.
Collapse
Affiliation(s)
- Mélissa Cizeron
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Institut NeuroMyoGène, Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR-5310, INSERM U-1217, 69008 Lyon, France
| | - Zhen Qiu
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Babis Koniaris
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- School of Computing, Edinburgh Napier University, Edinburgh EH10 5DT, UK
| | - Ragini Gokhale
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
| | - Noboru H Komiyama
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Erik Fransén
- Department of Computational Science and Technology, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden
- Science for Life Laboratory, KTH Royal Institute of Technology, SE-171 65 Solna, Sweden
| | - Seth G N Grant
- Genes to Cognition Program, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh EH16 4SB, UK.
- Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh EH8 9XD, UK
| |
Collapse
|
43
|
Liu G, Fiock KL, Levites Y, Golde TE, Hefti MM, Lee G. Fyn depletion ameliorates tau P301L-induced neuropathology. Acta Neuropathol Commun 2020; 8:108. [PMID: 32665013 PMCID: PMC7362472 DOI: 10.1186/s40478-020-00979-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/24/2020] [Indexed: 02/06/2023] Open
Abstract
The Src family non-receptor tyrosine kinase Fyn has been implicated in neurodegeneration of Alzheimer's disease through interaction with amyloid β (Aβ). However, the role of Fyn in the pathogenesis of primary tauopathies such as FTDP-17, where Aβ plaques are absent, is poorly understood. In the current study, we used AAV2/8 vectors to deliver tauP301L to the brains of WT and Fyn KO mice, generating somatic transgenic tauopathy models with the presence or absence of Fyn. Although both genotypes developed tau pathology, Fyn KO developed fewer neurofibrillary tangles on Bielschowsky and Thioflavin S stained sections and showed lower levels of phosphorylated tau. In addition, tauP301L-induced behavior abnormalities and depletion of synaptic proteins were not observed in the Fyn KO model. Our work provides evidence for Fyn being a critical protein in the disease pathogenesis of FTDP-17.
Collapse
Affiliation(s)
- Guanghao Liu
- Interdisciplinary Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Kimberly L. Fiock
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Yona Levites
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL USA
| | - Todd E. Golde
- Department of Neuroscience, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL USA
| | - Marco M. Hefti
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA USA
| | - Gloria Lee
- Interdisciplinary Program in Neuroscience, University of Iowa Carver College of Medicine, Iowa City, IA USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, 500 Newton Road, ML B191, Iowa City, IA 52242 USA
| |
Collapse
|
44
|
Barakat AK, Scholl C, Steffens M, Brandenburg K, Ising M, Lucae S, Holsboer F, Laje G, Kalayda GV, Jaehde U, Stingl JC. Citalopram-induced pathways regulation and tentative treatment-outcome-predicting biomarkers in lymphoblastoid cell lines from depression patients. Transl Psychiatry 2020; 10:210. [PMID: 32612257 PMCID: PMC7329820 DOI: 10.1038/s41398-020-00900-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 06/08/2020] [Accepted: 06/16/2020] [Indexed: 12/17/2022] Open
Abstract
Antidepressant therapy is still associated with delays in symptomatic improvement and low response rates. Incomplete understanding of molecular mechanisms underlying antidepressant effects hampered the identification of objective biomarkers for antidepressant response. In this work, we studied transcriptome-wide expression followed by pathway analysis in lymphoblastoid cell lines (LCLs) derived from 17 patients documented for response to SSRI antidepressants from the Munich Antidepressant Response Signatures (MARS) study upon short-term incubation (24 and 48 h) with citalopram. Candidate transcripts were further validated with qPCR in MARS LCLs from responders (n = 33) vs. non-responders (n = 36) and afterward in an independent cohort of treatment-resistant patients (n = 20) vs. first-line responders (n = 24) from the STAR*D study. In MARS cohort we observed significant associations of GAD1 (glutamate decarboxylase 1; p = 0.045), TBC1D9 (TBC1 Domain Family Member 9; p = 0.014-0.021) and NFIB (nuclear factor I B; p = 0.015-0.025) expression with response status, remission status and improvement in depression scale, respectively. Pathway analysis of citalopram-altered gene expression indicated response-status-dependent transcriptional reactions. Whereas in clinical responders neural function pathways were primarily up- or downregulated after incubation with citalopram, deregulated pathways in non-responders LCLs mainly involved cell adhesion and immune response. Results from the STAR*D study showed a marginal association of treatment-resistant depression with NFIB (p = 0.068) but not with GAD1 (p = 0.23) and TBC1D9 (p = 0.27). Our results propose the existence of distinct pathway regulation mechanisms in responders vs. non-responders and suggest GAD1, TBC1D9, and NFIB as tentative predictors for clinical response, full remission, and improvement in depression scale, respectively, with only a weak overlap in predictors of different therapy outcome phenotypes.
Collapse
Affiliation(s)
- Abdul Karim Barakat
- grid.414802.b0000 0000 9599 0422Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany ,grid.10388.320000 0001 2240 3300Department of Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Catharina Scholl
- grid.414802.b0000 0000 9599 0422Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | - Michael Steffens
- grid.414802.b0000 0000 9599 0422Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | - Kerstin Brandenburg
- grid.414802.b0000 0000 9599 0422Federal Institute for Drugs and Medical Devices (BfArM), Bonn, Germany
| | - Marcus Ising
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, Munich, Germany
| | - Susanne Lucae
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, Munich, Germany
| | - Florian Holsboer
- grid.419548.50000 0000 9497 5095Max Planck Institute of Psychiatry, Munich, Germany
| | - Gonzalo Laje
- Washington Behavioral Medicine Associates LLC, Chevy Chase, MD USA
| | - Ganna V. Kalayda
- grid.10388.320000 0001 2240 3300Department of Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Ulrich Jaehde
- grid.10388.320000 0001 2240 3300Department of Clinical Pharmacy, University of Bonn, Bonn, Germany
| | - Julia Carolin Stingl
- Institute of Clinical Pharmacology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
45
|
Fyn Tyrosine Kinase as Harmonizing Factor in Neuronal Functions and Dysfunctions. Int J Mol Sci 2020; 21:ijms21124444. [PMID: 32580508 PMCID: PMC7352836 DOI: 10.3390/ijms21124444] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/25/2022] Open
Abstract
Fyn is a non-receptor or cytoplasmatic tyrosine kinase (TK) belonging to the Src family kinases (SFKs) involved in multiple transduction pathways in the central nervous system (CNS) including synaptic transmission, myelination, axon guidance, and oligodendrocyte formation. Almost one hundred years after the original description of Fyn, this protein continues to attract extreme interest because of its multiplicity of actions in the molecular signaling pathways underlying neurodevelopmental as well as neuropathologic events. This review highlights and summarizes the most relevant recent findings pertinent to the role that Fyn exerts in the brain, emphasizing aspects related to neurodevelopment and synaptic plasticity. Fyn is a common factor in healthy and diseased brains that targets different proteins and shapes different transduction signals according to the neurological conditions. We will primarily focus on Fyn-mediated signaling pathways involved in neuronal differentiation and plasticity that have been subjected to considerable attention lately, opening the fascinating scenario to target Fyn TK for the development of potential therapeutic interventions for the treatment of CNS injuries and certain neurodegenerative disorders like Alzheimer’s disease.
Collapse
|
46
|
Transcriptomic Analysis Reveals Abnormal Expression of Prion Disease Gene Pathway in Brains from Patients with Autism Spectrum Disorders. Brain Sci 2020; 10:brainsci10040200. [PMID: 32235346 PMCID: PMC7226514 DOI: 10.3390/brainsci10040200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/22/2022] Open
Abstract
The role of infections in the pathogenesis of autism spectrum disorder (ASD) is still controversial. In this study, we aimed to evaluate markers of infections and immune activation in ASD by performing a meta-analysis of publicly available whole-genome transcriptomic datasets of brain samples from autistic patients and otherwise normal people. Among the differentially expressed genes, no significant enrichment was observed for infectious diseases previously associated with ASD, including herpes simplex virus-1 (HSV-1), cytomegalovirus and Epstein–Barr virus in brain samples, nor was it found in peripheral blood from ASD patients. Interestingly, a significant number of genes belonging to the “prion diseases” pathway were found to be modulated in our ASD brain meta-analysis. Overall, our data do not support an association between infection and ASD. However, the data do provide support for the involvement of pathways related to other neurodegenerative diseases and give input to uncover novel pathogenetic mechanisms underlying ASD.
Collapse
|
47
|
Glukhova XA, Trizna JA, Proussakova OV, Gogvadze VG, Beletsky IP. Dephosphorylation of Fas-ligand and caveolin-1 is a prerequisite step in Fas-ligand - caveolin-1 complex formation and cell death stimulation. Cell Signal 2020; 70:109590. [PMID: 32109550 DOI: 10.1016/j.cellsig.2020.109590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Revised: 02/11/2020] [Accepted: 02/25/2020] [Indexed: 10/24/2022]
Abstract
Fas-ligand/CD178 belongs to the TNF family proteins and is the well-characterized inducer of cell death. We showed previously that the interaction of Fas-ligand and caveolin-1 is necessary for Fas-ligand translocation to rafts, and the subsequent induction of Fas-ligand-dependent cell death. Both molecules can undergo phosphorylation, however the role of the phosphorylation state of Fas-ligand and caveolin-1 in their physical association, and consequently in of Fas - mediated cell death induction is currently unknown. In this study, we show that in control cells Fas-ligand interaction with caveolin-1 is not observed, and both molecules are phosphorylated. The intracellular part of Fas-ligand was shown to form a complex with p59Fyn-kinase. Upon cell death activation, the expression and activity of p59Fyn-kinase decreases substantially, leading to the disruption of Fas-ligand - p59Fyn-kinase association, dephosphorylation of Fas-ligand and caveolin-1, and formation of a complex between them (Fas-ligand - caveolin-1). The analysis of the effects of kinase and phosphatase inhibitors revealed that phosphorylation of Fas-ligand and caveolin-1 at tyrosine residues suppressed Fas-mediated cell death. Thus, dephosphorylation of Fas-ligand and caveolin-1 is critical for triggering Fas-ligand-mediated apoptotic pathway and cell death execution.
Collapse
Affiliation(s)
- Xenia A Glukhova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Institutskaya st., 3, 142290, Russia
| | - Julia A Trizna
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Institutskaya st., 3, 142290, Russia
| | - Olga V Proussakova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Institutskaya st., 3, 142290, Russia
| | - Vladimir G Gogvadze
- Faculty of Fundamental Medicine, MV Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Igor P Beletsky
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Puschino, Institutskaya st., 3, 142290, Russia.
| |
Collapse
|
48
|
Bland T, Zhu M, Dillon C, Sahin GS, Rodriguez-Llamas JL, Appleyard SM, Wayman GA. Leptin Controls Glutamatergic Synaptogenesis and NMDA-Receptor Trafficking via Fyn Kinase Regulation of NR2B. Endocrinology 2020; 161:5678106. [PMID: 31840160 PMCID: PMC7015580 DOI: 10.1210/endocr/bqz030] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 12/10/2019] [Indexed: 01/13/2023]
Abstract
Activation of the leptin receptor, LepRb, by the adipocytokine/neurotrophic factor leptin in the central nervous system has procognitive and antidepressive effects. Leptin has been shown to increase glutamatergic synaptogenesis in multiple brain regions. In contrast, mice that have a mutation in the LepRb gene show abnormal synapse development in the hippocampus as well as deficits in cognition and increased depressive-like symptoms. Leptin increases glutamatergic synaptogenesis, in part, through enhancement of N-methyl-D-aspartic acid (NMDA) receptor function; yet the underlying signaling pathway is not known. In this study, we examine how leptin regulates surface expression of NR2B-containing NMDA receptors in hippocampal neurons. Leptin stimulation increases NR2BY1472 phosphorylation, which is inhibited by the Src family kinase inhibitor, PP1. Moreover, we show that Fyn, a member of the Src family kinases, is required for leptin-stimulated NR2BY1472 phosphorylation. Furthermore, inhibiting Y1472 phosphorylation with either a dominant negative Fyn mutant or an NR2B mutant that lacks the phosphorylation site (NR2BY1472F) blocks leptin-stimulated synaptogenesis. Additionally, we show that LepRb forms a complex with NR2B and Fyn. Taken together, these findings expand our knowledge of the LepRb interactome and the mechanisms by which leptin stimulates glutamatergic synaptogenesis in the developing hippocampus. Comprehending these mechanisms is key for understanding dendritic spine development and synaptogenesis, alterations of which are associated with many neurological disorders.
Collapse
Affiliation(s)
- Tyler Bland
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Mingyan Zhu
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Crystal Dillon
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Gulcan Semra Sahin
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Jose Luis Rodriguez-Llamas
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Suzanne M Appleyard
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
| | - Gary A Wayman
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, Washington
- Correspondence: Gary A. Wayman, Department of Integrative Physiology and Neuroscience, Program in Neuroscience, Washington State University, Pullman Washington 99164. E-mail:
| |
Collapse
|
49
|
Brown AS, Meera P, Quinones G, Magri J, Otis TS, Pulst SM, Oro AE. Receptor protein tyrosine phosphatases control Purkinje neuron firing. Cell Cycle 2020; 19:153-159. [PMID: 31876231 PMCID: PMC6961678 DOI: 10.1080/15384101.2019.1695995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 10/23/2019] [Accepted: 11/07/2019] [Indexed: 10/25/2022] Open
Abstract
Spinocerebellar ataxias (SCA) are a genetically heterogeneous family of cerebellar neurodegenerative diseases characterized by abnormal firing of Purkinje neurons and degeneration. We recently demonstrated the slowed firing rates seen in several SCAs share a common etiology of hyper-activation of the Src family of non-receptor tyrosine kinases (SFKs). However, the lack of clinically available neuroactive SFK inhibitors lead us to investigate alternative mechanisms to modulate SFK activity. Previous studies demonstrate that SFK activity can be enhanced by the removal of inhibitory phospho-marks by receptor-protein-tyrosine phosphatases (RPTPs). In this Extra View we show that MTSS1 inhibits SFK activity through the binding and inhibition of a subset of the RPTP family members, and lowering RPTP activity in cerebellar slices with peptide inhibitors increases the suppressed Purkinje neuron basal firing rates seen in two different SCA models. Together these results identify RPTPs as novel effectors of Purkinje neuron basal firing, extending the MTSS1/SFK regulatory circuit we previously described and expanding the therapeutic targets for SCA patients.
Collapse
Affiliation(s)
- Alexander S. Brown
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Pratap Meera
- Department of Neurobiology, University of California, Los Angeles, CA, USA
| | - Gabe Quinones
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jessica Magri
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas S. Otis
- Sainsbury Wellcome Centre for Neural Circuits and Behavior, University College London, London, UK
| | - Stefan M. Pulst
- Department of Neurology, University of Utah Medical Center, Salt Lake City, UT, USA
| | - Anthony E. Oro
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
50
|
Liu AM, Chen BL, Yu LT, Liu T, Shi LL, Yu PP, Qu YB, So KF, Zhou LB. Human adipose tissue- and umbilical cord-derived stem cells: which is a better alternative to treat spinal cord injury? Neural Regen Res 2020; 15:2306-2317. [PMID: 32594054 PMCID: PMC7749492 DOI: 10.4103/1673-5374.284997] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Multiple types of stem cells have been proposed for the treatment of spinal cord injury, but their comparative information remains elusive. In this study, a rat model of T10 contusion spinal cord injury was established by the impactor method. Human umbilical cord-derived mesenchymal stem cells (UCMSCs) or human adipose tissue-derived mesenchymal stem cells (ADMSCs) (2.5 μL/injection site, 1 × 105 cells/μL) was injected on rostral and caudal of the injury segment on the ninth day after injury. Rats injected with mesenchymal stem cell culture medium were used as controls. Our results show that although transplanted UCMSCs and ADMSCs failed to differentiate into neurons or glial cells in vivo, both significantly improved motor and sensory function. After spinal cord injury, UCMSCs and ADMSCs similarly promoted spinal neuron survival and axonal regeneration, decreased glial scar and lesion cavity formation, and reduced numbers of active macrophages. Bio-Plex analysis of spinal samples showed a specific increase of interleukin-10 and decrease of tumor necrosis factor α in the ADMSC group, as well as a downregulation of macrophage inflammatory protein 3α in both UCMSC and ADMSC groups at 3 days after cell transplantation. Upregulation of interleukin-10 and interleukin-13 was observed in both UCMSC and ADMSC groups at 7 days after cell transplantation. Isobaric tagging for relative and absolute quantitation proteomics analyses showed that UCMSCs and ADMSCs induced changes of multiple genes related to axonal regeneration, neurotrophy, and cell apoptosis in common and specific manners. In conclusion, UCMSC and ADMSC transplants yielded quite similar contributions to motor and sensory recovery after spinal cord injury via anti-inflammation and improved axonal growth. However, there were some differences in cytokine and gene expression induced by these two types of transplanted cells. Animal experiments were approved by the Laboratory Animal Ethics Committee at Jinan University (approval No. 20180228026) on February 28, 2018, and the application of human stem cells was approved by the Medical Ethics Committee of Medical College of Jinan University of China (approval No. 2016041303) on April 13, 2016.
Collapse
Affiliation(s)
- Ai-Mei Liu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Bo-Li Chen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Ling-Tai Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Tao Liu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Ling-Ling Shi
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Pan-Pan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Yi-Bo Qu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Li-Bing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|