1
|
Bakhtazad A, Kabbaj M, Garmabi B, Joghataei MT. The role of CART peptide in learning and memory: A potential therapeutic target in memory-related disorders. Peptides 2024; 181:171298. [PMID: 39317295 DOI: 10.1016/j.peptides.2024.171298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/19/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
Cocaine and amphetamine-regulated transcript (CART) mRNA and peptide are vastly expressed in both cortical and subcortical brain areas and are involved in critical cognitive functions. CART peptide (CARTp), described in reward-related brain structures, regulates drug-induced learning and memory, and its role appears specific to psychostimulants. However, many other drugs of abuse, such as alcohol, opiates, nicotine, and caffeine, have been shown to alter the expression levels of CART mRNA and peptides in brain structures directly or indirectly associated with learning and memory processes. However, the number of studies demonstrating the contribution of CARTp in learning and memory is still minimal. Notably, the exact cellular and molecular mechanisms underlying CARTp effects are still unknown. The discoveries that CARTp effects are mediated through a putative G-protein coupled receptor and activation of cellular signaling cascades via NMDA receptor-coupled ERK have enhanced our knowledge about the action of this neuropeptide and allowed us to comprehend better CARTp exact cellular/molecular mechanisms that could mediate drug-induced changes in learning and memory functions. Unfortunately, these efforts have been impeded by the lack of suitable and specific CARTp receptor antagonists. In this review, following a short introduction about CARTp, we report on current knowledge about CART's roles in learning and memory processes and its recently described role in memory-related neurological disorders. We will also discuss the importance of further investigating how CARTp interacts with its receptor(s) and other neurotransmitter systems to influence learning and memory functions. This topic is sure to intrigue and motivate further exploration in the field of neuroscience.
Collapse
Affiliation(s)
- Atefeh Bakhtazad
- Cellular and Molecular Research Center, Deputy of Research and Technology, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohamed Kabbaj
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306-1270, United States; Program of Neuroscience, Florida State University, Tallahassee, FL 32306-1270, United States
| | - Behzad Garmabi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Deputy of Research and Technology, Iran University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Selås M. Language profiles in Noonan Syndrome - A multiple case study. CLINICAL LINGUISTICS & PHONETICS 2024; 38:1025-1039. [PMID: 38226429 DOI: 10.1080/02699206.2023.2298976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/11/2023] [Accepted: 12/19/2023] [Indexed: 01/17/2024]
Abstract
Noonan Syndrome (NS) is a genetic disorder associated with a diverse range of symptoms. This study aims to provide a comprehensive description of the linguistic profiles of children and adolescents with NS, focusing on vocabulary, grammar skills, phonological memory skills, working memory skills, and visuospatial skills. Sixteen participants aged 6-16 took part in the study. The findings reveal substantial variation in the affected linguistic areas, with some participants demonstrated normal findings, while inconsistent and overall weak language skills were observed in a large subgroup of participants.
Collapse
Affiliation(s)
- Magnhild Selås
- Department of Nordic and Media studies, University of Agder, Kristiansand, Norway
| |
Collapse
|
3
|
Walters JM, Noblet HA, Chung HJ. An emerging role of STriatal-Enriched protein tyrosine Phosphatase in hyperexcitability-associated brain disorders. Neurobiol Dis 2024; 200:106641. [PMID: 39159894 DOI: 10.1016/j.nbd.2024.106641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024] Open
Abstract
STriatal-Enriched protein tyrosine Phosphatase (STEP) is a brain-specific tyrosine phosphatase that is associated with numerous neurological and neuropsychiatric disorders. STEP dephosphorylates and inactivates various kinases and phosphatases critical for neuronal function and health including Fyn, Pyk2, ERK1/2, p38, and PTPα. Importantly, STEP dephosphorylates NMDA and AMPA receptors, two major glutamate receptors that mediate fast excitatory synaptic transmission. This STEP-mediated dephosphorylation leads to their internalization and inhibits both Hebbian synaptic potentiation and homeostatic synaptic scaling. Hence, STEP has been widely accepted to weaken excitatory synaptic strength. However, emerging evidence implicates a novel role of STEP in neuronal hyperexcitability and seizure disorders. Genetic deletion and pharmacological blockade of STEP reduces seizure susceptibility in acute seizure mouse models and audiogenic seizures in a mouse model of Fragile X syndrome. Pharmacologic inhibition of STEP also decreases hippocampal activity and neuronal intrinsic excitability. Here, we will highlight the divergent roles of STEP in excitatory synaptic transmission and neuronal intrinsic excitability, present the potential underlying mechanisms, and discuss their impact on STEP-associated neurologic and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jennifer M Walters
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hayden A Noblet
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hee Jung Chung
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Dept. of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Institute of Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
4
|
Montiel I, Bello-Medina PC, Prado-Alcalá RA, Quirarte GL, Verdín-Ruvalcaba LA, Marín-Juárez TA, Medina AC. Involvement of kinases in memory consolidation of inhibitory avoidance training. Rev Neurosci 2024:revneuro-2024-0093. [PMID: 39323086 DOI: 10.1515/revneuro-2024-0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/08/2024] [Indexed: 09/27/2024]
Abstract
The inhibitory avoidance (IA) task is a paradigm widely used to investigate the molecular and cellular mechanisms involved in the formation of long-term memory of aversive experiences. In this review, we discuss studies on different brain structures in rats associated with memory consolidation, such as the hippocampus, striatum, and amygdala, as well as some cortical areas, including the insular, cingulate, entorhinal, parietal and prefrontal cortex. These studies have shown that IA training triggers the release of neurotransmitters, hormones, growth factors, etc., that activate intracellular signaling pathways related to protein kinases, which induce intracellular non-genomic changes or transcriptional mechanisms in the nucleus, leading to the synthesis of proteins. We have summarized the temporal dynamics and crosstalk among protein kinase A, protein kinase C, mitogen activated protein kinase, extracellular-signal-regulated kinase, and Ca2+/calmodulin-dependent protein kinase II described in the hippocampus. Protein kinase activity has been associated with structural changes and synaptic strengthening, resulting in memory storage. However, little is known about the molecular mechanisms involved in intense IA training, which protects memory from typical amnestic treatments, such as protein synthesis inhibitors, and induces increased spinogenesis, suggesting an unexplored mechanism independent of the genomic pathway. This highly emotional experience causes an extinction-resistant memory, as has been observed in some pathological states such as post-traumatic stress disorder. We propose that the changes in spinogenesis observed after intense IA training could be generated by protein kinases via non-genomic pathways.
Collapse
Affiliation(s)
- Ivan Montiel
- Institut Pasteur, Université Paris Cité, Neural Circuits for Spatial Navigation and Memory, Department of Neuroscience, F-75015, Paris, France
- Sorbonne Université, Collège Doctoral, F-75005, Paris, France
| | - Paola C Bello-Medina
- Facultad de Ciencias, Universidad del Tolima, Altos de Santa Helena, Ibagué, Tolima, Colombia
| | - Roberto A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Gina L Quirarte
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Luis A Verdín-Ruvalcaba
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Tzitzi A Marín-Juárez
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Andrea C Medina
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| |
Collapse
|
5
|
Moseley-Alldredge M, Aragón C, Vargus M, Alley D, Somia N, Chen L. The L1CAM SAX-7 is an antagonistic modulator of Erk Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.14.613091. [PMID: 39345534 PMCID: PMC11429911 DOI: 10.1101/2024.09.14.613091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
L1CAMs are immunoglobulin superfamily cell adhesion molecules that ensure proper nervous system development and function. In addition to being associated with the autism and schizophrenia spectrum disorders, mutations in the L1CAM family of genes also underlie distinct developmental syndromes with neurological conditions, such as intellectual disability, spastic paraplegia, hypotonia and congenital hydrocephalus. Studies in both vertebrate and invertebrate model organisms have established conserved neurodevelopmental roles for L1CAMs; these include axon guidance, dendrite morphogenesis, synaptogenesis, and maintenance of neural architecture, among others. In Caenorhabditis elegans , L1CAMs, encoded by the sax-7 gene, are required for coordinated locomotion. We previously uncovered a genetic interaction between sax-7 and components of synaptic vesicle cycle, revealing a non-developmental role for sax-7 in regulating synaptic activity. More recently, we determined that sax-7 also genetically interacts with extracellular signal-related kinase (ERK) signaling in controlling coordinated locomotion. C. elegans ERK, encoded by the mpk-1 gene, is a serine/threonine protein kinase belonging to the mitogen-activated protein kinase (MAPK) family that governs multiple aspects of animal development and cellular homeostasis. Here, we show this genetic interaction between sax-7 and mpk-1 occurs not only in cholinergic neurons for coordinated locomotion, but also extends outside the nervous system, revealing novel roles for SAX-7/L1CAM in non-neuronal processes, including vulval development. Our genetic findings in both the nervous system and developing vulva are consistent with SAX-7/L1CAM acting as an antagonistic modulator of ERK signaling.
Collapse
|
6
|
Zhang H, Goedegebuure SP, Ding L, DeNardo D, Fields RC, Chen Y, Payne P, Li F. M3NetFlow: A novel multi-scale multi-hop graph AI model for integrative multi-omic data analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.15.545130. [PMID: 39282437 PMCID: PMC11398409 DOI: 10.1101/2023.06.15.545130] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Multi-omic data-driven studies, characterizing complex disease signaling system from multiple levels, are at the forefront of precision medicine and healthcare. The integration and interpretation of multi-omic data are essential for identifying molecular targets and deciphering core signaling pathways of complex diseases. However, it remains an open problem due the large number of biomarkers and complex interactions among them. In this study, we propose a novel Multi-scale Multi-hop Multi-omic graph model, M3NetFlow, to facilitate generic multi-omic data analysis to rank targets and infer core signaling flows/pathways. To evaluate M3NetFlow, we applied it in two independent multi-omic case studies: 1) uncovering mechanisms of synergistic drug combination response (defined as anchor-target guided learning), and 2) identifying biomarkers and pathways of Alzheimer 's disease (AD). The evaluation and comparison results showed M3NetFlow achieves the best prediction accuracy (accurate), and identifies a set of essential targets and core signaling pathways (interpretable). The model can be directly applied to other multi-omic data-driven studies. The code is publicly accessible at: https://github.com/FuhaiLiAiLab/M3NetFlow.
Collapse
Affiliation(s)
- Heming Zhang
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA
| | - S. Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Li Ding
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - David DeNardo
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ryan C. Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yixin Chen
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Philip Payne
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA
| | - Fuhai Li
- Institute for Informatics, Data Science and Biostatistics (I2DB), Washington University School of Medicine, St. Louis, MO, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
7
|
Xie S, Zhu M, Zhu H, Wang W. Effects of β-asarone on spatial learning and memory impairment by exhaustive exercise-induced fatigue: Role of NR-CaMKII-ERK/CREB signal in hippocampus of rats. Behav Brain Res 2024; 471:115076. [PMID: 38825021 DOI: 10.1016/j.bbr.2024.115076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/04/2024]
Abstract
OBJECTIVE It is to investigate the effects of β-asarone on learning and memory, hippocampal morphology, synaptophysin (SYP) and postsynaptic density 95(PSD95) protein expression, N-methyl-D-aspartic acid receptor 2B (NR2B)- Ca2+/calmodulin (CaM)-dependent protein kinase II (CaMKII) - Extracellular signal-regulated kinase (ERK) / Cyclic-AMP response element binding protein (CREB) signal in hippocampus of rats with exhaustive exercise-induced fatigue. METHODS Fifty Sprague-Dawley male rats were randomly divided into five groups: normal group, exercise group, exercise and β-asarone (2.5, 10, 40 mg/kg)-treated groups. The learning and memory in rats were tested by Morris water maze experiment. We measured the hippocampal morphology by Nissl staining. The levels of SYP, PSD95, NR2B, CaMKII, ERK1/2, CREB, p-NR2B, p-CaMKII, p-ERK1/2 and p-CREB expression were measured by western blot analysis. RESULTS The results demonstrated that β-asarone (10, 40 mg/kg) treatment significantly decreased the latency to find the platform, increased the time spent in the target quadrant and the number of crossing the platform of rats with exhaustive exercise-induced fatigue. β-asarone (10, 40 mg/kg) treatment increased the cell density in the hippocampus CA1 region, significantly up-regulated NR2B-CaMKII-ERK/CREB signal and improved the protein expression levels of SYP and PSD95 in hippocampus of rats with exhaustive exercise-induced fatigue. CONCLUSIONS It suggests that β-asarone could improve learning and memory of rats with exhaustive exercise-induced fatigue. The mechanism might be related to β-asarone protecting the morphology of hippocampus, increasing the protein expression levels of SYP and PSD95 and up-regulating NR2B-CaMKII-ERK/CREB signal in hippocampus of rats with exhaustive exercise-induced fatigue.
Collapse
Affiliation(s)
- Shifeng Xie
- School of Sports Science, Jinggangshan University, Jian, Jiangxi 343009, China
| | - Meiju Zhu
- School of Sports Science, Jinggangshan University, Jian, Jiangxi 343009, China.
| | - Hongzhu Zhu
- School of Sports Science, Jinggangshan University, Jian, Jiangxi 343009, China
| | - Wei Wang
- School of Sports Science, Jinggangshan University, Jian, Jiangxi 343009, China
| |
Collapse
|
8
|
Ribeiro FC, Cozachenco D, Argyrousi EK, Staniszewski A, Wiebe S, Calixtro JD, Soares‐Neto R, Al‐Chami A, Sayegh FE, Bermudez S, Arsenault E, Cossenza M, Lacaille J, Nader K, Sun H, De Felice FG, Lourenco MV, Arancio O, Aguilar‐Valles A, Sonenberg N, Ferreira ST. The ketamine metabolite (2R,6R)-hydroxynorketamine rescues hippocampal mRNA translation, synaptic plasticity and memory in mouse models of Alzheimer's disease. Alzheimers Dement 2024; 20:5398-5410. [PMID: 38934107 PMCID: PMC11350050 DOI: 10.1002/alz.14034] [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: 12/02/2023] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Impaired brain protein synthesis, synaptic plasticity, and memory are major hallmarks of Alzheimer's disease (AD). The ketamine metabolite (2R,6R)-hydroxynorketamine (HNK) has been shown to modulate protein synthesis, but its effects on memory in AD models remain elusive. METHODS We investigated the effects of HNK on hippocampal protein synthesis, long-term potentiation (LTP), and memory in AD mouse models. RESULTS HNK activated extracellular signal-regulated kinase 1/2 (ERK1/2), mechanistic target of rapamycin (mTOR), and p70S6 kinase 1 (S6K1)/ribosomal protein S6 signaling pathways. Treatment with HNK rescued hippocampal LTP and memory deficits in amyloid-β oligomers (AβO)-infused mice in an ERK1/2-dependent manner. Treatment with HNK further corrected aberrant transcription, LTP and memory in aged APP/PS1 mice. DISCUSSION Our findings demonstrate that HNK induces signaling and transcriptional responses that correct synaptic and memory deficits in AD mice. These results raise the prospect that HNK could serve as a therapeutic approach in AD. HIGHLIGHTS The ketamine metabolite HNK activates hippocampal ERK/mTOR/S6 signaling pathways. HNK corrects hippocampal synaptic and memory defects in two mouse models of AD. Rescue of synaptic and memory impairments by HNK depends on ERK signaling. HNK corrects aberrant transcriptional signatures in APP/PS1 mice.
Collapse
Affiliation(s)
- Felipe C. Ribeiro
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Danielle Cozachenco
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Elentina K. Argyrousi
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Agnieszka Staniszewski
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | - Shane Wiebe
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Joao D. Calixtro
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Rubens Soares‐Neto
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Aycheh Al‐Chami
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Fatema El Sayegh
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Sara Bermudez
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Emily Arsenault
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Marcelo Cossenza
- Department of Physiology and Pharmacology, Fluminense Federal UniversityBiomedical InstituteNiteróiRio de JaneiroBrazil
| | - Jean‐Claude Lacaille
- Department of Neurosciences, Université de MontréalCentre for Interdisciplinary Research on Brain and Learning and Research Group on Neural Signaling and CircuitsMontrealQuebecCanada
| | - Karim Nader
- Department of PsychologyMcGill UniversityMontrealQuebecCanada
| | - Hongyu Sun
- Department of NeuroscienceCarleton UniversityOttawaOntarioCanada
| | - Fernanda G. De Felice
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
- Department of Biomedical and Molecular Sciences, Centre for Neuroscience StudiesQueen's UniversityKingstonOntarioCanada
- Department of PsychiatryQueen's UniversityKingstonOntarioCanada
- D'Or Institute for Research and EducationRio de JaneiroRio de JaneiroBrazil
| | - Mychael V. Lourenco
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
| | | | - Nahum Sonenberg
- Department of BiochemistryMcGill UniversityMontrealQuebecCanada
| | - Sergio T. Ferreira
- Institute of Medical Biochemistry Leopoldo de MeisFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
- D'Or Institute for Research and EducationRio de JaneiroRio de JaneiroBrazil
- Institute of Biophysics Carlos Chagas FilhoFederal University of Rio de JaneiroRio de JaneiroRio de JaneiroBrazil
| |
Collapse
|
9
|
Latif‐Hernandez A, Yang T, Butler RR, Losada PM, Minhas PS, White H, Tran KC, Liu H, Simmons DA, Langness V, Andreasson KI, Wyss‐Coray T, Longo FM. A TrkB and TrkC partial agonist restores deficits in synaptic function and promotes activity-dependent synaptic and microglial transcriptomic changes in a late-stage Alzheimer's mouse model. Alzheimers Dement 2024; 20:4434-4460. [PMID: 38779814 PMCID: PMC11247716 DOI: 10.1002/alz.13857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 05/25/2024]
Abstract
INTRODUCTION Tropomyosin related kinase B (TrkB) and C (TrkC) receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid beta (Aβ) toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. METHODS PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APPL/S) and wild-type controls. Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA sequencing. RESULTS In APPL/S mice, BD10-2 treatment improved memory and LTP deficits. This was accompanied by normalized phosphorylation of protein kinase B (Akt), calcium-calmodulin-dependent kinase II (CaMKII), and AMPA-type glutamate receptors containing the subunit GluA1; enhanced activity-dependent recruitment of synaptic proteins; and increased excitatory synapse number. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. DISCUSSION BD10-2 prevented APPL/S/Aβ-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response. HIGHLIGHTS Small molecule modulation of tropomyosin related kinase B (TrkB) and C (TrkC) restores long-term potentiation (LTP) and behavior in an Alzheimer's disease (AD) model. Modulation of TrkB and TrkC regulates synaptic activity-dependent transcription. TrkB and TrkC receptors are candidate targets for translational therapeutics. Electrophysiology combined with transcriptomics elucidates synaptic restoration. LTP identifies neuron and microglia AD-relevant human-mouse co-expression modules.
Collapse
Affiliation(s)
- Amira Latif‐Hernandez
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Tao Yang
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Robert R. Butler
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Patricia Moran Losada
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
| | - Paras S. Minhas
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Halle White
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Kevin C. Tran
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Harry Liu
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Danielle A. Simmons
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Vanessa Langness
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
| | - Katrin I. Andreasson
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
- Chan Zuckerberg BiohubSan FranciscoCaliforniaUSA
| | - Tony Wyss‐Coray
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
- The Phil and Penny Knight Initiative for Brain ResilienceStanford UniversityStanfordCaliforniaUSA
| | - Frank M. Longo
- Department of Neurology & Neurological SciencesStanford University School of MedicinePalo AltoCaliforniaUSA
- Wu Tsai Neurosciences Institute, Stanford UniversityStanfordCaliforniaUSA
| |
Collapse
|
10
|
Tan L, Zhang H, Li H, Sun S, Lyu Q, Jiang Y. Blueberry extracts antagonize Aβ 25-35 neurotoxicity and exert a neuroprotective effect through MEK-ERK-BDNF/UCH-L1 signaling pathway in rat and mouse hippocampus. Nutr Neurosci 2024; 27:745-760. [PMID: 37647279 DOI: 10.1080/1028415x.2023.2252640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
BACKGROUND The neuroprotective potential of blueberry (BB) extracts against Alzheimer's disease (AD) has been previously hinted at, while its exact mechanism has remained largely enigmatic. OBJECTIVE Our study endeavored to unravel the impacts and mechanisms by which BB extracts ameliorated the learning and memory prowess of AD-afflicted mice, with a specific focus on the MEK-ERK pathway. METHODS We employed 3-month-old APP/PS1 transgenic mice and stratified them into three distinct groups: AD+BB, AD, and control (CT). The Morris Water Maze Test (MWMT) was then administered to gauge their learning and memory faculties. In vitro experiments were executed on Aβ25-35-afflicted rat hippocampal neurons, which were subsequently treated with varying concentrations of BB extracts. We then assessed the expression levels of genes and proteins integral to the MEK-ERKBDNF/UCH-L1 pathway. RESULTS The data showed that the AD mice demonstrated compromised learning and memory faculties in MWMT. However, the AD+BB cohort showcased marked improvements in performance. Furthermore, in the AD subset, significant elevations in the expressions of MEK2 and ERK1/2 were observed, both at the mRNA and protein levels. Conversely, UCH-L1 mRNA expressions exhibited a decline, while BDNF expressions surged significantly. However, post BB extract treatment, the expressions of MEK2 and ERK1/2 were subdued, with UCH-L1 and BDNF mRNA expressions reverting to control levels. CONCLUSIONS Our findings propounded that BB extracts could offer therapeutic promise for AD by bolstering learning and memory capacities. The unwarranted activation of the MEK-ERK pathway, coupled with the aberrant expressions of BDNF and UCH-L1, might underpin AD's pathogenesis.
Collapse
Affiliation(s)
- Long Tan
- Department of Nutrition and Food Hygiene, School of Public Health, Tianjin Medical University, Tianjin, People's Republic of China
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
| | - Han Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
| | - Haiqiang Li
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
- Yantai Economic and Technological Development Area Hospital, Yantai Economic and Technological Development Area, Yantai, People's Republic of China
| | - Shoudan Sun
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
- Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Quanjun Lyu
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, People's Republic of China
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yugang Jiang
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, People's Republic of China
| |
Collapse
|
11
|
Cheng KM, Hsu WL, Ma YL, Liu YC, Lee EHY. Novel role of NCoR1 in impairing spatial memory through the mediation of a novel interacting protein DEC2. Cell Mol Life Sci 2024; 81:273. [PMID: 38900294 PMCID: PMC11335199 DOI: 10.1007/s00018-024-05321-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/06/2024] [Accepted: 06/13/2024] [Indexed: 06/21/2024]
Abstract
Long-term memory formation requires de novo RNA and protein synthesis. Using differential display PCR, we found that the NCoR1 cDNA fragment is differentially expressed between fast learners and slow learners, with fast learners showing a lower expression level than slow learners in the water maze learning task. Fast learners also show lower NCoR1 mRNA and protein expression levels. In addition, spatial training decreases both NCoR1 mRNA and protein expression, whereas NCoR1 conditional knockout (cKO) mice show enhanced spatial memory. In studying the molecular mechanism, we found that spatial training decreases the association between NCoR1 and DEC2. Both NCoR1 and DEC2 suppress the expression of BDNF, integrin α3 and SGK1 through C/EBPα binding to their DNA promoters, but overexpression of DEC2 in NCoR1 cKO mice rescues the decreased expression of these proteins compared with NCoR1 loxP mice overexpressing DEC2. Further, spatial training decreases DEC2 expression. Spatial training also enhances C/EBPα binding to Bdnf, Itga3 and Sgk1 promoters, an effect also observed in fast learners, and both NCoR1 and DEC2 control C/EBPα activity. Whereas knockdown of BDNF, integrin α3 or SGK1 expression impairs spatial learning and memory, it does not affect Y-maze performance, suggesting that BDNF, integrin α3 and SGK1 are involved in long-term memory formation, but not short-term memory formation. Moreover, NCoR1 expression is regulated by the JNK/c-Jun signaling pathway. Collectively, our findings identify DEC2 as a novel interacting protein of NCoR1 and elucidate the novel roles and mechanisms of NCoR1 and DEC2 in negative regulation of spatial memory formation.
Collapse
Affiliation(s)
- Kuang-Min Cheng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Wei-Lun Hsu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Yun-Li Ma
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Yen-Chen Liu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan
| | - Eminy H Y Lee
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
- Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan.
| |
Collapse
|
12
|
Rahamim N, Liran M, Aronovici C, Flumin H, Gordon T, Urshansky N, Barak S. Inhibition of ERK1/2 or CRMP2 Disrupts Alcohol Memory Reconsolidation and Prevents Relapse in Rats. Int J Mol Sci 2024; 25:5478. [PMID: 38791516 PMCID: PMC11122309 DOI: 10.3390/ijms25105478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Relapse to alcohol abuse, often caused by cue-induced alcohol craving, is a major challenge in alcohol addiction treatment. Therefore, disrupting the cue-alcohol memories can suppress relapse. Upon retrieval, memories transiently destabilize before they reconsolidate in a process that requires protein synthesis. Evidence suggests that the mammalian target of rapamycin complex 1 (mTORC1), governing the translation of a subset of dendritic proteins, is crucial for memory reconsolidation. Here, we explored the involvement of two regulatory pathways of mTORC1, phosphoinositide 3-kinase (PI3K)-AKT and extracellular regulated kinase 1/2 (ERK1/2), in the reconsolidation process in a rat (Wistar) model of alcohol self-administration. We found that retrieval of alcohol memories using an odor-taste cue increased ERK1/2 activation in the amygdala, while the PI3K-AKT pathway remained unaffected. Importantly, ERK1/2 inhibition after alcohol memory retrieval impaired alcohol-memory reconsolidation and led to long-lasting relapse suppression. Attenuation of relapse was also induced by post-retrieval administration of lacosamide, an inhibitor of collapsin response mediator protein-2 (CRMP2)-a translational product of mTORC1. Together, our findings indicate the crucial role of ERK1/2 and CRMP2 in the reconsolidation of alcohol memories, with their inhibition as potential treatment targets for relapse prevention.
Collapse
Affiliation(s)
- Nofar Rahamim
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Mirit Liran
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
- Faculty of Life Sciences, Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Coral Aronovici
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Hila Flumin
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Tamar Gordon
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Nataly Urshansky
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
| | - Segev Barak
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel; (N.R.)
- School of Psychological Sciences, Tel Aviv University, Tel Aviv 69978, Israel (N.U.)
- Faculty of Life Sciences, Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
13
|
Lalancette E, Cantin É, Routhier MÈ, Mailloux C, Bertrand MC, Kiaei DS, Larouche V, Tabori U, Hawkins C, Ellezam B, Décarie JC, Théoret Y, Métras MÉ, McKeown T, Ospina LH, Vairy S, Ramaswamy V, Coltin H, Sultan S, Legault G, Bouffet É, Lafay-Cousin L, Hukin J, Erker C, Caru M, Dehaes M, Jabado N, Perreault S, Lippé S. Impact of trametinib on the neuropsychological profile of NF1 patients. J Neurooncol 2024; 167:447-454. [PMID: 38443693 DOI: 10.1007/s11060-024-04624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024]
Abstract
PURPOSE The use of trametinib in the treatment of pediatric low-grade gliomas (PLGG) and plexiform neurofibroma (PN) is being investigated in an ongoing multicenter phase II trial (NCT03363217). Preliminary data shows potential benefits with significant response in the majority of PLGG and PN and an overall good tolerance. Moreover, possible benefits of MEK inhibitor therapy on cognitive functioning in neurofibromatosis type 1 (NF1) were recently shown which supports the need for further evaluation. METHODS Thirty-six patients with NF1 (age range 3-19 years) enrolled in the phase II study of trametinib underwent a neurocognitive assessment at inclusion and at completion of the 72-week treatment. Age-appropriate Wechsler Intelligence Scales and the Trail Making Test (for children over 8 years old) were administered at each assessment. Paired t-tests and Reliable Change Index (RCI) analyses were performed to investigate change in neurocognitive outcomes. Regression analyses were used to investigate the contribution of age and baseline score in the prediction of change. RESULTS Stable performance on neurocognitive tests was revealed at a group-level using paired t-tests. Clinically significant improvements were however found on specific indexes of the Wechsler intelligence scales and Trail Making Test, using RCI analyses. No significant impact of age on cognitive change was evidenced. However, lower initial cognitive performance was associated with increased odds of presenting clinically significant improvements on neurocognitive outcomes. CONCLUSION These preliminary results show a potential positive effect of trametinib on cognition in patients with NF1. We observed significant improvements in processing speed, visuo-motor and verbal abilities. This study demonstrates the importance of including neuropsychological evaluations into clinical trial when using MEK inhibitors for patients with NF1.
Collapse
Affiliation(s)
- Eve Lalancette
- CHU Sainte-Justine Research Center, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada.
| | - Édith Cantin
- Division of Neuropsychology, Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, QC, Canada
| | - Marie-Ève Routhier
- Division of Neuropsychology, Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, QC, Canada
| | - Chantal Mailloux
- Division of Neuropsychology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Marie-Claude Bertrand
- Division of Neuropsychology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Dorsa Sadat Kiaei
- CHU Sainte-Justine Research Center, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Valérie Larouche
- Division of Hemato-Oncology, Department of Pediatrics, Centre Hospitalier Universitaire de Québec-Université Laval, Quebec City, QC, Canada
| | - Uri Tabori
- Division of Hemato-Oncology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Cynthia Hawkins
- Department of Pathology, Hospital for Sick Children, Toronto, ON, Canada
| | - Benjamin Ellezam
- Department of Pathology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Jean-Claude Décarie
- Department of Radiology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Yves Théoret
- Department of Pharmacology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Marie-Élaine Métras
- Department of Pharmacology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Tara McKeown
- Division of Hemato-Oncology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Luis H Ospina
- Department of Ophthalmology, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Stéphanie Vairy
- Division of Hemato-Oncology, CHU Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Vijay Ramaswamy
- Division of Hemato-Oncology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Hallie Coltin
- Department of Hemato-Oncology, CHU Sainte Justine, Université de Montréal, Montréal, QC, Canada
| | - Serge Sultan
- CHU Sainte-Justine Research Center, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Geneviève Legault
- Division of Neurology, Department of Pediatrics, McGill University Health Center, Montreal Children's Hospital, Montréal, QC, Canada
| | - Éric Bouffet
- Division of Hemato-Oncology, Department of Pediatrics, Hospital for Sick Children, Toronto, ON, Canada
| | - Lucie Lafay-Cousin
- Departments of Oncology and Pediatrics, Alberta Children's Hospital, University of Calgary, Cumming School of Medicine, Calgary, AB, Canada
| | - Juliette Hukin
- Department of Pediatrics, Divisions of Neurology and Oncology, BC Children's Hospital, University of British Columbia, Vancouver, BCBC, Canada
| | - Craig Erker
- Division of Hemato-Oncology, Department of Pediatrics, IWK Health Centre, Dalhousie University, Halifax, NS, Canada
| | - Maxime Caru
- Department of Pediatrics, Division of Hematology and Oncology, Pennsylvania State Health Children's Hospital, Hershey, PA, USA
| | - Mathieu Dehaes
- CHU Sainte-Justine Research Center, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
- Department of Radiology, Radio-Oncology and Nuclear Medicine, University of Montréal, Montréal, Canada
| | - Nada Jabado
- Division of Hemato-Oncology, Department of Pediatrics, McGill University Health Center, Montreal Children's Hospital, Montréal, QC, Canada
| | - Sébastien Perreault
- Division of Child Neurology, Department of Pediatrics, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
| | - Sarah Lippé
- CHU Sainte-Justine Research Center, CHU Sainte-Justine, Université de Montréal, Montréal, QC, Canada
- Department of Psychology, Faculty of Arts and Sciences, University of Montréal, Montréal, Canada
| |
Collapse
|
14
|
Kannon T, Murashige S, Nishioka T, Amano M, Funahashi Y, Tsuboi D, Yamahashi Y, Nagai T, Kaibuchi K, Yoshimoto J. KANPHOS: Kinase-associated neural phospho-signaling database for data-driven research. Front Mol Neurosci 2024; 17:1379089. [PMID: 38628370 PMCID: PMC11018961 DOI: 10.3389/fnmol.2024.1379089] [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: 01/31/2024] [Accepted: 03/11/2024] [Indexed: 04/19/2024] Open
Abstract
Protein phosphorylation, a key regulator of cellular processes, plays a central role in brain function and is implicated in neurological disorders. Information on protein phosphorylation is expected to be a clue for understanding various neuropsychiatric disorders and developing therapeutic strategies. Nonetheless, existing databases lack a specific focus on phosphorylation events in the brain, which are crucial for investigating the downstream pathway regulated by neurotransmitters. To overcome the gap, we have developed a web-based database named "Kinase-Associated Neural PHOspho-Signaling (KANPHOS)." This paper presents the design concept, detailed features, and a series of improvements for KANPHOS. KANPHOS is designed to support data-driven research by fulfilling three key objectives: (1) enabling the search for protein kinases and their substrates related to extracellular signals or diseases; (2) facilitating a consolidated search for information encompassing phosphorylated substrate genes, proteins, mutant mice, diseases, and more; and (3) offering integrated functionalities to support pathway and network analysis. KANPHOS is also equipped with API functionality to interact with external databases and analysis tools, enhancing its utility in data-driven investigations. Those key features represent a critical step toward unraveling the complex landscape of protein phosphorylation in the brain, with implications for elucidating the molecular mechanisms underlying neurological disorders. KANPHOS is freely accessible to all researchers at https://kanphos.jp.
Collapse
Affiliation(s)
- Takayuki Kannon
- Department of Biomedical Data Science, Fujita Health University School of Medicine, Toyoake, Japan
- Division of Computational Science, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Satoshi Murashige
- Department of Biomedical Data Science, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tomoki Nishioka
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Mutsuki Amano
- Department of Cell Pharmacology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yasuhiro Funahashi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Daisuke Tsuboi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Yukie Yamahashi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| | - Junichiro Yoshimoto
- Department of Biomedical Data Science, Fujita Health University School of Medicine, Toyoake, Japan
- Division of Computational Science, International Center for Brain Science, Fujita Health University, Toyoake, Japan
| |
Collapse
|
15
|
Yang Y, Ma Q, Jin S, Huang B, Wang Z, Chen G. Identification of mapk genes, and their expression profiles in response to low salinity stress, in cobia (Rachycentron canadum). Comp Biochem Physiol B Biochem Mol Biol 2024; 271:110950. [PMID: 38307403 DOI: 10.1016/j.cbpb.2024.110950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/04/2024]
Abstract
Mitogen-activated protein kinases (MAPKs) are a class of protein kinases that regulate various physiological processes, and play a crucial role in maintaining the osmotic equilibrium of fish. The objective of this study was to identify and characterize the mapk family genes in cobia (Rachycentron canadum) and examine their expression profiles under different low salinity stress regimes (acute: from 30‰ to 10‰ in 1 h, sub-chronic: from 30‰ to 10‰ over 4 d). A total of 12 cobia mapk genes (Rcmapks) were identified and cloned, including six erk subfamily genes (Rcmapk1/3/4/6/7/15), three jnk subfamily genes (Rcmapk8/9/10) and three p38 mapk subfamily genes (Rcmapk 11/13/14). Domain analysis indicated that the RcMAPKs possessed the typical domains including S_TKc and PKc_like domain. Phylogenetic analysis revealed that the Rcmapks were most closely related to those of the turbot (Scophthalmus maximus). The tissue distribution of mapk genes in adult cobia and the expression patterns of Rcmapks under different low salinity stress regimes were investigated using quantitative real-time PCR (qRT-PCR). The results revealed that Rcmapk3/9/10/11/13/14 exhibited a relatively broad expression distribution across 14 different tissues. For all these genes the highest expression level was in the brain, except for Rcmapk14 (highly expressed in the stomach, gill, and skin). The genes Rcmapk1/6/15 showed significantly higher expression in the testis. Under acute low salinity stress, expression of Rcmapk1/3/6/7/9/11/13/14 was significantly altered in the gill, intestine, and trunk kidney, however, the aforementioned genes exhibited very different expression patterns among the three tissues. In the gill, most of the genes from the erk (Rcmapk3/6/7) and p38 mapk subfamily (Rcmapk11/13/14) were significantly up-regulated at almost all the time points (P < 0.05); Similarly, the expression of Rcmapk3/9/11/13/14 genes were significantly increased in the trunk kidney; while in the intestine, most of the altered genes (Rcmapk6/7/9/11/13/14) were significantly down-regulated at 1 h. Following the sub-chronic low salinity stress, expression of Rcmapk1/3/6/7/9/11/13/14 genes were significantly altered in all three tissues. These findings provide important reference data for elucidating the roles of cobia mapk family genes in response to low salinity stress.
Collapse
Affiliation(s)
- Yunsheng Yang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Qian Ma
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Shulei Jin
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Baosong Huang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Zhongliang Wang
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| | - Gang Chen
- College of Fisheries, Guangdong Ocean University, Zhanjiang 524088, China
| |
Collapse
|
16
|
Chen Y, Zhou Y, Cai J, Xu J, Hu C, Chen H, Hong Y, Pan N, Jiang Y, Zhou C, Wei H, Xu Z, Liu L, Wu X, Cui W. The activation of RARα prevents surgery-induced cognitive impairments via the inhibition of neuroinflammation and the restoration of synaptic proteins in elderly mice. Int Immunopharmacol 2024; 130:111772. [PMID: 38432148 DOI: 10.1016/j.intimp.2024.111772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Post-operative cognitive dysfunction (POCD) is a multi-etiological symptom mainly occurred in elderly people after surgery. The activation of retinoic acid receptor α (RARα), a transcriptional factor, was previously predicated to be negatively associated with the occurrence of POCD. However, the mechanisms underlying anti-POCD effects of RARα were still unclear. In this study, AM580, a selective agonist of RARα, and all-trans-retinoic acid (ATRA), a pan agonist of RAR, significantly alleviated cognitive dysfunction and increased the expression of RARα in elderly mice after surgery, which was decreased by RO41-5253, an antagonist of RARα. A bioinformatic study further predicted that the activation of RARα might produce anti-POCD effects via the restoration of synaptic proteins. Both agonists inhibited the expression of Toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (Myd88) and the phosphorylation of nuclear factorkappa-B (NF-κB), leading to the prevention of microglial over-activation and pro-inflammatory cytokines secretion in the hippocampal regions of elderly mice after surgery. Moreover, AM580 and ATRA increased the expression of brain-derived neurotrophic factor (BDNF) and postsynaptic density protein 95 (PSD95), and the phosphorylation of extracellular signal-regulated kinase (ERK) and cAMP-response element binding protein (CREB). All these results suggested that the activation of RARα prevented surgery-induced cognitive impairments via the inhibition of neuroinflammation by the reduction of the TLR4/Myd88/NF-κB pathway and the restoration of synaptic proteins by the activation of the BDNF/ERK/CREB pathway, providing a further support that RARα could be developed as a therapeutic target for POCD.
Collapse
Affiliation(s)
- Yuan Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, China; Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yi Zhou
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Jinhan Cai
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Jiayi Xu
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Chenwei Hu
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Huiyue Chen
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yirui Hong
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Nanyi Pan
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yujie Jiang
- Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China
| | - Chenhui Zhou
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, China
| | - Hua Wei
- Ningbo College of Health Sciences, Ningbo 315100, China
| | - Zhipeng Xu
- Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 200120, China
| | - Lin Liu
- Women and Children's Hospital of Ningbo University, Ningbo 315211, China
| | - Xiang Wu
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, China; Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China.
| | - Wei Cui
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, China; Translational Medicine Center of Pain, Emotion and Cognition, Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, China; Ningbo Kangning Hospital, Ningbo 315211, China.
| |
Collapse
|
17
|
Chen PJ, Yao CA, Chien PC, Tsai HJ, Chen YR, Chuang JH, Chou PL, Lee GC, Lin W, Lin Y. Paeonol Derivative, 6'-Methyl Paeonol, Attenuates Aβ-Induced Pathophysiology in Cortical Neurons and in an Alzheimer's Disease Mice Model. ACS Chem Neurosci 2024; 15:724-734. [PMID: 38290213 DOI: 10.1021/acschemneuro.3c00633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024] Open
Abstract
Herbs themselves and various herbal medicines are great resources for discovering therapeutic drugs for various diseases, including Alzheimer's disease (AD), one of the common neurodegenerative diseases. Utilizing mouse primary cortical neurons and DiBAC4(3), a voltage-sensitive indicator, we have set up a drug screening system and identified an herbal extraction compound, paeonol, obtained from Paeonia lactiflora; this compound is able to ameliorate the abnormal depolarization induced by Aβ42 oligomers. Our aim was to further find effective paeonol derivatives since paeonol has been previously studied. 6'-Methyl paeonol, one of the six paeonol derivatives surveyed, is able to inhibit the abnormal depolarization induced by Aβ oligomers. Furthermore, 6'-methyl paeonol is able to alleviate the NMDA- and AMPA-induced depolarization. When a molecular mechanism was investigated, 6'-methyl paeonol was found to reverse the Aβ-induced increase in ERK phosphorylation. At the animal level, mice injected with 6'-methyl paeonol showed little change in their basic physical parameters compared to the control mice. 6'-Methyl paeonol was able to ameliorate the impairment of memory and learning behavior in J20 mice, an AD mouse model, as measured by the Morris water maze. Thus, paeonol derivatives could provide a structural foundation for developing and designing an effective compound with promising clinical benefits.
Collapse
Affiliation(s)
| | - Chien-An Yao
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100225, Taiwan
| | | | | | | | | | - Pei-Li Chou
- Department of Family Medicine, National Taiwan University Hospital, Taipei, 100225, Taiwan
| | | | | | | |
Collapse
|
18
|
Jiang SZ, Shahoha M, Zhang HY, Brancaleone W, Elkahloun A, Tejeda HA, Ashery U, Eiden LE. The guanine nucleotide exchange factor RapGEF2 is required for ERK-dependent immediate-early gene (Egr1) activation during fear memory formation. Cell Mol Life Sci 2024; 81:48. [PMID: 38236296 PMCID: PMC11071968 DOI: 10.1007/s00018-023-04999-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 09/16/2023] [Accepted: 10/04/2023] [Indexed: 01/19/2024]
Abstract
The MAP kinase ERK is important for neuronal plasticity underlying associative learning, yet specific molecular pathways for neuronal ERK activation are undetermined. RapGEF2 is a neuron-specific cAMP sensor that mediates ERK activation. We investigated whether it is required for cAMP-dependent ERK activation leading to other downstream neuronal signaling events occurring during associative learning, and if RapGEF2-dependent signaling impairments affect learned behavior. Camk2α-cre+/-::RapGEF2fl/fl mice with depletion of RapGEF2 in hippocampus and amygdala exhibit impairments in context- and cue-dependent fear conditioning linked to corresponding impairment in Egr1 induction in these two brain regions. Camk2α-cre+/-::RapGEF2fl/fl mice show decreased RapGEF2 expression in CA1 and dentate gyrus associated with abolition of pERK and Egr1, but not of c-Fos induction, following fear conditioning, impaired freezing to context after fear conditioning, and impaired cAMP-dependent long-term potentiation at perforant pathway and Schaffer collateral synapses in hippocampal slices ex vivo. RapGEF2 expression is largely eliminated in basolateral amygdala, also involved in fear memory, in Camk2α-cre+/-::RapGEF2fl/fl mice. Neither Egr1 nor c-fos induction in BLA after fear conditioning, nor cue-dependent fear learning, are affected by ablation of RapGEF2 in BLA. However, Egr1 induction (but not that of c-fos) in BLA is reduced after restraint stress-augmented fear conditioning, as is freezing to cue after restraint stress-augmented fear conditioning, in Camk2α-cre+/-::RapGEF2fl/fl mice. Cyclic AMP-dependent GEFs have been genetically associated as risk factors for schizophrenia, a disorder associated with cognitive deficits. Here we show a functional link between one of them, RapGEF2, and cognitive processes involved in associative learning in amygdala and hippocampus.
Collapse
Affiliation(s)
- Sunny Zhihong Jiang
- Section On Molecular Neuroscience, NIMH Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, 20892, USA
| | - Meishar Shahoha
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, and Sagol School of Neuroscience, Tel Aviv University, Sherman Building Rm 719, Ramat Aviv, 69978, Tel Aviv, Israel
| | - Hai-Ying Zhang
- Section On Molecular Neuroscience, NIMH Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, 20892, USA
| | - William Brancaleone
- Section On Molecular Neuroscience, NIMH Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, 20892, USA
| | | | - Hugo A Tejeda
- Unit on Neuromodulation and Synaptic Integration, NIMH-IRP, Bethesda, MD, USA
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, and Sagol School of Neuroscience, Tel Aviv University, Sherman Building Rm 719, Ramat Aviv, 69978, Tel Aviv, Israel.
| | - Lee E Eiden
- Section On Molecular Neuroscience, NIMH Intramural Research Program, 9000 Rockville Pike, Building 49, Room 5A38, Bethesda, MD, 20892, USA.
| |
Collapse
|
19
|
Lee IB, Lee E, Han NE, Slavuj M, Hwang JW, Lee A, Sun T, Jeong Y, Baik JH, Park JY, Choi SY, Kwag J, Yoon BJ. Persistent enhancement of basolateral amygdala-dorsomedial striatum synapses causes compulsive-like behaviors in mice. Nat Commun 2024; 15:219. [PMID: 38191518 PMCID: PMC10774417 DOI: 10.1038/s41467-023-44322-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/08/2023] [Indexed: 01/10/2024] Open
Abstract
Compulsive behaviors are observed in a range of psychiatric disorders, however the neural substrates underlying the behaviors are not clearly defined. Here we show that the basolateral amygdala-dorsomedial striatum (BLA-DMS) circuit activation leads to the manifestation of compulsive-like behaviors. We revealed that the BLA neurons projecting to the DMS, mainly onto dopamine D1 receptor-expressing neurons, largely overlap with the neuronal population that responds to aversive predator stress, a widely used anxiogenic stressor. Specific optogenetic activation of the BLA-DMS circuit induced a strong anxiety response followed by compulsive grooming. Furthermore, we developed a mouse model for compulsivity displaying a wide spectrum of compulsive-like behaviors by chronically activating the BLA-DMS circuit. In these mice, persistent molecular changes at the BLA-DMS synapses observed were causally related to the compulsive-like phenotypes. Together, our study demonstrates the involvement of the BLA-DMS circuit in the emergence of enduring compulsive-like behaviors via its persistent synaptic changes.
Collapse
Affiliation(s)
- In Bum Lee
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Eugene Lee
- Department of Brain and Cognitive Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Na-Eun Han
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Marko Slavuj
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Jeong Wook Hwang
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Ahrim Lee
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Taeyoung Sun
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Yehwan Jeong
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Ja-Hyun Baik
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Se-Young Choi
- Department of Physiology, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, Republic of Korea
| | - Jeehyun Kwag
- Department of Brain and Cognitive Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Bong-June Yoon
- Department of Life Sciences, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
20
|
Gómez-Pineda VG, Nieto-Mendoza E, Torres-Cruz FM, Hernández-Echeagaray E. Neurotrophin-3 Rescues Striatal Synaptic Plasticity in Model of Neurodegeneration by PLC Signaling Activation. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1488-1498. [PMID: 38859788 DOI: 10.2174/0118715273298919240531110022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Neurotrophins are essential factors for neural growth and function; they play a crucial role in neurodegenerative diseases where their expression levels are altered. Our previous research has demonstrated changes in synaptic plasticity and neurotrophin expression levels in a pharmacological model of Huntington's disease (HD) induced by 3-nitropropionic acid (3-NP). In the 3-NP-induced HD model, corticostriatal Long Term Depression (LTD) was impaired, but neurotrophin- 3 (NT-3) restored striatal LTD. This study delves into the NT-3-induced signaling pathways involved in modulating and restoring striatal synaptic plasticity in cerebral slices from 3-NPinduced striatal degeneration in mice in vivo. METHODS Phospholipase C (PLC), phosphatidylinositol-3-kinase (PI3K), and mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways activated by NT-3 were analyzed by means of field electrophysiological recordings in brain slices from control and 3-NP treated in the presence of specific inhibitors of the signaling pathways. RESULTS Using specific inhibitors, PLC, PI3K, and MEK/ERK signaling pathways contribute to NT-3-mediated plasticity modulation in striatal tissue slices recorded from control animals. However, in the neurodegeneration model induced by 3-NP, the recovery of striatal LTD induced by NT-3 was prevented only by the PLC inhibitor. Moreover, the PLC signaling pathway appeared to trigger downstream activation of the endocannabinoid system, evidenced by AM 251, an inhibitor of the CB1 receptor, also hindered NT-3 plasticity recovery. CONCLUSION Our finding highlights the specific involvement of the PLC pathway in the neuroprotective effects of NT-3 in mitigating synaptic dysfunction under neurodegenerative conditions.
Collapse
Affiliation(s)
- Victor G Gómez-Pineda
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Facultad de Estudios Superiores Iztacala, Unidad de Investigación en Biomedicina, Universidad Nacional Autónoma de México, Av. De los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, C.P. 54090, México
| | - Elizabeth Nieto-Mendoza
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Facultad de Estudios Superiores Iztacala, Unidad de Investigación en Biomedicina, Universidad Nacional Autónoma de México, Av. De los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, C.P. 54090, México
| | - Francisco M Torres-Cruz
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Facultad de Estudios Superiores Iztacala, Unidad de Investigación en Biomedicina, Universidad Nacional Autónoma de México, Av. De los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, C.P. 54090, México
| | - Elizabeth Hernández-Echeagaray
- Laboratorio de Neurofisiología del Desarrollo y la Neurodegeneración, Facultad de Estudios Superiores Iztacala, Unidad de Investigación en Biomedicina, Universidad Nacional Autónoma de México, Av. De los Barrios No. 1, Los Reyes Iztacala, Tlalnepantla, Estado de México, C.P. 54090, México
| |
Collapse
|
21
|
He Y, Nan D, Wang H. Role of Non-Receptor-Type Tyrosine Phosphatases in Brain-Related Diseases. Mol Neurobiol 2023; 60:6530-6541. [PMID: 37458988 DOI: 10.1007/s12035-023-03487-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/05/2023] [Indexed: 09/28/2023]
Abstract
The non-receptor protein tyrosine phosphatase is a class of enzymes that catalyze the dephosphorylation of phosphotyrosines in protein molecules. They are involved in cellular signaling by regulating the phosphorylation status of a variety of receptors and signaling molecules within the cell, thereby influencing cellular physiological and pathological processes. In this article, we detail multiple non-receptor tyrosine phosphatase and non-receptor tyrosine phosphatase genes involved in the pathological process of brain disease. These include PTPN6, PTPN11, and PTPN13, which are involved in glioma signaling; PTPN1, PTPN5, and PTPN13, which are involved in the pathogenesis of Alzheimer's disease Tau protein lesions, PTPN23, which may be involved in the pathogenesis of Epilepsy and PTPN1, which is involved in the pathogenesis of Parkinson's disease. The role of mitochondrial tyrosine phosphatase in brain diseases was also discussed. Non-receptor tyrosine phosphatases have great potential for targeted therapies in brain diseases and are highly promising research areas.
Collapse
Affiliation(s)
- Yatong He
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Ding Nan
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China
| | - Hongmei Wang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
22
|
Forrest MP, Penzes P. Mechanisms of copy number variants in neuropsychiatric disorders: From genes to therapeutics. Curr Opin Neurobiol 2023; 82:102750. [PMID: 37515924 PMCID: PMC10529795 DOI: 10.1016/j.conb.2023.102750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 06/01/2023] [Accepted: 06/27/2023] [Indexed: 07/31/2023]
Abstract
Copy number variants (CNVs) are genomic imbalances strongly linked to the aetiology of neuropsychiatric disorders such as schizophrenia and autism. By virtue of their large size, CNVs often contain many genes, providing a multi-genic view of disease processes that can be dissected in model systems. Thus, CNV research provides an important stepping stone towards understanding polygenic disease mechanisms, positioned between monogenic and polygenic risk models. In this review, we will outline hypothetical models for gene interactions occurring within CNVs and discuss different approaches used to study rodent and stem cell disease models. We highlight recent work showing that genetic and pharmacological strategies can be used to rescue important aspects of CNV-mediated pathophysiology, which often converges onto synaptic pathways. We propose that using a rescue approach in complete CNV models provides a new path forward for precise mechanistic understanding of complex disorders and a tangible route towards therapeutic development.
Collapse
Affiliation(s)
- Marc P Forrest
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Peter Penzes
- Department of Neuroscience, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
23
|
Latif-Hernandez A, Yang T, Raymond-Butler R, Losada PM, Minhas P, White H, Tran KC, Liu H, Simmons DA, Langness V, Andreasson K, Wyss-Coray T, Longo FM. A TrkB and TrkC partial agonist restores deficits in synaptic function and promotes activity-dependent synaptic and microglial transcriptomic changes in a late-stage Alzheimer's mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558138. [PMID: 37781573 PMCID: PMC10541128 DOI: 10.1101/2023.09.18.558138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
Introduction TrkB and TrkC receptor signaling promotes synaptic plasticity and interacts with pathways affected by amyloid-β (Aβ)-toxicity. Upregulating TrkB/C signaling could reduce Alzheimer's disease (AD)-related degenerative signaling, memory loss, and synaptic dysfunction. Methods PTX-BD10-2 (BD10-2), a small molecule TrkB/C receptor partial agonist, was orally administered to aged London/Swedish-APP mutant mice (APP L/S ) and wild-type controls (WT). Effects on memory and hippocampal long-term potentiation (LTP) were assessed using electrophysiology, behavioral studies, immunoblotting, immunofluorescence staining, and RNA-sequencing. Results Memory and LTP deficits in APP L/S mice were attenuated by treatment with BD10-2. BD10-2 prevented aberrant AKT, CaMKII, and GLUA1 phosphorylation, and enhanced activity-dependent recruitment of synaptic proteins. BD10-2 also had potentially favorable effects on LTP-dependent complement pathway and synaptic gene transcription. Conclusions BD10-2 prevented APP L/S /Aβ-associated memory and LTP deficits, reduced abnormalities in synapse-related signaling and activity-dependent transcription of synaptic genes, and bolstered transcriptional changes associated with microglial immune response.
Collapse
|
24
|
Zhao J, Bang S, Furutani K, McGinnis A, Jiang C, Roberts A, Donnelly CR, He Q, James ML, Berger M, Ko MC, Wang H, Palmiter RD, Ji RR. PD-L1/PD-1 checkpoint pathway regulates hippocampal neuronal excitability and learning and memory behavior. Neuron 2023; 111:2709-2726.e9. [PMID: 37348508 PMCID: PMC10529885 DOI: 10.1016/j.neuron.2023.05.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/15/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023]
Abstract
Programmed death protein 1 (PD-1) and its ligand PD-L1 constitute an immune checkpoint pathway. We report that neuronal PD-1 signaling regulates learning/memory in health and disease. Mice lacking PD-1 (encoded by Pdcd1) exhibit enhanced long-term potentiation (LTP) and memory. Intraventricular administration of anti-mouse PD-1 monoclonal antibody (RMP1-14) potentiated learning and memory. Selective deletion of PD-1 in excitatory neurons (but not microglia) also enhances LTP and memory. Traumatic brain injury (TBI) impairs learning and memory, which is rescued by Pdcd1 deletion or intraventricular PD-1 blockade. Conversely, re-expression of Pdcd1 in PD-1-deficient hippocampal neurons suppresses memory and LTP. Exogenous PD-L1 suppresses learning/memory in mice and the excitability of mouse and NHP hippocampal neurons through PD-1. Notably, neuronal activation suppresses PD-L1 secretion, and PD-L1/PD-1 signaling is distinctly regulated by learning and TBI. Thus, conditions that reduce PD-L1 levels or PD-1 signaling could promote memory in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Junli Zhao
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Sangsu Bang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Kenta Furutani
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Aidan McGinnis
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Changyu Jiang
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Alexus Roberts
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Christopher R Donnelly
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Qianru He
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Michael L James
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Miles Berger
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mei-Chuan Ko
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Haichen Wang
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| | - Richard D Palmiter
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Ru-Rong Ji
- Center for Translational Pain Medicine, Department of Anesthesiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
25
|
Samadi M, Hales CA, Lustberg DJ, Farris S, Ross MR, Zhao M, Hepler JR, Harbin NH, Robinson ESJ, Banks PJ, Bashir ZI, Dudek SM. Mechanisms of mGluR-dependent plasticity in hippocampal area CA2. Hippocampus 2023; 33:730-744. [PMID: 36971428 PMCID: PMC10213158 DOI: 10.1002/hipo.23529] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/29/2023]
Abstract
Pyramidal cells in hippocampal area CA2 have synaptic properties that are distinct from the other CA subregions. Notably, this includes a lack of typical long-term potentiation of stratum radiatum synapses. CA2 neurons express high levels of several known and potential regulators of metabotropic glutamate receptor (mGluR)-dependent signaling including Striatal-Enriched Tyrosine Phosphatase (STEP) and several Regulator of G-protein Signaling (RGS) proteins, yet the functions of these proteins in regulating mGluR-dependent synaptic plasticity in CA2 are completely unknown. Thus, the aim of this study was to examine mGluR-dependent synaptic depression and to determine whether STEP and the RGS proteins RGS4 and RGS14 are involved. Using whole cell voltage-clamp recordings from mouse pyramidal cells, we found that mGluR agonist-induced long-term depression (mGluR-LTD) is more pronounced in CA2 compared with that observed in CA1. This mGluR-LTD in CA2 was found to be protein synthesis and STEP dependent, suggesting that CA2 mGluR-LTD shares mechanistic processes with those seen in CA1, but in addition, RGS14, but not RGS4, was essential for mGluR-LTD in CA2. In addition, we found that exogenous application of STEP could rescue mGluR-LTD in RGS14 KO slices. Supporting a role for CA2 synaptic plasticity in social cognition, we found that RGS14 KO mice had impaired social recognition memory as assessed in a social discrimination task. These results highlight possible roles for mGluRs, RGS14, and STEP in CA2-dependent behaviors, perhaps by biasing the dominant form of synaptic plasticity away from LTP and toward LTD in CA2.
Collapse
Affiliation(s)
- Mahsa Samadi
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences BuildingUniversity Walk, University of BristolBristolUKBS8 1TD
- Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIH)111 T.W. Alexander Drive, Research Triangle ParkDurhamNorth Carolina27709USA
- Present address:
Faculty Education Office, Faculty of Medicine, Imperial College London, Hammersmith Campus, Wolfson Education CentreLondonUKW12 0NN
| | - Claire A. Hales
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences BuildingUniversity Walk, University of BristolBristolUKBS8 1TD
- Present address:
Department of Psychology, Djavad Mowafaghian Centre for Brain HealthUniversity of British Columbia2215, Wesbrook MallVancouverBritish ColumbiaV6T 1Z3Canada
| | - Daniel J. Lustberg
- Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIH)111 T.W. Alexander Drive, Research Triangle ParkDurhamNorth Carolina27709USA
- Present address:
Mouse Pharmacology GroupPsychogenics Inc215 College RoadParamusNew Jersey07652USA
| | - Shannon Farris
- Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIH)111 T.W. Alexander Drive, Research Triangle ParkDurhamNorth Carolina27709USA
- Present address:
Fralin Biomedical Research Institute at Virginia TechRoanokeVirginia24014USA
| | - Madeleine R. Ross
- Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIH)111 T.W. Alexander Drive, Research Triangle ParkDurhamNorth Carolina27709USA
| | - Meilan Zhao
- Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIH)111 T.W. Alexander Drive, Research Triangle ParkDurhamNorth Carolina27709USA
| | - John R. Hepler
- Department of Pharmacology and Chemical BiologyEmory University School of Medicine100 Woodruff CircleAtlantaGeorgia30322USA
| | - Nicholas H. Harbin
- Department of Pharmacology and Chemical BiologyEmory University School of Medicine100 Woodruff CircleAtlantaGeorgia30322USA
| | - Emma S. J. Robinson
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences BuildingUniversity Walk, University of BristolBristolUKBS8 1TD
| | - Paul J. Banks
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences BuildingUniversity Walk, University of BristolBristolUKBS8 1TD
| | - Zafar I. Bashir
- School of Physiology, Pharmacology and Neuroscience, Biomedical Sciences BuildingUniversity Walk, University of BristolBristolUKBS8 1TD
| | - Serena M. Dudek
- Neurobiology Laboratory, National Institute of Environmental Health Sciences (NIH)111 T.W. Alexander Drive, Research Triangle ParkDurhamNorth Carolina27709USA
| |
Collapse
|
26
|
Quan J, Yang H, Qin F, He Y, Liu J, Zhao Y, Ma C, Cheng M. Discovery of novel tryptamine derivatives as GluN2B subunit-containing NMDA receptor antagonists via pharmacophore-merging strategy with orally available therapeutic effect of cerebral ischemia. Eur J Med Chem 2023; 253:115318. [PMID: 37037139 DOI: 10.1016/j.ejmech.2023.115318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/16/2023] [Accepted: 03/27/2023] [Indexed: 04/12/2023]
Abstract
A series of tryptamine derivatives has been designed and synthesized as novel GluN2B subunit-containing NMDA receptor (GluN2B-NMDAR) antagonists, which could simultaneously manifest the receptor-ligand interactions of representative GluN2B-NMDAR antagonists ifenprodil (1) and EVT-101 (3). In the present study, the neuroprotective potential of these compounds was explored through chemical synthesis and pharmacological characterization. Compound Z25 with significantly better neuroprotective activity than the positive control drug (percentage of protection: 55.8 ± 0.6% vs. 41.0 ± 2.7%) was considered to be an effective antagonist of the human GluN2B-NMDA receptor. Judging from in vitro pharmacological profiling, Z25 could downregulate NMDA-induced increased intracellular Ca2+ concentration, and Z25 could also upregulate NMDA-induced decreased intracellular p-ERK 1/2 expression, which suggested that Z25 is an antagonist of the GluN2B-NMDA receptor. Furthermore, the in vitro preliminary evaluation of the drug-like properties of compound Z25 showed remarkable plasma stability. Based on in vivo pharmacokinetic and pharmacodynamic studies in C57 mice, compound Z25 exhibited a relatively short half-life and a low F value (3.12 ± 0.01%), while administration of Z25 substantially improved the cognitive performance of mice in a series of tests of cerebral ischemic injury. Overall, these results support the further development of compound Z25 as a potential lead compound to treat the cerebral ischemic injury by antagonizing GluN2B-NMDA receptor.
Collapse
Affiliation(s)
- Jishun Quan
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, People's Republic of China
| | - Huali Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, People's Republic of China
| | - Fengyun Qin
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, People's Republic of China
| | - Yeli He
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, People's Republic of China
| | - Jiao Liu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, People's Republic of China
| | - Ying Zhao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, People's Republic of China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, People's Republic of China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, People's Republic of China.
| |
Collapse
|
27
|
Kumar H, Chakrabarti A, Sarma P, Modi M, Banerjee D, Radotra BD, Bhatia A, Medhi B. Novel therapeutic mechanism of action of metformin and its nanoformulation in Alzheimer's disease and role of AKT/ERK/GSK pathway. Eur J Pharm Sci 2023; 181:106348. [PMID: 36496166 DOI: 10.1016/j.ejps.2022.106348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Brain Insulin-resistance plays a critical role in pathogenesis of Alzheimer's disease (AD). Current study explored the therapeutic mechanism of metformin (insulin sensitizer) and its solid-lipid nanoformulation (SLN) in rat-model of AD. In our study, SLN was prepared using microemulsion method. AD was induced with ICV-Aβ whereas the control-group (sham) received ICV-NS. Treatment arms included, disease-control (no treatment), Metformin (50 mg/kg, 100 mg/kg and 150 mg/kg), SLN-metformin 50 mg/kg and memantine 1.8 mg/kg (positive-control). Animals were tested for cognitive performance (EPM, MWM) after 21 days of therapy and sacrificed. Aβ (1-42), hyperphosphorylated tau, pAKTser473, GSK-3β, p-ERK (ELISA), metformin level(HPLC), neuronal injury score(H&E), Bcl2 and Bax(IHC) was evaluated in isolated brain. In our study, metformin-SLN were of spherical shape (size<200 nm) with 94.08% entrapment efficiency. Metformin was detectable in brain. Compared to sham, the disease-control group showed significantly higher (p ≤ 0.05) memory impairment(MWM and EPM), hyperphosphorylated tau, Aβ(1-42), neuronal-injury, Bax and lower Bcl-2 expression. Treatment with metformin and nanoformulation significantly reverse these parameters. AKT-ERK-GSK3β-Hyperphosphorylated tau pathway was found to be involved in the protective efficacy of metformin. To conclude, both metformin and its SLN were found to be effective as therapeutic agents in AD which act through the AKT-ERK-GSK3β-Hyperphosphorylated tau pathway. We need population based studies to confirm the same.
Collapse
Affiliation(s)
| | | | | | | | | | - B D Radotra
- Department of Histopathology, PGIMER, Chandigarh
| | - Alka Bhatia
- Department of Experimental Medicine & Biotechnology, PGIMER, Chandigarh.
| | | |
Collapse
|
28
|
Basavarajappa BS, Subbanna S. Synaptic Plasticity Abnormalities in Fetal Alcohol Spectrum Disorders. Cells 2023; 12:442. [PMID: 36766783 PMCID: PMC9913617 DOI: 10.3390/cells12030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/10/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
The brain's ability to strengthen or weaken synaptic connections is often termed synaptic plasticity. It has been shown to function in brain remodeling following different types of brain damage (e.g., drugs of abuse, alcohol use disorders, neurodegenerative diseases, and inflammatory conditions). Although synaptic plasticity mechanisms have been extensively studied, how neural plasticity can influence neurobehavioral abnormalities in alcohol use disorders (AUDs) is far from being completely understood. Alcohol use during pregnancy and its harmful effects on the developing offspring are major public health, social, and economic challenges. The significant attribute of prenatal alcohol exposure on offspring is damage to the central nervous system (CNS), causing a range of synaptic structural, functional, and behavioral impairments, collectively called fetal alcohol spectrum disorder (FASD). Although the synaptic mechanisms in FASD are limited, emerging evidence suggests that FASD pathogenesis involves altering a set of molecules involved in neurotransmission, myelination, and neuroinflammation. These studies identify several immediate and long-lasting changes using many molecular approaches that are essential for synaptic plasticity and cognitive function. Therefore, they can offer potential synaptic targets for the many neurobehavioral abnormalities observed in FASD. In this review, we discuss the substantial research progress in different aspects of synaptic and molecular changes that can shed light on the mechanism of synaptic dysfunction in FASD. Increasing our understanding of the synaptic changes in FASD will significantly advance our knowledge and could provide a basis for finding novel therapeutic targets and innovative treatment strategies.
Collapse
Affiliation(s)
- Balapal S. Basavarajappa
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
- Molecular Imaging and Neuropathology Area, New York State Psychiatric Institute, New York, NY 10032, USA
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY 10032, USA
- Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA
| | - Shivakumar Subbanna
- Center for Dementia Research, Nathan Kline Institute for Psychiatric Research, Orangeburg, NY 10962, USA
| |
Collapse
|
29
|
G-protein coupled estrogen receptor (GPER1) activation promotes synaptic insertion of AMPA receptors and induction of chemical LTP at hippocampal temporoammonic-CA1 synapses. Mol Brain 2023; 16:16. [PMID: 36709268 PMCID: PMC9883958 DOI: 10.1186/s13041-023-01003-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/12/2023] [Indexed: 01/30/2023] Open
Abstract
It is well documented that 17β estradiol (E2) regulates excitatory synaptic transmission at hippocampal Shaffer-collateral (SC)-CA1 synapses, via activation of the classical estrogen receptors (ERα and ERβ). Hippocampal CA1 pyramidal neurons are also innervated by the temporoammonic (TA) pathway, and excitatory TA-CA1 synapses are reported to be regulated by E2. Recent studies suggest a role for the novel G-protein coupled estrogen receptor (GPER1) at SC-CA1 synapses, however, the role of GPER1 in mediating the effects of E2 at juvenile TA-CA1 synapses is unclear. Here we demonstrate that the GPER1 agonist, G1 induces a persistent, concentration-dependent (1-10 nM) increase in excitatory synaptic transmission at TA-CA1 synapses and this effect is blocked by selective GPER1 antagonists. The ability of GPER1 to induce this novel form of chemical long-term potentiation (cLTP) was prevented following blockade of N-methyl-D-aspartate (NMDA) receptors, and it was not accompanied by any change in paired pulse facilitation ratio (PPR). GPER1-induced cLTP involved activation of ERK but was independent of phosphoinositide 3-kinase (PI3K) signalling. Prior treatment with philanthotoxin prevented the effects of G1, indicating that synaptic insertion of GluA2-lacking α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors underlies GPER1-induced cLTP. Furthermore, activity-dependent LTP occluded G1-induced cLTP and vice versa, indicating that these processes have overlapping expression mechanisms. Activity-dependent LTP was blocked by the GPER1 antagonist, G15, suggesting that GPER1 plays a role in NMDA-dependent LTP at juvenile TA-CA1 synapses. These findings add a new dimension to our understanding of GPER1 in modulating neuronal plasticity with relevance to age-related neurodegenerative conditions.
Collapse
|
30
|
Yáñez-Gómez F, Ramos-Miguel A, García-Sevilla JA, Manzanares J, Femenía T. Regulation of Cortico-Thalamic JNK1/2 and ERK1/2 MAPKs and Apoptosis-Related Signaling Pathways in PDYN Gene-Deficient Mice Following Acute and Chronic Mild Stress. Int J Mol Sci 2023; 24:ijms24032303. [PMID: 36768626 PMCID: PMC9916432 DOI: 10.3390/ijms24032303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
The crosstalk between the opioidergic system and mitogen-activated protein kinases (MAPKs) has a critical role in mediating stress-induced behaviors related to the pathophysiology of anxiety. The present study evaluated the basal status and stress-induced alterations of cortico-thalamic MAPKs and other cell fate-related signaling pathways potentially underlying the anxiogenic endophenotype of PDYN gene-deficient mice. Compared to littermates, PDYN knockout (KO) mice had lower cortical and or thalamic amounts of the phospho-activated MAPKs c-Jun N-terminal kinase (JNK1/2) and extracellular signal-regulated kinase (ERK1/2). Similarly, PDYN-KO animals displayed reduced cortico-thalamic densities of total and phosphorylated (at Ser191) species of the cell fate regulator Fas-associated protein with death domain (FADD) without alterations in the Fas receptor. Exposure to acute restraint and chronic mild stress stimuli induced the robust stimulation of JNK1/2 and ERK1/2 MAPKs, FADD, and Akt-mTOR pathways, without apparent increases in apoptotic rates. Interestingly, PDYN deficiency prevented stress-induced JNK1/2 and FADD but not ERK1/2 or Akt-mTOR hyperactivations. These findings suggest that cortico-thalamic MAPK- and FADD-dependent neuroplasticity might be altered in PDYN-KO mice. In addition, the results also indicate that the PDYN gene (and hence dynorphin release) may be required to stimulate JNK1/2 and FADD (but not ERK1/2 or Akt/mTOR) pathways under environmental stress conditions.
Collapse
Affiliation(s)
- Fernando Yáñez-Gómez
- Laboratorio de Neurofarmacología, IUNICS, Universitat de les Illes Balears, Crta. Valldemossa km 7.5, 07122 Palma de Mallorca, Spain
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain
| | - Alfredo Ramos-Miguel
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barrio Sarriena S/N, 48940 Leioa, Spain
- BioCruces Bizkaia Health Research Institute, Plaza de Cruces 12, 48903 Barakaldo, Spain
- Correspondence:
| | - Jesús A. García-Sevilla
- Laboratorio de Neurofarmacología, IUNICS, Universitat de les Illes Balears, Crta. Valldemossa km 7.5, 07122 Palma de Mallorca, Spain
| | - Jorge Manzanares
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Teresa Femenía
- Instituto de Neurociencias de Alicante, Universidad Miguel Hernández-CSIC, Avda. de Ramón y Cajal s/n, San Juan de Alicante, 03550 Alicante, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Red de Investigación en Atención Primaria de Adicciones (RIAPAd), Instituto de Salud Carlos III, MICINN and FEDER, 28029 Madrid, Spain
| |
Collapse
|
31
|
Opioid-Modulated Receptor Localization and Erk1/2 Phosphorylation in Cells Coexpressing μ-Opioid and Nociceptin Receptors. Int J Mol Sci 2023; 24:ijms24021048. [PMID: 36674576 PMCID: PMC9865058 DOI: 10.3390/ijms24021048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
We attempted to examine the alterations elicited by opioids via coexpressed μ-opioid (MOP) and nociceptin/orphanin FQ (NOP) receptors for receptor localization and Erk1/2 (p44/42 MAPK) in human embryonic kidney (HEK) 293 cells. Through two-photon microscopy, the proximity of MOP and NOP receptors was verified by fluorescence resonance energy transfer (FRET), and morphine but not buprenorphine facilitated the process of MOP-NOP heterodimerization. Single-particle tracking (SPT) further revealed that morphine or buprenorphine hindered the movement of the MOP-NOP heterodimers. After exposure to morphine or buprenorphine, receptor localization on lipid rafts was detected by immunocytochemistry, and phosphorylation of Erk1/2 was determined by immunoblotting in HEK 293 cells expressing MOP, NOP, or MOP+NOP receptors. Colocalization of MOP and NOP on lipid rafts was enhanced by morphine but not buprenorphine. Morphine stimulated the phosphorylation of Erk1/2 with a similar potency in HEK 293 cells expressing MOP and MOP+NOP receptors, but buprenorphine appeared to activate Erk1/2 solely through NOP receptors. Our results suggest that opioids can fine-tune the cellular localization of opioid receptors and phosphorylation of Erk1/2 in MOP+NOP-expressing cells.
Collapse
|
32
|
Sciaccaluga M, Ruffolo G, Palma E, Costa C. Traditional and Innovative Anti-seizure Medications Targeting Key Physiopathological Mechanisms: Focus on Neurodevelopment and Neurodegeneration. Curr Neuropharmacol 2023; 21:1736-1754. [PMID: 37143270 PMCID: PMC10514539 DOI: 10.2174/1570159x21666230504160948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 05/06/2023] Open
Abstract
Despite the wide range of compounds currently available to treat epilepsy, there is still no drug that directly tackles the physiopathological mechanisms underlying its development. Indeed, antiseizure medications attempt to prevent seizures but are inefficacious in counteracting or rescuing the physiopathological phenomena that underlie their onset and recurrence, and hence do not cure epilepsy. Classically, the altered excitation/inhibition balance is postulated as the mechanism underlying epileptogenesis and seizure generation. This oversimplification, however, does not account for deficits in homeostatic plasticity resulting from either insufficient or excessive compensatory mechanisms in response to a change in network activity. In this respect, both neurodevelopmental epilepsies and those associated with neurodegeneration may share common underlying mechanisms that still need to be fully elucidated. The understanding of these molecular mechanisms shed light on the identification of new classes of drugs able not only to suppress seizures, but also to present potential antiepileptogenic effects or "disease-modifying" properties.
Collapse
Affiliation(s)
- Miriam Sciaccaluga
- Section of Neurology, S.M. della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06129, Italy
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome, Sapienza, Rome, 00185, Italy
- IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Eleonora Palma
- Department of Physiology and Pharmacology, Istituto Pasteur—Fondazione Cenci Bolognetti, University of Rome, Sapienza, Rome, 00185, Italy
- IRCCS San Raffaele Roma, Rome, 00166, Italy
| | - Cinzia Costa
- Section of Neurology, S.M. della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Piazzale Gambuli 1, Perugia, 06129, Italy
| |
Collapse
|
33
|
Leahy SN, Song C, Vita DJ, Broadie K. FMRP activity and control of Csw/SHP2 translation regulate MAPK-dependent synaptic transmission. PLoS Biol 2023; 21:e3001969. [PMID: 36701299 PMCID: PMC9879533 DOI: 10.1371/journal.pbio.3001969] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 12/16/2022] [Indexed: 01/27/2023] Open
Abstract
Noonan syndrome (NS) and NS with multiple lentigines (NSML) cognitive dysfunction are linked to SH2 domain-containing protein tyrosine phosphatase-2 (SHP2) gain-of-function (GoF) and loss-of-function (LoF), respectively. In Drosophila disease models, we find both SHP2 mutations from human patients and corkscrew (csw) homolog LoF/GoF elevate glutamatergic transmission. Cell-targeted RNAi and neurotransmitter release analyses reveal a presynaptic requirement. Consistently, all mutants exhibit reduced synaptic depression during high-frequency stimulation. Both LoF and GoF mutants also show impaired synaptic plasticity, including reduced facilitation, augmentation, and post-tetanic potentiation. NS/NSML diseases are characterized by elevated MAPK/ERK signaling, and drugs suppressing this signaling restore normal neurotransmission in mutants. Fragile X syndrome (FXS) is likewise characterized by elevated MAPK/ERK signaling. Fragile X Mental Retardation Protein (FMRP) binds csw mRNA and neuronal Csw protein is elevated in Drosophila fragile X mental retardation 1 (dfmr1) nulls. Moreover, phosphorylated ERK (pERK) is increased in dfmr1 and csw null presynaptic boutons. We find presynaptic pERK activation in response to stimulation is reduced in dfmr1 and csw nulls. Trans-heterozygous csw/+; dfmr1/+ recapitulate elevated presynaptic pERK activation and function, showing FMRP and Csw/SHP2 act within the same signaling pathway. Thus, a FMRP and SHP2 MAPK/ERK regulative mechanism controls basal and activity-dependent neurotransmission strength.
Collapse
Affiliation(s)
- Shannon N. Leahy
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Chunzhu Song
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Dominic J. Vita
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
34
|
Jee HJ, Ryu D, Kim S, Yeon SH, Son RH, Hwang SH, Jung YS. Fermented Perilla frutescens Ameliorates Depression-like Behavior in Sleep-Deprivation-Induced Stress Model. Int J Mol Sci 2022; 24:ijms24010622. [PMID: 36614066 PMCID: PMC9820360 DOI: 10.3390/ijms24010622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
Excessive stress plays a critical role in the pathogenesis of mood disorders such as depression. Fermented natural products have recently attracted attention because of their health benefits. We evaluated the antidepressant-like efficacy of fermented Perilla frutescens (FPF), and its underlying mechanisms, in sleep deprivation (SD)-induced stress mice. SD-stressed mice revealed a remarkable increase in the immobility time in both forced swimming test and tail suspension test; this increase was ameliorated by treatment with FPF at doses of 100 and 150 mg/kg. FPF treatment also reduced the level of stress hormones such as corticosterone and adrenocorticotropic hormone. Additionally, FPF increased the levels of serotonin and dopamine which were significantly decreased in the brain tissues of SD-stressed mice. The increased expression of proinflammatory cytokines, such as TNF-α and IL1β, and the decreased expression of brain-derived neurotrophic factor (BDNF) in the stressed mice were significantly reversed by FPF treatment. Furthermore, FPF also increased phosphorylation of tropomyosin receptor kinase B (TrkB), extracellular regulated protein kinase (ERK), and cAMP response element binding protein (CREB). Among the six components isolated from FPF, protocatechuic acid and luteolin-7-O-glucuronide exhibited significant antidepressant-like effects, suggesting that they are major active components. These findings suggest that FPF has therapeutic potential for SD-induced stress, by correcting dysfunction of hypothalamic-pituitary-adrenal axis and modulating the BDNF/TrkB/ERK/CREB signaling pathway.
Collapse
Affiliation(s)
- Hye Jin Jee
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- AI-Super Convergence KIURI Translational Research Center, School of Medicine, Ajou University, Suwon 16499, Republic of Korea
| | - Dajung Ryu
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Suyeon Kim
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Sung Hum Yeon
- R&D Center, Huons Co., Ltd., 55 Hanyangdaehak-ro, Ansan 15588, Republic of Korea
| | - Rak Ho Son
- R&D Center, Huons Co., Ltd., 55 Hanyangdaehak-ro, Ansan 15588, Republic of Korea
| | - Seung Hwan Hwang
- R&D Center, Huons Co., Ltd., 55 Hanyangdaehak-ro, Ansan 15588, Republic of Korea
| | - Yi-Sook Jung
- College of Pharmacy, Ajou University, Suwon 16499, Republic of Korea
- Research Institute of Pharmaceutical Sciences and Technology, Ajou University, Suwon 16499, Republic of Korea
- Correspondence: ; Tel.: +82-31-219-3444
| |
Collapse
|
35
|
Liang Y, Wang L. Carthamus tinctorius L.: A natural neuroprotective source for anti-Alzheimer's disease drugs. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115656. [PMID: 36041691 DOI: 10.1016/j.jep.2022.115656] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 07/24/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alzheimer's disease (AD) is a multicausal neurodegenerative disease clinically characterized by generalized dementia. The pathogenic process of AD not only is progressive and complex but also involves multiple factors and mechanisms, including β-amyloid (Aβ) aggregation, tau protein hyperphosphorylation, oxidative stress, and neuroinflammation. As the first-line treatment for AD, cholinesterase inhibitors can, to a certain extent, relieve AD symptoms and delay AD progression. Nonetheless, the current treatment strategies for AD are far from meeting clinical expectations, and more options for AD treatment should be applied in clinical practice. AIM OF THE REVIEW The aim of this review was to investigate published reports of C. tinctorius L. and its active constituents in AD treatment through a literature review. MATERIALS AND METHODS Information was retrieved from scientific databases including Web of Science, ScienceDirect, Scopus, Google Scholar, Chemical Abstracts Services and books, PubMed, dissertations and technical reports. Keywords used for the search engines were "Honghua" or "Carthamus tinctorius L." or "safflower" in conjunction with "(native weeds OR alien invasive)"AND "Chinese herbal medicine". RESULTS A total of 47 literatures about C. tinctorius L. and its active constituents in AD treatment through signaling pathways, immune cells, and disease-related mediators and systematically elucidates potential mechanisms from the point of anti-Aβ aggregation, suppressing tau protein hyperphosphorylation, increasing cholinergic neurotransmitters levels, inhibiting oxidative stress, anti-neuroinflammation, ameliorating synaptic plasticity, and anti-apoptosis. CONCLUSIONS Chinese herbal medicine (CHM) is a treasure endowed by nature to mankind. Emerging studies have confirmed that CHM and its active constituents play a positive role in AD treatment. Carthamus tinctorius L., the most commonly used CHM, can be used with medicine and food, with the effect of activating blood circulation and eliminating blood stasis. In the paper, we have concluded that the existing therapeutic mechanisms of C. tinctorius L. and summarized the potential mechanisms of C. tinctorius L. and its active constituents in AD treatment through a literature review.
Collapse
Affiliation(s)
- Yuanyuan Liang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
36
|
Ojea Ramos S, Feld M, Fustiñana MS. Contributions of extracellular-signal regulated kinase 1/2 activity to the memory trace. Front Mol Neurosci 2022; 15:988790. [PMID: 36277495 PMCID: PMC9580372 DOI: 10.3389/fnmol.2022.988790] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/02/2022] [Indexed: 11/15/2022] Open
Abstract
The ability to learn from experience and consequently adapt our behavior is one of the most fundamental capacities enabled by complex and plastic nervous systems. Next to cellular and systems-level changes, learning and memory formation crucially depends on molecular signaling mechanisms. In particular, the extracellular-signal regulated kinase 1/2 (ERK), historically studied in the context of tumor growth and proliferation, has been shown to affect synaptic transmission, regulation of neuronal gene expression and protein synthesis leading to structural synaptic changes. However, to what extent the effects of ERK are specifically related to memory formation and stabilization, or merely the result of general neuronal activation, remains unknown. Here, we review the signals leading to ERK activation in the nervous system, the subcellular ERK targets associated with learning-related plasticity, and how neurons with activated ERK signaling may contribute to the formation of the memory trace.
Collapse
Affiliation(s)
- Santiago Ojea Ramos
- Instituto de Fisiología, Biología Molecular y Neurociencias, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Mariana Feld
- Instituto de Fisiología, Biología Molecular y Neurociencias, Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | | |
Collapse
|
37
|
Mohammed RA, Sayed RH, El-Sahar AE, Khattab MA, Saad MA. Insights into the role of pERK1/2 signaling in post-cerebral ischemia reperfusion sexual dysfunction in rats. Eur J Pharmacol 2022; 933:175258. [PMID: 36096157 DOI: 10.1016/j.ejphar.2022.175258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/15/2022] [Accepted: 09/05/2022] [Indexed: 11/15/2022]
Abstract
The purpose of the present study was to investigate the effects of ERK1/2 inhibition on both the amygdala and hippocampal structures, and to investigate its role in regulating memory for sexual information. This study utilized a cerebral ischemia reperfusion (IR) model to produce a stressful brain condition that highlights the possible involvement of a hippocampal GC/pERK1/2/BDNF pathway in the resulting sexual consequences of this ailment. Male Wistar rats were divided into four groups: (1) sham; (2) IR: subjected to 45 min of ischemia followed by 48 h of reperfusion; (3) PD98059: received PD98059 at 0.3 mg/kg, i.p.; (4) IR + PD98059. This study provides new evidence for cerebral IR-induced amygdala injury and the sexual impairments that are associated with motor and cognitive deficits in rats. These findings were correlated with histopathological changes that are defined by extensive neuronal loss in both the hippocampus and the amygdala. The current study postulated that the ERK inhibitor PD98059 could reverse IR-induced injury in the amygdala as well as reversing IR-induced sexual impairments. This hypothesis is supported by the ability of PD98059 to: (1) restore luteinizing hormone and testosterone levels; (2) increase sexual arousal and copulatory performance (as evidenced by modulating mount, intromission, ejaculation latencies, and post-ejaculatory intervals); (3) improve the histological profile in the amygdala that is associated with reduced glutamate levels, c-Fos expression, and elevated gamma aminobutyric acid levels. In conclusion, the present findings introduce pERK1/2 inhibition as a possible strategy for enhancing sexual activity in survivors of IR.
Collapse
Affiliation(s)
- Reham A Mohammed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Ayman E El-Sahar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mohamed A Khattab
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Muhammed A Saad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, Ajman, 4184, United Arab Emirates
| |
Collapse
|
38
|
Reich N, Hölscher C. The neuroprotective effects of glucagon-like peptide 1 in Alzheimer's and Parkinson's disease: An in-depth review. Front Neurosci 2022; 16:970925. [PMID: 36117625 PMCID: PMC9475012 DOI: 10.3389/fnins.2022.970925] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 12/16/2022] Open
Abstract
Currently, there is no disease-modifying treatment available for Alzheimer's and Parkinson's disease (AD and PD) and that includes the highly controversial approval of the Aβ-targeting antibody aducanumab for the treatment of AD. Hence, there is still an unmet need for a neuroprotective drug treatment in both AD and PD. Type 2 diabetes is a risk factor for both AD and PD. Glucagon-like peptide 1 (GLP-1) is a peptide hormone and growth factor that has shown neuroprotective effects in preclinical studies, and the success of GLP-1 mimetics in phase II clinical trials in AD and PD has raised new hope. GLP-1 mimetics are currently on the market as treatments for type 2 diabetes. GLP-1 analogs are safe, well tolerated, resistant to desensitization and well characterized in the clinic. Herein, we review the existing evidence and illustrate the neuroprotective pathways that are induced following GLP-1R activation in neurons, microglia and astrocytes. The latter include synaptic protection, improvements in cognition, learning and motor function, amyloid pathology-ameliorating properties (Aβ, Tau, and α-synuclein), the suppression of Ca2+ deregulation and ER stress, potent anti-inflammatory effects, the blockage of oxidative stress, mitochondrial dysfunction and apoptosis pathways, enhancements in the neuronal insulin sensitivity and energy metabolism, functional improvements in autophagy and mitophagy, elevated BDNF and glial cell line-derived neurotrophic factor (GDNF) synthesis as well as neurogenesis. The many beneficial features of GLP-1R and GLP-1/GIPR dual agonists encourage the development of novel drug treatments for AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical and Life Sciences Division, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, Second Associated Hospital, Shanxi Medical University, Taiyuan, China
- Henan University of Chinese Medicine, Academy of Chinese Medical Science, Zhengzhou, China
| |
Collapse
|
39
|
Riyahi J, Abdoli B, Gelfo F, Petrosini L, Khatami L, Meftahi GH, Haghparast A. Multigenerational effects of paternal spatial training are lasting in the F1 and F2 male offspring. Behav Pharmacol 2022; 33:342-354. [PMID: 35502983 DOI: 10.1097/fbp.0000000000000682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Recent studies on intergenerational transmission of learning and memory performances demonstrated that parental spatial training before fertilization could facilitate learning and memory in the offspring, but many questions remain unclarified. Essential issues regarding whether and how long the effects of parental training in a task can last in several generations, and whether learning a task repeated in the successive generations can enhance a load of multigenerational effects. In the present study, the spatial performances of F1 and F2 generations of male offspring of fathers or grandfathers spatially trained in the Morris Water Maze were evaluated and compared with the performance of a control sample matched for age and sex. Further, to investigate the memory process in F1 and F2 male offspring, brain-derived neurotrophic factor (BDNF), p-ERK1/2 and acetylated histone 3 lysine 14 (H3K14) expression levels in the hippocampus were analyzed. The findings showed that paternal training reduced escape latencies and increased time spent in the target quadrant by F1 and F2 male offspring. Besides, paternal spatial training repeated in two generations did not enhance the beneficial effects on offspring's spatial performances. These findings were supported by neurobiologic data showing that paternal training increased BDNF and p-ERK1/2 in the hippocampus of F1 and F2 male offspring. Furthermore, the hippocampal level of acetylated H3K14 increased in the offspring of spatially trained fathers, reinforcing the hypothesis that the augmented histone acetylation might play an essential role in the inheritance of spatial competence.
Collapse
Affiliation(s)
- Javad Riyahi
- Neuroscience Research Center, Baqiyatallah University of Medical Sciences
| | - Behrouz Abdoli
- Department of Cognitive and Behavioral Science and Technology in Sport, Faculty of Sport Sciences and Health, Shahid Beheshti University, Tehran, Iran
| | - Francesca Gelfo
- IRCCS Santa Lucia Foundation
- Department of Human Sciences, Guglielmo Marconi University, Rome, Italy
| | | | - Leila Khatami
- School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical sciences, Tehran, Iran
| |
Collapse
|
40
|
Geng YN, Zhao M, Yang JL, Cheng X, Han Y, Wang CB, Jiang XF, Fan M, Zhu LL. GP-14 protects against severe hypoxia-induced neuronal injury through the AKT and ERK pathways and its induced transcriptome profiling alteration. Toxicol Appl Pharmacol 2022; 448:116092. [PMID: 35654276 DOI: 10.1016/j.taap.2022.116092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
Abstract
Gypenosides are major bioactive ingredients of G. pentaphyllum. In our previous study, we found that gypenosides had neuroprotective effects against hypoxia-induced injury. In the current study, we focused on the protective effects of gypenoside-14 (GP-14), which is one of the newly identified bioactive components, on neuronal injury caused by severe hypoxia (0.3% O2). The results showed that GP-14 pretreatment alleviated the cell viability damage and apoptosis induced by hypoxia in PC12 cells. Moreover, GP-14 pretreatment also attenuated primary neuron injuries under hypoxic conditions. Additionally, GP-14 pretreatment significantly ameliorated neuronal damage in the hippocampal region induced by high-altitude cerebral edema (HACE). At the molecular level, GP-14 pretreatment reversed the decreased activities of the AKT and ERK signaling pathways caused by hypoxia in PC12 cells and primary neurons. To comprehensively explore the possible mechanisms, transcriptome sequencing was conducted, and these results indicated that GP-14 could alter the transcriptional profiles of primary neuron. Taken together, our results suggest that GP-14 acts as a neuroprotective agent to protect against neuronal damage induced by severe hypoxia and it is a promising compound for the development of neuroprotective drugs.
Collapse
Affiliation(s)
- Ya-Nan Geng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ming Zhao
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Jun-Li Yang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences (CAS), Lanzhou 730000, China
| | - Xiang Cheng
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ying Han
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Cheng-Bo Wang
- CAS Key Laboratory of Chemistry of Northwestern Plant Resources and Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences (CAS), Lanzhou 730000, China
| | - Xiu-Fang Jiang
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China
| | - Ming Fan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing 100069, China; Beijing Institute of Basic Medical Sciences, Beijing 100850, China; School of information Science & Engineering, Lanzhou University, Lanzhou 730000, China.
| | - Ling-Ling Zhu
- Beijing Institute of Basic Medical Sciences, Beijing 100850, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China; College of Life Sciences, Anhui Medical University, Hefei 230032, China; Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
41
|
Ramalingam M, Jeong HS, Hwang J, Cho HH, Kim BC, Kim E, Jang S. Autophagy Signaling by Neural-Induced Human Adipose Tissue-Derived Stem Cell-Conditioned Medium during Rotenone-Induced Toxicity in SH-SY5Y Cells. Int J Mol Sci 2022; 23:ijms23084193. [PMID: 35457010 PMCID: PMC9031864 DOI: 10.3390/ijms23084193] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/07/2022] [Accepted: 04/07/2022] [Indexed: 12/04/2022] Open
Abstract
Rotenone (ROT) inhibits mitochondrial complex I, leading to reactive oxygen species formation, which causes neurodegeneration and alpha-synuclein (α-syn) aggregation and, consequently, Parkinson’s disease. We previously found that a neurogenic differentiated human adipose tissue-derived stem cell-conditioned medium (NI-hADSC-CM) was protective against ROT-induced toxicity in SH-SY5Y cells. In the present study, ROT significantly decreased the phospho (p)-mTORC1/total (t)-mTOR, p-mTORC2/t-mTOR, and p-/t-ULK1 ratios and the ATG13 level by increasing the DEPTOR level and p-/t-AMPK ratio. Moreover, ROT increased the p-/t-Akt ratio and glycogen synthase kinase-3β (GSK3β) activity by decreasing the p-/t-ERK1/2 ratios and beclin-1 level. ROT also promoted the lipidation of LC3B-I to LC3B-II by inducing autophagosome formation in Triton X-100-soluble and -insoluble cell lysate fractions. Additionally, the levels of ATG3, 5, 7, and 12 were decreased, along with those of lysosomal LAMP1, LAMP2, and TFEB, leading to lysosomal dysfunction. However, NI-hADSC-CM treatment increased the p-mTORC1, p-mTORC2, p-ULK1, p-Akt, p-ERK1/2, ATG13, and beclin-1 levels and decreased the p-AMPK level and GSK3β activity in response to ROT-induced toxicity. Additionally, NI-hADSC-CM restored the LC3B-I level, increased the p62 level, and normalized the ATG and lysosomal protein amounts to control levels. Autophagy array revealed that the secreted proteins in NI-hADSC-CM could be crucial in the neuroprotection. Taken together, our results showed that the neuroprotective effects of NI-hADSC-CM on the autophagy signaling pathways could alleviate the aggregation of α-syn in Parkinson’s disease and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea; (H.-S.J.); (J.H.)
- Correspondence: (M.R.); (S.J.)
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea; (H.-S.J.); (J.H.)
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea; (H.-S.J.); (J.H.)
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Korea;
| | - Byeong C. Kim
- Department of Neurology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Korea;
| | - Eungpil Kim
- Jeonnam Biopharmaceutical Research Center, Hwasun 58141, Korea;
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Hwasun 58128, Korea; (H.-S.J.); (J.H.)
- Correspondence: (M.R.); (S.J.)
| |
Collapse
|
42
|
Yan J, Zhang F, Le Niu, Wang X, Lu X, Ma C, Zhang C, Song J, Zhang Z. High-frequency repetitive transcranial magnetic stimulation mitigates depression-like behaviors in CUMS-induced rats via FGF2/FGFR1/p-ERK signaling pathway. Brain Res Bull 2022; 183:94-103. [PMID: 35247488 DOI: 10.1016/j.brainresbull.2022.02.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 12/11/2022]
Abstract
High-frequency repetitive transcranial magnetic stimulation (rTMS) is a widely used and effective biological treatment for depression. Although previous studies have shown that astrocyte function may be modified by rTMS, the specific neurobiological mechanisms underlying its antidepressant action are not clear. Substantial evidence has accumulated indicating that neurotrophin dysfunction and neuronal apoptosis play a role in the development of depression. To evaluate this hypothesis, we applied a chronical unpredictable mild stress (CUMS) protocol to induce depression-like behaviors in rats, followed by the delivery of 10-Hz rTMS for 3 weeks. Behavioral outcome measures consisted of a sucrose preference test, forced swimming test, and open field test. Histological analysis focused on apoptosis, expression of GFAP and FGF2, and FGF2 pathway-related proteins. The results showed that after rTMS treatment, the rats' sucrose preference increased, open field performance improved while the immobility time of forced swimming decreased. The behavioral changes seen in rTMS treated rats were accompanied by marked reductions in the number of TUNEL-positive neural cells and the level of expression of BAX and by an increase in Bcl2. Furthermore, the expression of GFAP and FGF2 was increased, along with activation of FGF2 downstream pathway. These results suggest that rTMS treatment can improve depression-like behavior, attenuate neural apoptosis, and reverse reduction of astrocytes in a rat model of depression. We hypothesize that the therapeutic action of rTMS in CUMS-induced rats is linked to the activation of the FGF2/FGFR1/p-ERK signaling pathway.
Collapse
Affiliation(s)
- Junni Yan
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Henan Key Lab of Biological Psychiatry, Xinxiang, Henan 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, Xinxiang, Henan 453002, China
| | - Fuping Zhang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Henan Key Lab of Biological Psychiatry, Xinxiang, Henan 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, Xinxiang, Henan 453002, China
| | - Le Niu
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihui, Henan 453100, China; The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Henan Key Lab of Biological Psychiatry, Xinxiang, Henan 453002, China
| | - Xiaonan Wang
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Henan Key Lab of Biological Psychiatry, Xinxiang, Henan 453002, China
| | - Xinxin Lu
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Henan Key Lab of Biological Psychiatry, Xinxiang, Henan 453002, China
| | - Chaoyue Ma
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihui, Henan 453100, China; The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Henan Key Lab of Biological Psychiatry, Xinxiang, Henan 453002, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Clinical Neuroscience Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Research Center for Brain Science and Brain-Inspired technology, Shanghai, China
| | - Jinggui Song
- The Second Affiliated Hospital of Xinxiang Medical University (Henan Mental Hospital), Henan Key Lab of Biological Psychiatry, Xinxiang, Henan 453002, China; Henan Engineering Research Center of Physical Diagnostics and Treatment Technology for the Mental and Neurological Diseases, Xinxiang, Henan 453002, China.
| | - Zhaohui Zhang
- The First Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Neurorestoratology, Weihui, Henan 453100, China.
| |
Collapse
|
43
|
Proteomic analysis of hypothalamus in prepubertal and pubertal female goat. J Proteomics 2022; 251:104411. [PMID: 34728423 DOI: 10.1016/j.jprot.2021.104411] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/28/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022]
Abstract
The functions of proteins at the onset of puberty in goats remain largely unexplored. To identify the proteins regulating puberty in goats, we analysed protein abundance and pathways in the hypothalamus of female goats. We applied tandem mass tag (TMT) labelling, liquid chromatography-tandem mass spectrometry (LC-MS/MS), and parallel reaction monitoring (PRM) to examine hypothalamus of pubertal (cases; n = 3) and prepubertal (controls; n = 3) goats. We identified 5119 proteins, including 69 differentially abundant proteins (DAPs), of which 35 were upregulated and 34 were downregulated. Fourteen DAPs were randomly selected to verify these results using PRM, and the results were consistent with the TMT quantitative results. DAPs were enriched in MAPK signalling pathway, Ras signalling pathway, Autophagy-animal, Endocytosis, and PI3K/Akt/mTOR signalling pathway categories. These pathways are related to embryogenesis, cell proliferation, cell differentiation, and promoting the release of gonadotropin-releasing hormone (GnRH) in the hypothalamus. In particular, PDGFRβ and MAP3K7 occupied important locations in the protein-protein interaction network. The results demonstrate that DAPs and their related signalling pathways are crucial in regulating puberty in goats. However, further research is needed to explore the functions of DAPs and their pathways to provide new insights into the mechanism of puberty onset. SIGNIFICANCE: In domestic animals, reaching the age of puberty is an event that contributes significantly to lifetime reproductive potential. And the hypothalamus functions directly in the complex systemic changes that control puberty. Our study was the first TMT proteomics analysis on hypothalamus tissues of pubertal goats, which revealed the changes of protein and pathways that are related to the onset of puberty. We identified 69 DAPs, which were enriched in the MAPK signaling pathway, the Ras signaling pathway, and the IGF-1/PI3K/Akt/mTOR pathway, suggesting that these processes were probably involved in the onset of puberty.
Collapse
|
44
|
Faucher P, Huguet C, Mons N, Micheau J. Acute pre-learning stress selectively impairs hippocampus-dependent fear memory consolidation: Behavioral and molecular evidence. Neurobiol Learn Mem 2022; 188:107585. [PMID: 35021061 DOI: 10.1016/j.nlm.2022.107585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/22/2021] [Accepted: 01/04/2022] [Indexed: 11/30/2022]
Abstract
Despite compelling evidence that stress or stress-related hormones influence fear memory consolidation processes, the understanding of molecular mechanisms underlying the effects of stress is still fragmentary. The release of corticosterone in response to pre-learning stress exposure has been demonstrated to modulate positively or negatively memory encoding and/or consolidation according to many variables such as stress intensity, the emotional valence of the learned material or the interval between stressful episode and learning experience. Here, we report that contextual but not cued fear memory consolidation was selectively impaired in male mice exposed to a 50 min-period of restraint stress just before the unpaired fear conditioning session. In addition to behavioral impairment, acute stress down-regulated activated/phosphorylated ERK1/2 (pERK1/2) in dorsal hippocampal area CA1 in mice sacrificed 60 min and 9 h after unpaired conditioning. In lateral amygdala, although acute stress by itself increased the level of pERK1/2 it nevertheless blocked the peak of pERK1/2 that was normally observed 15 min after unpaired conditioning. To examine whether stress-induced corticosterone overflow was responsible of these detrimental effects, the corticosterone synthesis inhibitor, metyrapone, was administered 30 min before stress exposure. Metyrapone abrogated the stress-induced contextual fear memory deficits but did not alleviate the effects of stress on pERK1/2 and its downstream target phosphorylated CREB (pCREB) in hippocampus CA1 and lateral amygdala. Collectively, our observations suggest that consolidation of hippocampus-dependent memory and the associated signaling pathway are particularly sensitive to stress. However, behavioral normalization by preventive metyrapone treatment was not accompanied by renormalization of the canonical signaling pathway. A new avenue would be to consider surrogate mechanisms involving proper metyrapone influence on both nongenomic and genomic actions of glucocorticoid receptors.
Collapse
Affiliation(s)
- Pierre Faucher
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Célia Huguet
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Nicole Mons
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France
| | - Jacques Micheau
- Université de Bordeaux, CNRS, INCIA, UMR 5287, F-33000 Bordeaux, France.
| |
Collapse
|
45
|
Effects of docosanyl ferulate, a constituent of Withania somnifera, on ethanol- and morphine-elicited conditioned place preference and ERK phosphorylation in the accumbens shell of CD1 mice. Psychopharmacology (Berl) 2022; 239:795-806. [PMID: 35088095 PMCID: PMC8891193 DOI: 10.1007/s00213-022-06069-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 01/17/2022] [Indexed: 10/27/2022]
Abstract
BACKGROUND Docosanyl ferulate (DF) is a behaviourally active GABAA receptor complex (GABAAR) agonist, recently isolated from the standardized methanolic extract of Withania somnifera Dunal (WSE) root. Previous studies have shown that WSE prevents both ethanol- and morphine-dependent acquisition and expression of conditioned place preference (CPP) and stimulation of dopamine release in the nucleus accumbens shell (AcbSh). AIMS The study aimed at determining (a) whether DF contributes to WSE's ability to affect the acquisition and expression of ethanol- and morphine-elicited CPP and, given that phosphorylation of extracellular signal-regulated kinase (pERK) in the AcbSh is involved in associative learning and motivated behaviours, (b) whether WSE and DF may affect ethanol- and morphine-induced ERKs phosphorylation in the AcbSh. METHODS In adult male CD1 mice, DF's effects on the acquisition and expression of ethanol- and morphine-elicited CPP were evaluated by a classical place conditioning paradigm, whereas the effects of WSE and DF on ethanol- and morphine-elicited pERK in the AcbSh were evaluated by immunohistochemistry. RESULTS AND CONCLUSIONS The study shows that DF, differently from WSE, affects only the acquisition but not the expression of ethanol- and morphine-induced CPP. Moreover, the study shows that both WSE and DF can prevent ethanol- and morphine-elicited pERK expression in the AcbSh. Overall, these results highlight subtle but critical differences for the role of GABAARs in the mechanism by which WSE affects these ethanol- and morphine-dependent behavioural and molecular/cellular responses and support the suggestion of WSE and DF for the control of different components of drug addiction.
Collapse
|
46
|
Banhasasim-Tang Ameliorates Spatial Memory by Suppressing Oxidative Stress through Regulation of ERK/p38 Signaling in Hippocampus of Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6970578. [PMID: 34900088 PMCID: PMC8660254 DOI: 10.1155/2021/6970578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/26/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022]
Abstract
Since ancient times, Banhasasim-tang (BHS) has been used to treat functional dyspepsia in East Asia. Here, we aimed to determine the protective action of BHS on hippocampal neurons against oxidative stress. We investigated the functional effect of BHS on a scopolamine-induced mouse model, and molecular analysis was performed in glutamate-induced HT22 cells. We observed that BHS administration ameliorated memory dysfunction in scopolamine-treated mice. BHS administration also increased neuronal survival and acetylcholine activity and phosphorylation of extracellular signal-regulated kinase (ERK) and cAMP response element-binding protein (CREB) in the hippocampus of mice. In hippocampal cells, BHS treatment rescued glutamate-induced cytotoxicity, apoptosis, and oxidative stress. We observed an increase of HO-1 and a decrease of Nrf2 protein expression in glutamate-induced oxidative stress; however, the expression level of these proteins was significantly rescued by BHS treatment. BHS treatment also regulated phosphorylation of p38, p53, ERK, and CREB. Therefore, our data indicated that BHS may reduce oxidative stress through regulation of ERK-CREB and p38-p53 signaling in the hippocampus, resulting in decreased neuronal damage and improved memory in rodent models of neurodegenerative disease.
Collapse
|
47
|
Shi L, Li J, Liang XF, He S, Dou Y, Peng J, Cai W, Liang H. Memory regulation in feeding habit transformation to dead prey fish of Chinese perch (Siniperca chuatsi). FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:1893-1907. [PMID: 34581919 DOI: 10.1007/s10695-021-01001-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/10/2021] [Indexed: 06/13/2023]
Abstract
Memory drove a critical process of feeding habit transformation in Chinese perch when they re-trained to eat dead prey fish. To investigate the regulatory mechanism of cAMP-response element-binding protein (CREB) signaling pathway on the memory of Chinese perch during feeding habit transformation, the phosphorylation levels of upstream signal proteins of CREB between the control group (trained once) and the experimental group (trained twice) were measured. The results illustrated that the re-training was correlated to phosphorylation of extracellular regulated protein kinase (ERK1/2) and calcium/calmodulin-dependent protein kinase II (CaMKII), and dephosphorylation of protein kinase A (PKA) of Chinese perch. Inhibition of ERK1/2-CREB pathway decreased the mRNA levels of memory-related genes ((fos-related antigen 2 (fra2), CCAAT enhancer-binding protein delta (c/ebpb), immediate-early gene zif268 (zif268), proto-oncogenes c-fos (c-fox) and synaptotagmin-IV (sytIV)) and mRNA levels of appetite-related genes (agouti-related peptide (agrp) and ghrelin), and activation of PP1-CREB pathway increased the phosphorylated levels of CREB, the mRNA levels of memory-related genes (fra2, c/ebpb, zif268, and c-fox), and the mRNA levels of appetite-related genes (pro-opiomelanocortin (pomc) and leptin) in primary brain cells of Chinese perch. The memory in Chinese perch feeding habit transformation was associated with the ERK1/2-CREB and PP1-CREB pathways, which could regulate the transcription of memory-related genes and appetite-related genes.
Collapse
Affiliation(s)
- Linjie Shi
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan, 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Jiao Li
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan, 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Xu-Fang Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan, 430070, China.
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China.
| | - Shan He
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan, 430070, China.
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China.
| | - Yaqi Dou
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan, 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Jian Peng
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan, 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Wenjing Cai
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan, 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| | - Hui Liang
- College of Fisheries, Chinese Perch Research Center, Huazhong Agricultural University, Wuhan, 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan, 430070, China
| |
Collapse
|
48
|
Cardoso FDS, Mansur FCB, Lopes-Martins RÁB, Gonzalez-Lima F, Gomes da Silva S. Transcranial Laser Photobiomodulation Improves Intracellular Signaling Linked to Cell Survival, Memory and Glucose Metabolism in the Aged Brain: A Preliminary Study. Front Cell Neurosci 2021; 15:683127. [PMID: 34539346 PMCID: PMC8446546 DOI: 10.3389/fncel.2021.683127] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 08/11/2021] [Indexed: 11/16/2022] Open
Abstract
Aging is often accompanied by exacerbated activation of cell death-related signaling pathways and decreased energy metabolism. We hypothesized that transcranial near-infrared laser may increase intracellular signaling pathways beneficial to aging brains, such as those that regulate brain cell proliferation, apoptosis, and energy metabolism. To test this hypothesis, we investigated the expression and activation of intracellular signaling proteins in the cerebral cortex and hippocampus of aged rats (20 months old) treated with the transcranial near-infrared laser for 58 consecutive days. As compared to sham controls, transcranial laser treatment increased intracellular signaling proteins related to cell proliferation and cell survival, such as signal transducer and activator of transcription 3 (STAT3), extracellular signal-regulated protein kinase (ERK), c-Jun N-terminal kinase (JNK), p70 ribosomal protein S6 kinase (p70S6K) and protein kinase B (PKB), also known as Akt that is linked to glucose metabolism. In addition, ERK is linked to memory, while ERK and JNK signaling pathways regulate glucose metabolism. Specifically, the laser treatment caused the activation of STAT3, ERK, and JNK signaling proteins in the cerebral cortex. In the hippocampus, the laser treatment increased the expression of p70S6K and STAT3 and the activation of Akt. Taken together, the data support the hypothesis that transcranial laser photobiomodulation improves intracellular signaling pathways linked to cell survival, memory, and glucose metabolism in the brain of aged rats.
Collapse
Affiliation(s)
- Fabrízio Dos Santos Cardoso
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Mogi das Cruzes, Brazil.,Department of Psychology and Institute for Neuroscience, University of Texas at Austin, Austin, TX, United States
| | | | | | - Francisco Gonzalez-Lima
- Department of Psychology and Institute for Neuroscience, University of Texas at Austin, Austin, TX, United States
| | - Sérgio Gomes da Silva
- Núcleo de Pesquisas Tecnológicas, Universidade de Mogi das Cruzes, Mogi das Cruzes, Brazil.,Centro Universitário UNIFAMINAS (UNIFAMINAS), Muriaé, Brazil.,Hospital do Câncer de Muriaé, Fundação Cristiano Varella (FCV), Muriaé, Brazil
| |
Collapse
|
49
|
Navarro-Lobato I, Masmudi-Martín M, López-Aranda MF, Quiros-Ortega ME, Carretero-Rey M, Garcia-Garrido MF, Gallardo-Martínez C, Martín-Montañez E, Gaona-Romero C, Delgado G, Torres-Garcia L, Terrón-Melguizo J, Posadas S, Muñoz LR, Rios CV, Zoidakis J, Vlahou A, López JC, Khan ZU. RGS14414-Mediated Activation of the 14-3-3ζ in Rodent Perirhinal Cortex Induces Dendritic Arborization, an Increase in Spine Number, Long-Lasting Memory Enhancement, and the Prevention of Memory Deficits. Cereb Cortex 2021; 32:1894-1910. [PMID: 34519346 DOI: 10.1093/cercor/bhab322] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The remedy of memory deficits has been inadequate, as all potential candidates studied thus far have shown limited to no effects and a search for an effective strategy is ongoing. Here, we show that an expression of RGS14414 in rat perirhinal cortex (PRh) produced long-lasting object recognition memory (ORM) enhancement and that this effect was mediated through the upregulation of 14-3-3ζ, which caused a boost in BDNF protein levels and increase in pyramidal neuron dendritic arborization and dendritic spine number. A knockdown of the 14-3-3ζ gene in rat or the deletion of the BDNF gene in mice caused complete loss in ORM enhancement and increase in BDNF protein levels and neuronal plasticity, indicating that 14-3-3ζ-BDNF pathway-mediated structural plasticity is an essential step in RGS14414-induced memory enhancement. We further observed that RGS14414 treatment was able to prevent deficits in recognition, spatial, and temporal memory, which are types of memory that are particularly affected in patients with memory dysfunctions, in rodent models of aging and Alzheimer's disease. These results suggest that 14-3-3ζ-BDNF pathway might play an important role in the maintenance of the synaptic structures in PRh that support memory functions and that RGS14414-mediated activation of this pathway could serve as a remedy to treat memory deficits.
Collapse
Affiliation(s)
- Irene Navarro-Lobato
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Mariam Masmudi-Martín
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Manuel F López-Aranda
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - María E Quiros-Ortega
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Marta Carretero-Rey
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - María F Garcia-Garrido
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Carmen Gallardo-Martínez
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Elisa Martín-Montañez
- Department of Pharmacology, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Celia Gaona-Romero
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Gloria Delgado
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Laura Torres-Garcia
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Javier Terrón-Melguizo
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Sinforiano Posadas
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Lourdes Rodríguez Muñoz
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Carlos Vivar Rios
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain
| | - Jerome Zoidakis
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Juan C López
- Animal Behavior and Neuroscience Lab., Department of Experimental Psychology, Faculty of Psychology, University of Seville, Seville 41018, Spain
| | - Zafar U Khan
- Laboratory of Neurobiology, CIMES, University of Malaga, Malaga 29010, Spain.,Department of Medicine, Faculty of Medicine, University of Malaga, Malaga 29010, Spain.,CIBERNED, Institute of Health Carlos III, Madrid 28031, Spain
| |
Collapse
|
50
|
Gomes KC, Lima FWB, da Silva Aguiar HQ, de Araújo SS, de Cordova CAS, de Cordova FM. Thiamine deficiency and recovery: impact of recurrent episodes and beneficial effect of treatment with Trolox and dimethyl sulfoxide. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:2289-2307. [PMID: 34468817 DOI: 10.1007/s00210-021-02148-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
At present, thiamine deficiency (TD) is managed with administration of high doses of thiamine. Even so, severe and permanent neurological disorders can occur in recurrent episodes of TD. In this study, we used a murine model to assess the efficacy of TD recovery treatments using thiamine with or without additional administration of the antioxidant Trolox or the anti-inflammatory dimethyl sulfoxide (DMSO) after a single or recurrent episode of TD. TD was induced for 9 days with deficient chow and pyrithiamine, and the recovery period was 7 days with standard amounts of chow and thiamine, Trolox, and/or DMSO. After these periods, we evaluated behavior, histopathology, and ERK1/2 modulation in the brain. Deficient animals showed reductions in locomotor activity, motor coordination, and spatial memory. Morphologically, after a single episode of TD and recovery, deficient mice showed neuronal vacuolization in the dorsal thalamus and, after two episodes, a reduction in neuronal cell number. These effects were attenuated or reversed by the recovery treatments, mainly in the treatments with thiamine associated with Trolox or DMSO. Deficient animals showed a strong increase in ERK1/2 phosphorylation in the thalamus, hippocampus, and cerebral cortex after one deficiency episode and recovery. Interestingly, after recurrent TD and recovery, ERK1/2 phosphorylation remained high only in the deficient mice treated with thiamine and/or Trolox or thiamine with DMSO. Our data suggest that a protocol for TD treatment with thiamine in conjunction with Trolox or DMSO enhances the recovery of animals and possibly minimizes the late neurological sequelae.
Collapse
Affiliation(s)
- Ketren Carvalho Gomes
- Programa de Pós-Graduação em Sanidade Animal e Saúde Pública nos Trópicos , Universidade Federal do Tocantins, BR-153, km 112, Araguaína, TO, 77804-970, Brazil
| | | | - Helen Quézia da Silva Aguiar
- Curso de Medicina Veterinária, Universidade Federal do Tocantins, BR-153, km 112, Araguaína, TO, 77804-970, Brazil
| | - Suiane Silva de Araújo
- Curso de Medicina Veterinária, Universidade Federal do Tocantins, BR-153, km 112, Araguaína, TO, 77804-970, Brazil
| | | | - Fabiano Mendes de Cordova
- Programa de Pós-Graduação em Sanidade Animal e Saúde Pública nos Trópicos , Universidade Federal do Tocantins, BR-153, km 112, Araguaína, TO, 77804-970, Brazil.
| |
Collapse
|