1
|
Martínez Sánchez I, Spielbauer J, Diaz Heijtz R. Maternal peptidoglycan overexposure during late pregnancy alters neurodevelopment and behavior in juvenile offspring. Brain Behav Immun 2025; 127:96-102. [PMID: 40058669 DOI: 10.1016/j.bbi.2025.03.014] [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: 01/13/2025] [Revised: 02/19/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Emerging evidence suggests that maternal gut microbiota-derived metabolites and components influence fetal brain development and subsequent neurodevelopment. This study investigates the effects of maternal overexposure to muramyl dipeptide (MDP)-a bacterial peptidoglycan (PGN) motif recognized by Nod2 receptors-on offspring neurodevelopment and behavior. Time-mated C57BL/6J female mice received MDP via drinking water from gestational days 16-19. Nod2 activation in amniotic fluid was assessed using a Nod2 cell-based reporter assay, showing a significant increase in males 24 h after MDP exposure. Gene expression analysis revealed upregulation of PGN transporters in fetal brains, with males showing higher levels of Slc15a1/PepT1, Slc15a2/PepT2, and Slc46a2. No changes in inflammatory or microglia-related markers were found. Behavioral assessments during the juvenile period revealed sex-specific effects: prenatally exposed males showed reduced social interaction, while females exhibited reduced novelty-induced locomotion and impaired social recognition. These behavioral changes were linked to altered expression of synaptic (Dlg4, Ppp1r9b, Darpp-32) and microglial (Trem-2, Cx3cr1) genes in the prefrontal cortex. Our findings underscore the sex-specific effects of maternal PGN overexposure on offspring neurodevelopment, highlighting the potential role of the maternal microbiome in the neurobiology of neurodevelopmental disorders, even in the absence of infection or robust inflammation.
Collapse
Affiliation(s)
| | - Julia Spielbauer
- Department of Neuroscience, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | |
Collapse
|
2
|
Erkizia-Santamaría I, Horrillo I, Martínez-Álvarez N, Pérez-Martínez D, Rivero G, Erdozain AM, Meana JJ, Ortega JE. Evaluation of behavioural and neurochemical effects of psilocybin in mice subjected to chronic unpredictable mild stress. Transl Psychiatry 2025; 15:201. [PMID: 40517150 PMCID: PMC12167372 DOI: 10.1038/s41398-025-03421-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/16/2025] [Accepted: 06/04/2025] [Indexed: 06/16/2025] Open
Abstract
Depression and anxiety are disabling and high incidence mental disorders characterized by phenotypic heterogeneity. Currently available treatments show severe limitations. Thus, there is an urgent need for effective treatments in this population. In the search for novel rapid-acting antidepressants, the psychedelic psilocybin has emerged as a promising therapy in several clinical trials. However, its antidepressant mechanism of action is still not well understood. The aim of the present study was to evaluate the therapeutic potential of psilocybin in ameliorating the adverse behavioural and neurochemical consequences of chronic stress. To this end, a chronic unpredictable mild stress (CUMS) animal model was used, and psilocybin treatment was administered (two doses of 1 mg/kg, i.p., administered 7 days apart). Psilocybin reversed impairments in anhedonia and behavioural despair dimensions of depressive phenotype but not in apathy-related behaviour. Psilocybin administration was also able to exert an anxiolytic-like effect on treated animals. Physiological alterations caused by stress, indicative of a hyperactive hypothalamic-pituitary-adrenal axis (HPA), were not reversed by psilocybin. When neuroplasticity-related proteins were assessed in cerebral cortex, brain-derived neurotrophic factor (BDNF) was found to be decreased in stressed animals, and treatment did not reverse such impairment. Psilocybin administration increased the expression and function of serotonin-2A-receptor (5HT2AR) in brain cortex of control and CUMS groups. Furthermore, psilocybin treatment caused a selective increase in the expression of glucocorticoid-receptor (GR) in brain cortex of CUMS mice. In conclusion, psilocybin was able to rescue impairments in the depressive phenotype, and to induce anxiolytic-like effects. Furthermore, an enhancement in sensitivity to psilocybin-induced HTR was observed following a booster dose. Altogether, this work provides new knowledge on the putative benefit/risk actions of psilocybin and contributes to the understanding of the therapeutic mechanism of action of psychedelics.
Collapse
Affiliation(s)
| | - Igor Horrillo
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Leioa, Spain
- Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Nerea Martínez-Álvarez
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
| | - Daniel Pérez-Martínez
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
| | - Guadalupe Rivero
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Leioa, Spain
- Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Leioa, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Leioa, Spain
- Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Jorge E Ortega
- Department of Pharmacology, University of the Basque Country UPV/EHU, Leioa, Bizkaia, Spain.
- Centro de Investigación Biomédica en Red de Salud Mental, Instituto de Salud Carlos III, Leioa, Spain.
- Biobizkaia Health Research Institute, Barakaldo, Bizkaia, Spain.
| |
Collapse
|
3
|
Li C, Yan Y, Stork O, Shen R, Behnisch T. The E3 Ubiquitin Ligase PRAJA1: A Key Regulator of Synaptic Dynamics and Memory Processes with Implications for Alzheimer's Disease. Int J Mol Sci 2025; 26:2909. [PMID: 40243483 PMCID: PMC11988436 DOI: 10.3390/ijms26072909] [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: 02/05/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
The precise regulation of synaptic function by targeted protein degradation is fundamental to learning and memory, yet the roles of many brain-enriched E3 ubiquitin ligases in this process remain elusive. Here, we uncover a critical and previously unappreciated role for the E3 ubiquitin ligase PRAJA1 in orchestrating synaptic plasticity and hippocampus-dependent memory. Utilizing C57BL/6 and 5xFAD male mice and employing a multi-faceted approach including protein biochemistry, molecular biology, in vitro electrophysiology, and behavioral assays, we demonstrate that long-term potentiation (LTP) induction triggers a rapid, proteasome-dependent downregulation of PRAJA1 within the CA1 region of the hippocampus. Critically, selective knockdown of PRAJA1 in vivo profoundly enhanced both object recognition and spatial memory, while disrupting normal exploratory behavior. Mechanistically, we reveal that PRAJA1 acts as a key regulator of synaptic architecture and transmission: its downregulation leads to a reduction in key synaptic proteins and spine density, influencing the excitatory/inhibitory balance and facilitating synaptic plasticity. Conversely, increased PRAJA1 expression potentiates GABAergic transmission. Furthermore, we identify spinophilin as a novel substrate of PRAJA1, suggesting a direct molecular link between PRAJA1 and synaptic remodeling. Strikingly, our findings implicate dysregulation of PRAJA1 in the pathogenesis of Alzheimer's disease, positioning PRAJA1 as a potential therapeutic target for cognitive enhancement in neurodegenerative conditions. These results unveil PRAJA1 as a critical molecular brake on synaptic plasticity and memory formation, offering a promising new avenue for understanding and potentially treating memory impairment.
Collapse
Affiliation(s)
- Chuhan Li
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yan Yan
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany;
| | - Ruling Shen
- Shanghai Laboratory Animal Research Center, Shanghai 201203, China
| | - Thomas Behnisch
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
4
|
Ahn EH, Park JB. Molecular Mechanisms of Alzheimer's Disease Induced by Amyloid-β and Tau Phosphorylation Along with RhoA Activity: Perspective of RhoA/Rho-Associated Protein Kinase Inhibitors for Neuronal Therapy. Cells 2025; 14:89. [PMID: 39851517 PMCID: PMC11764136 DOI: 10.3390/cells14020089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/30/2024] [Accepted: 01/06/2025] [Indexed: 01/26/2025] Open
Abstract
Amyloid-β peptide (Aβ) is a critical cause of Alzheimer's disease (AD). It is generated from amyloid precursor protein (APP) through cleavages by β-secretase and γ-secretase. γ-Secretase, which includes presenilin, is regulated by several stimuli. Tau protein has also been identified as a significant factor in AD. In particular, Tau phosphorylation is crucial for neuronal impairment, as phosphorylated Tau detaches from microtubules, leading to the formation of neurofibrillary tangles and the destabilization of the microtubule structure. This instability in microtubules damages axons and dendrites, resulting in neuronal impairment. Notably, Aβ is linked to Tau phosphorylation. Another crucial factor in AD is neuroinflammation, primarily occurring in the microglia. Microglia possess several receptors that bind with Aβ, triggering the expression and release of an inflammatory factor, although their main physiological function is to phagocytose debris and pathogens in the brain. NF-κB activation plays a major role in neuroinflammation. Additionally, the production of reactive oxygen species (ROS) in the microglia contributes to this neuroinflammation. In microglia, superoxide is produced through NADPH oxidase, specifically NOX2. Rho GTPases play an essential role in regulating various cellular processes, including cytoskeletal rearrangement, morphology changes, migration, and transcription. The typical function of Rho GTPases involves regulating actin filament formation. Neurons, with their complex processes and synapse connections, rely on cytoskeletal dynamics for structural support. Other brain cells, such as astrocytes, microglia, and oligodendrocytes, also depend on specific cytoskeletal structures to maintain their unique cellular architectures. Thus, the aberrant regulation of Rho GTPases activity can disrupt actin filaments, leading to altered cell morphology, including changes in neuronal processes and synapses, and potentially contributing to brain diseases such as AD.
Collapse
Affiliation(s)
- Eun Hee Ahn
- Department of Physiology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea;
- Department of Neurology, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- Institute of Cell Differentiation and Aging, Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
- ELMED Co., Hallym University College of Medicine, Chuncheon 24252, Kangwon-do, Republic of Korea
| |
Collapse
|
5
|
Gutiérrez-Esparza BL, González-Torres ML, Quintanar-Stephano A, Quintanar JL. Neurological recovery in rats with portocaval anastomosis-induced hepatic encephalopathy treated with leuprolide acetate, a GnRH agonist. Metab Brain Dis 2024; 39:1447-1458. [PMID: 39177865 DOI: 10.1007/s11011-024-01413-9] [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: 06/07/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024]
Abstract
Hepatic encephalopathy (HE) is a neuropsychiatric complication of acute liver failure or chronic liver injury. Liver dysfunction impairs ammonia detoxification, allowing it to cross the blood-brain barrier (BBB) and disrupt brain function. The hippocampus becomes a crucial target during elevated ammonia levels, causing spatial memory impairment and decreased learning ability. Leuprolide acetate (LA), a GnRH agonist, has been implicated in neuroprotection and neuroregeneration in several regions of the central nervous system (CNS) including hippocampus. In this study, we aim to evaluate the effects of LA treatment on hippocampus of rats with HE induced by portocaval anastomosis (PCA) trough cognitive tests, histology analysis and expression of neuronal recovery marker proteins, such as neurofilament (NF200) and neurabin II, and astrocyte marker glial fibrillary acidic protein (GFAP). Rats were divided into three groups: SHAM, portocaval anastomosis with saline solution (PCA + SS) and portocaval anastomosis treated with LA (PCA + LA). To evaluate learning and spatial memory elevated T-maze (ETM) and Y-maze test (YMT) were respectively used. Results indicated that LA-treated rats performed significantly better in ETM and YMT than untreated rats. Histological analysis of hippocampus showed increased neuron density, nuclear area, and layer thickness in dentate gyrus of PCA + LA group compared to PCA + SS. Additionally, neurabin II and NF200 expression were higher in LA-treated rats, while GFAP expression was elevated in the PCA + SS group compared to control and PCA + LA groups. In conclusion, LA enhances hippocampal neuron recovery and reduces astrogliosis, suggesting its potential as a therapeutic intervention for attenuating hippocampal damage during HE.
Collapse
Affiliation(s)
- Brenda Lizeth Gutiérrez-Esparza
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad, No. 940, Ciudad Universitaria, C.P. 20100, Aguascalientes, Aguascalientes, México
| | - Marina Liliana González-Torres
- Departamento de Psicología, Centro de Ciencias Sociales y Humanidades, Universidad Autónoma de Aguascalientes, Av. Universidad, No. 940, Ciudad Universitaria, C.P. 20100, Aguascalientes, Aguascalientes, México
| | - Andrés Quintanar-Stephano
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad, No. 940, Ciudad Universitaria, C.P. 20100, Aguascalientes, Aguascalientes, México
| | - J Luis Quintanar
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Av. Universidad, No. 940, Ciudad Universitaria, C.P. 20100, Aguascalientes, Aguascalientes, México.
| |
Collapse
|
6
|
Higa GSV, Viana FJC, Francis-Oliveira J, Cruvinel E, Franchin TS, Marcourakis T, Ulrich H, De Pasquale R. Serotonergic neuromodulation of synaptic plasticity. Neuropharmacology 2024; 257:110036. [PMID: 38876308 DOI: 10.1016/j.neuropharm.2024.110036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024]
Abstract
Synaptic plasticity constitutes a fundamental process in the reorganization of neural networks that underlie memory, cognition, emotional responses, and behavioral planning. At the core of this phenomenon lie Hebbian mechanisms, wherein frequent synaptic stimulation induces long-term potentiation (LTP), while less activation leads to long-term depression (LTD). The synaptic reorganization of neuronal networks is regulated by serotonin (5-HT), a neuromodulator capable of modify synaptic plasticity to appropriately respond to mental and behavioral states, such as alertness, attention, concentration, motivation, and mood. Lately, understanding the serotonergic Neuromodulation of synaptic plasticity has become imperative for unraveling its impact on cognitive, emotional, and behavioral functions. Through a comparative analysis across three main forebrain structures-the hippocampus, amygdala, and prefrontal cortex, this review discusses the actions of 5-HT on synaptic plasticity, offering insights into its role as a neuromodulator involved in emotional and cognitive functions. By distinguishing between plastic and metaplastic effects, we provide a comprehensive overview about the mechanisms of 5-HT neuromodulation of synaptic plasticity and associated functions across different brain regions.
Collapse
Affiliation(s)
- Guilherme Shigueto Vilar Higa
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil; Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Felipe José Costa Viana
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - José Francis-Oliveira
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Emily Cruvinel
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Thainá Soares Franchin
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Tania Marcourakis
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química (USP), Butantã, São Paulo, SP, 05508-900, Brazil
| | - Roberto De Pasquale
- Laboratório de Neurofisiologia, Departamento de Fisiologia e Biofísica, Universidade de São Paulo, Butantã, São Paulo, SP, 05508-000, Brazil.
| |
Collapse
|
7
|
Stickel KC, Shah NR, Claeboe ET, Orr KS, Mosley AL, Doud EH, Belecky-Adams TL, Baucum AJ. Mechanisms of spinophilin-dependent pancreas dysregulation in obesity. Am J Physiol Endocrinol Metab 2024; 327:E155-E171. [PMID: 38630048 PMCID: PMC11427100 DOI: 10.1152/ajpendo.00099.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 03/18/2024] [Accepted: 04/04/2024] [Indexed: 05/07/2024]
Abstract
Spinophilin is an F-actin binding and protein phosphatase 1 (PP1) targeting protein that acts as a scaffold of PP1 to its substrates. Spinophilin knockout (Spino-/-) mice have decreased fat mass, increased lean mass, and improved glucose tolerance, with no difference in feeding behaviors. Although spinophilin is enriched in neurons, its roles in nonneuronal tissues, such as β cells of the pancreatic islets, are unclear. We have corroborated and expanded upon previous studies to determine that Spino-/- mice have decreased weight gain and improved glucose tolerance in two different models of obesity. We have identified multiple putative spinophilin-interacting proteins isolated from intact pancreas and observed increased interactions of spinophilin with exocrine, ribosomal, and cytoskeletal protein classes that normally act to mediate peptide hormone production, processing, and/or release in Leprdb/db and/or high-fat diet-fed (HFF) models of obesity. In addition, we have found that spinophilin interacts with proteins from similar classes in isolated islets, suggesting a role for spinophilin in the pancreatic islet. Consistent with a pancreatic β cell type-specific role for spinophilin, using our recently described conditional spinophilin knockout mice, we found that loss of spinophilin specifically in pancreatic β cells improved glucose tolerance without impacting body weight in chow-fed mice. Our data further support the role of spinophilin in mediating pathophysiological changes in body weight and whole body metabolism associated with obesity. Our data provide the first evidence that pancreatic spinophilin protein interactions are modulated by obesity and that loss of spinophilin specifically in pancreatic β cells impacts whole body glucose tolerance.NEW & NOTEWORTHY To our knowledge, these data are the first to demonstrate that obesity impacts spinophilin protein interactions in the pancreas and identify spinophilin specifically in pancreatic β cells as a modulator of whole body glucose tolerance.
Collapse
Affiliation(s)
- Kaitlyn C Stickel
- Department of Biology, Indiana University-Indianapolis, Indianapolis, Indiana, United States
| | - Nikhil R Shah
- Medical Neurosciences and Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Emily T Claeboe
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Kara S Orr
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Amber L Mosley
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Center for Proteome Analysis, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Emma H Doud
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Center for Proteome Analysis, Indiana University School of Medicine, Indianapolis, Indiana, United States
| | - Teri L Belecky-Adams
- Department of Biology, Indiana University-Indianapolis, Indianapolis, Indiana, United States
| | - Anthony J Baucum
- Department of Biology, Indiana University-Indianapolis, Indianapolis, Indiana, United States
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, Indiana, United States
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, Indiana, United States
| |
Collapse
|
8
|
Mitra A, Deats SP, Dickson PE, Zhu J, Gardin J, Nieman BJ, Henkelman RM, Tsai NP, Chesler EJ, Zhang ZW, Kumar V. Tmod2 Is a Regulator of Cocaine Responses through Control of Striatal and Cortical Excitability and Drug-Induced Plasticity. J Neurosci 2024; 44:e1389232024. [PMID: 38508714 PMCID: PMC11063827 DOI: 10.1523/jneurosci.1389-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 02/12/2024] [Accepted: 02/24/2024] [Indexed: 03/22/2024] Open
Abstract
Drugs of abuse induce neuroadaptations, including synaptic plasticity, that are critical for transition to addiction, and genes and pathways that regulate these neuroadaptations are potential therapeutic targets. Tropomodulin 2 (Tmod2) is an actin-regulating gene that plays an important role in synapse maturation and dendritic arborization and has been implicated in substance abuse and intellectual disability in humans. Here, we mine the KOMP2 data and find that Tmod2 knock-out mice show emotionality phenotypes that are predictive of addiction vulnerability. Detailed addiction phenotyping shows that Tmod2 deletion does not affect the acute locomotor response to cocaine administration. However, sensitized locomotor responses are highly attenuated in these knock-outs, indicating perturbed drug-induced plasticity. In addition, Tmod2 mutant animals do not self-administer cocaine indicating lack of hedonic responses to cocaine. Whole-brain MR imaging shows differences in brain volume across multiple regions, although transcriptomic experiments did not reveal perturbations in gene coexpression networks. Detailed electrophysiological characterization of Tmod2 KO neurons showed increased spontaneous firing rate of early postnatal and adult cortical and striatal neurons. Cocaine-induced synaptic plasticity that is critical for sensitization is either missing or reciprocal in Tmod2 KO nucleus accumbens shell medium spiny neurons, providing a mechanistic explanation of the cocaine response phenotypes. Combined, these data, collected from both males and females, provide compelling evidence that Tmod2 is a major regulator of plasticity in the mesolimbic system and regulates the reinforcing and addictive properties of cocaine.
Collapse
Affiliation(s)
| | | | | | - Jiuhe Zhu
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | | | - Brian J Nieman
- Mouse Imaging Centre and Translational Medicine, Hospital for Sick Children; Ontario Institute for Cancer Research; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T 3H7, Canada
| | - R Mark Henkelman
- Mouse Imaging Centre and Translational Medicine, Hospital for Sick Children; Ontario Institute for Cancer Research; Department of Medical Biophysics, University of Toronto, Toronto, Ontario M5T 3H7, Canada
| | - Nien-Pei Tsai
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | | | | | - Vivek Kumar
- The Jackson Laboratory, Bar Harbor, Maine 04609
| |
Collapse
|
9
|
Macedo-Mendoza M, Calderón-Vallejo D, González-Torres M, Martínez-Martínez A, Gasca-Martínez D, Orta Salazar E, Quintanar J. SPATIAL MEMORY RECOVERY IN AGED MALE RATS TREATED WITH LEUPROLIDE ACETATE, A GNRH AGONIST. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2024; 20:127-135. [PMID: 39845751 PMCID: PMC11750219 DOI: 10.4183/aeb.2024.127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Context Studies indicate a decrease in spatial memory across species as they age. Moreover, consistent administration of Gonadotropin-releasing hormone (GnRH) improves learning abilities in older rats that have undergone gonadectomy. Objective The aim of this study was to investigate the effects of the GnRH agonist, leuprolide acetate (LA) on spatial memory in aged intact male rats and the expression of proteins associated with hippocampal plasticity. Subjects and methods Aged male rats were injected with LA or saline every three days for nine weeks to assess the effects of LA on spatial memory. The aged rats were trained in the Morris Water Maze for four days, and the evaluation took place on the fifth day. Additionally, the Y-maze test was used to investigate short-term spatial memory. The expression of spinophilin and microtubule-associated protein 2 (MAP2) in the hippocampus was measured. Results Behavioral tests revealed that LA improves spatial memory in aged rats. Additionally, we observed increased expression of spinophilin and MAP2 in the hippocampus of aged male rats following LA treatment. Based on these results, the administration of LA holds is a potential treatment for restoring cognitive function in aged individuals.
Collapse
Affiliation(s)
- M. Macedo-Mendoza
- Universidad Autónoma de Aguascalientes, Centro de Ciencias Básicas, Department of Physiology and Pharmacology
| | - D. Calderón-Vallejo
- Universidad Autónoma de Aguascalientes, Centro de Ciencias Básicas, Department of Physiology and Pharmacology
- Department of Morphology
| | - M.L. González-Torres
- Universidad Autónoma de Aguascalientes, Department of Psychology, Aguascalientes
| | - A. Martínez-Martínez
- Universidad Autónoma de Aguascalientes, Department of Psychology, Aguascalientes
| | - D. Gasca-Martínez
- Universidad Nacional Autónoma de México, Instituto de Neurobiología, Juriquilla, Querétaro, México
| | - E.M. Orta Salazar
- Universidad Nacional Autónoma de México, Instituto de Neurobiología, Juriquilla, Querétaro, México
| | - J.L. Quintanar
- Universidad Autónoma de Aguascalientes, Centro de Ciencias Básicas, Department of Physiology and Pharmacology
| |
Collapse
|
10
|
Li G, McLaughlin DW, Peskin CS. A biochemical description of postsynaptic plasticity-with timescales ranging from milliseconds to seconds. Proc Natl Acad Sci U S A 2024; 121:e2311709121. [PMID: 38324573 PMCID: PMC10873618 DOI: 10.1073/pnas.2311709121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 12/29/2023] [Indexed: 02/09/2024] Open
Abstract
Synaptic plasticity [long-term potentiation/depression (LTP/D)], is a cellular mechanism underlying learning. Two distinct types of early LTP/D (E-LTP/D), acting on very different time scales, have been observed experimentally-spike timing dependent plasticity (STDP), on time scales of tens of ms; and behavioral time scale synaptic plasticity (BTSP), on time scales of seconds. BTSP is a candidate for a mechanism underlying rapid learning of spatial location by place cells. Here, a computational model of the induction of E-LTP/D at a spine head of a synapse of a hippocampal pyramidal neuron is developed. The single-compartment model represents two interacting biochemical pathways for the activation (phosphorylation) of the kinase (CaMKII) with a phosphatase, with ion inflow through channels (NMDAR, CaV1,Na). The biochemical reactions are represented by a deterministic system of differential equations, with a detailed description of the activation of CaMKII that includes the opening of the compact state of CaMKII. This single model captures realistic responses (temporal profiles with the differing timescales) of STDP and BTSP and their asymmetries. The simulations distinguish several mechanisms underlying STDP vs. BTSP, including i) the flow of [Formula: see text] through NMDAR vs. CaV1 channels, and ii) the origin of several time scales in the activation of CaMKII. The model also realizes a priming mechanism for E-LTP that is induced by [Formula: see text] flow through CaV1.3 channels. Once in the spine head, this small additional [Formula: see text] opens the compact state of CaMKII, placing CaMKII ready for subsequent induction of LTP.
Collapse
Affiliation(s)
- Guanchun Li
- Courant Institute and Center for Neural Science, Department of Mathematics, New York University, New York, NY10012
| | - David W. McLaughlin
- Courant Institute and Center for Neural Science, Department of Mathematics, New York University, New York, NY10012
- Center for Neural Science, Department of Neural Science, New York University, New York, NY10012
- Institute of Mathematical Science, Mathematics Department, New York University-Shanghai, Shanghai200122, China
- Neuroscience Institute of New York University Langone Health, New York University, New York, NY10016
| | - Charles S. Peskin
- Courant Institute and Center for Neural Science, Department of Mathematics, New York University, New York, NY10012
- Center for Neural Science, Department of Neural Science, New York University, New York, NY10012
| |
Collapse
|
11
|
Li C, Ma Y, Chai X, Feng X, Feng W, Zhao Y, Cui C, Wang J, Zhao S, Zhu X. Ketogenic diet attenuates cognitive dysfunctions induced by hypoglycemia via inhibiting endoplasmic reticulum stress-dependent pathways. Food Funct 2024; 15:1294-1309. [PMID: 38197246 DOI: 10.1039/d3fo04007k] [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: 01/11/2024]
Abstract
Hypoglycemia can potentially cause severe damage to the central nervous system. The ketogenic diet (KD), characterized by high-fat and extremely low-carbohydrate content, can modulate homeostasis and nutrient metabolism, thereby influencing body health. However, the effects and underlying mechanisms of KD on hypoglycemia-induced brain injury have not been thoroughly investigated. We aimed to explore the modulating effects of KD on cognitive functions and elucidate the underlying mechanisms. In this study, one-month-old mice were fed with KD for 2 weeks, and the changes in the gut microbiota were detected using the 16S rRNA gene amplicon sequencing method. The hypoglycemic model of mice was established using insulin, and the potential protective effect of KD on hypoglycemia-induced brain injury in mice was evaluated through immunofluorescence staining, western blotting, transmission electron microscopy, and Golgi staining. Our results showed that the intestinal flora of Dorea increased and Rikenella decreased in KD-fed mice. KD can not only alleviate anxiety-like behavior induced by hypoglycemia, but also increase the proportion of mushroom dendritic spines in the hippocampus by modulating changes in the gut microbiota. KD regulated synaptic plasticity by increasing the levels of SPN, PSD95, and SYP, which relieve cognitive impairment caused by hypoglycemia. Moreover, KD can promote the proliferation and survival of adult neural stem cells in the hippocampus, while reducing apoptosis by suppressing the activation of the IRE1-XBP1 and ATF6 endoplasmic reticulum stress pathways in mice with hypoglycemia. This study provides new evidence for demonstrating that KD may alleviate cognitive dysfunctions caused by hypoglycemia by modulating the gut microbiota.
Collapse
Affiliation(s)
- Cixia Li
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Xuejun Chai
- College of Basic Medicine, Xi'An Medical University, Xi'An, Shaanxi 710021, PR China
| | - Xiao Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Wenyu Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Yongkang Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Chaochu Cui
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
12
|
Jahanbakhshi H, Moghaddam MH, Sani M, Parvardeh S, Boroujeni ME, Vakili K, Fathi M, Azimi H, Mehranpour M, Abdollahifar MA, Ghafghazi S, Sadidi M, Aliaghaei A, Bayat AH, Peyvandi AA. The elderberry diet protection against intrahippocampal Aβ-induced memory dysfunction; the abrogated apoptosis and neuroinflammation. Toxicol Res (Camb) 2023; 12:1063-1076. [PMID: 38145093 PMCID: PMC10734613 DOI: 10.1093/toxres/tfad097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/31/2023] [Accepted: 10/04/2023] [Indexed: 12/26/2023] Open
Abstract
This study evaluates whether elderberry (EB) effectively decreases the inflammation and oxidative stress in the brain cells to reduce Aβ toxicity. In the Aβ + EB group, EB powder was added to rats' routine diet for eight consecutive weeks. Then, spatial memory, working memory, and long-term memory, were measured using the Morris water maze, T-maze, and passive avoidance test. Also, in this investigation immunohistopathology, distribution of hippocampal cells, and gene expression was carried out. Voronoi tessellation method was used to estimate the spatial distribution of the cells in the hippocampus. In addition to improving the memory functions of rats with Aβ toxicity, a reduction in astrogliosis and astrocytes process length and the number of branches and intersections distal to the soma was observed in their hippocampus compared to the control group. Further analysis indicated that the EB diet decreased the caspase-3 expression in the hippocampus of rats with Aβ toxicity. Also, EB protected hippocampal pyramidal neurons against Aβ toxicity and improved the spatial distribution of the hippocampal neurons. Moreover, EB decreased the expression of inflammatory and apoptotic genes. Overall, our study suggest that EB can be considered a potent modifier of astrocytes' reactivation and inflammatory responses.
Collapse
Affiliation(s)
- Hadiseh Jahanbakhshi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mojtaba Sani
- Department of Educational Neuroscience, Aras International Campus, University of Tabriz, Tabriz, Iran
| | - Siavash Parvardeh
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Eskandarian Boroujeni
- Laboratory of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Helia Azimi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Mehranpour
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shiva Ghafghazi
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Sadidi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir-Hossein Bayat
- Department of Neuroscience, School of Sciences and Advanced Technology in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ali Asghar Peyvandi
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Wang C, Zhou W, Zhang L, Fu L, Shi W, Qing Y, Lu F, Tang J, Gao X, Zhang A, Jia Z, Zhang Y, Zhao X, Zheng B. Diagnostic yield and novel candidate genes for neurodevelopmental disorders by exome sequencing in an unselected cohort with microcephaly. BMC Genomics 2023; 24:422. [PMID: 37501076 PMCID: PMC10373276 DOI: 10.1186/s12864-023-09505-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 07/04/2023] [Indexed: 07/29/2023] Open
Abstract
OBJECTIVES Microcephaly is caused by reduced brain volume and most usually associated with a variety of neurodevelopmental disorders (NDDs). To provide an overview of the diagnostic yield of whole exome sequencing (WES) and promote novel candidates in genetically unsolved families, we studied the clinical and genetic landscape of an unselected Chinese cohort of patients with microcephaly. METHODS We performed WES in an unselected cohort of 103 NDDs patients with microcephaly as one of the features. Full evaluation of potential novel candidate genes was applied in genetically undiagnosed families. Functional validations of selected variants were conducted in cultured cells. To augment the discovery of novel candidates, we queried our genomic sequencing data repository for additional likely disease-causing variants in the identified candidate genes. RESULTS In 65 families (63.1%), causative sequence variants (SVs) and clinically relevant copy number variants (CNVs) with a pathogenic or likely pathogenic (P/LP) level were identified. By incorporating coverage analysis to WES, a pathogenic or likely pathogenic CNV was detected in 15 families (16/103, 15.5%). In another eight families (8/103, 7.8%), we identified variants in newly reported gene (CCND2) and potential novel neurodevelopmental disorders /microcephaly candidate genes, which involved in cell cycle and division (PWP2, CCND2), CDC42/RAC signaling related actin cytoskeletal organization (DOCK9, RHOF), neurogenesis (ELAVL3, PPP1R9B, KCNH3) and transcription regulation (IRF2BP1). By looking into our data repository of 5066 families with NDDs, we identified additional two cases with variants in DOCK9 and PPP1R9B, respectively. CONCLUSION Our results expand the morbid genome of monogenic neurodevelopmental disorders and support the adoption of WES as a first-tier test for individuals with microcephaly.
Collapse
Affiliation(s)
- Chunli Wang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhou
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Luyan Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Luhan Fu
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Shi
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yan Qing
- Department of Neurosurgery, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Fen Lu
- Department of Rehabilitation Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Tang
- Department of Rehabilitation Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Xiucheng Gao
- Department of Radiology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.
| | - Xiaoke Zhao
- Department of Rehabilitation Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics, Children's Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
14
|
Salek AB, Claeboe ET, Bansal R, Berbari NF, Baucum AJ. Spinophilin-dependent regulation of GluN2B-containing NMDAR-dependent calcium influx, GluN2B surface expression, and cleaved caspase expression. Synapse 2023; 77:e22264. [PMID: 36738175 PMCID: PMC11648995 DOI: 10.1002/syn.22264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/09/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023]
Abstract
N-methyl-d-aspartate receptors (NMDARs) are calcium-permeable ion channels that are ubiquitously expressed within the glutamatergic postsynaptic density. Phosphorylation of NMDAR subunits defines receptor conductance and surface localization, two alterations that can modulate overall channel activity. Modulation of NMDAR phosphorylation by kinases and phosphatases regulates the amount of calcium entering the cell and subsequent activation of calcium-dependent processes. The dendritic spine enriched protein, spinophilin, is the major synaptic protein phosphatase 1 (PP1) targeting protein. Depending on the substrate, spinophilin can act as either a PP1 targeting protein, to permit substrate dephosphorylation, or a PP1 inhibitory protein, to enhance substrate phosphorylation. Spinophilin limits NMDAR function in a PP1-dependent manner. Specifically, we have previously shown that spinophilin sequesters PP1 away from the GluN2B subunit of the NMDAR, which results in increased phosphorylation of Ser-1284 on GluN2B. However, how spinophilin modifies NMDAR function is unclear. Herein, we utilize a Neuro2A cell line to detail that Ser-1284 phosphorylation increases calcium influx via GluN2B-containing NMDARs. Moreover, overexpression of spinophilin decreases GluN2B-containing NMDAR activity by decreasing its surface expression, an effect that is independent of Ser-1284 phosphorylation. In hippocampal neurons isolated from spinophilin knockout animals, there is an increase in cleaved caspase-3 levels, a marker of calcium-associated apoptosis, compared with wildtype mice. Taken together, our data demonstrate that spinophilin regulates GluN2B containing NMDAR phosphorylation, channel function, and trafficking and that loss of spinophilin enhances neuronal cleaved caspase-3 expression.
Collapse
Affiliation(s)
- Asma B. Salek
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Emily T. Claeboe
- Department of Pharmacology and Toxicology, Indiana University School of Medicine Indianapolis, IN, USA
| | - Ruchi Bansal
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Nicolas F. Berbari
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine. Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine. Indianapolis, IN, USA
| | - Anthony J. Baucum
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
- Department of Pharmacology and Toxicology, Indiana University School of Medicine Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine. Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine. Indianapolis, IN, USA
| |
Collapse
|
15
|
Wang YY, Zhou YN, Jiang L, Wang S, Zhu L, Zhang SS, Yang H, He Q, Liu L, Xie YH, Liang X, Tang J, Chao FL, Tang Y. Long-term voluntary exercise inhibited AGE/RAGE and microglial activation and reduced the loss of dendritic spines in the hippocampi of APP/PS1 transgenic mice. Exp Neurol 2023; 363:114371. [PMID: 36871860 DOI: 10.1016/j.expneurol.2023.114371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/22/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Alzheimer's disease (AD) is closely related to hippocampal synapse loss, which can be alleviated by running exercise. However, further studies are needed to determine whether running exercise reduces synapse loss in the hippocampus in an AD model by regulating microglia. Ten-month-old male wild-type mice and APP/PS1 mice were randomly divided into control and running groups. All mice in the running groups were subjected to voluntary running exercise for four months. After the behavioral tests, immunohistochemistry, stereological methods, immunofluorescence staining, 3D reconstruction, western blotting and RNA-Seq were performed. Running exercise improved the spatial learning and memory abilities of APP/PS1 mice and increased the total number of dendritic spines, the levels of the PSD-95 and Synapsin Ia/b proteins, the colocalization of PSD-95 and neuronal dendrites (MAP-2) and the number of PSD-95-contacting astrocytes (GFAP) in the hippocampi of APP/PS1 mice. Moreover, running exercise reduced the relative expression of CD68 and Iba-1, the number of Iba-1+ microglia and the colocalization of PSD-95 and Iba-1+ microglia in the hippocampi of APP/PS1 mice. The RNA-Seq results showed that some differentially expressed genes (DEGs) related to the complement system (Cd59b, Serping1, Cfh, A2m, and Trem2) were upregulated in the hippocampi of APP/PS1 mice, while running exercise downregulated the C3 gene. At the protein level, running exercise also reduced the expression of advanced glycation end products (AGEs), receptor for advanced glycation end products (RAGE), C1q and C3 in the hippocampus and AGEs and RAGE in hippocampal microglia in APP/PS1 mice. Furthermore, the Col6a3, Scn5a, Cxcl5, Tdg and Clec4n genes were upregulated in the hippocampi of APP/PS1 mice but downregulated after running, and these genes were associated with the C3 and RAGE genes according to protein-protein interaction (PPI) analysis. These findings indicate that long-term voluntary exercise might protect hippocampal synapses and affect the function and activation of microglia, the AGE/RAGE signaling pathway in microglia and the C1q/C3 complement system in the hippocampus in APP/PS1 mice, and these effects may be related to the Col6a3, Scn5a, Cxcl5, Tdg and Clec4n genes. The current results provide an important basis for identifying targets for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Yi-Ying Wang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Yu-Ning Zhou
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Jiang
- Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Shun Wang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Lin Zhu
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Shan-Shan Zhang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Hao Yang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Qi He
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Li Liu
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Yu-Han Xie
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Xin Liang
- Department of Pathophysiology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Jing Tang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China
| | - Feng-Lei Chao
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China.
| | - Yong Tang
- Department of Histology and Embryology, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China; Laboratory of Stem Cell and Tissue Engineering, Faculty of Basic Medical College, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
16
|
Dysregulated Signaling at Postsynaptic Density: A Systematic Review and Translational Appraisal for the Pathophysiology, Clinics, and Antipsychotics' Treatment of Schizophrenia. Cells 2023; 12:cells12040574. [PMID: 36831241 PMCID: PMC9954794 DOI: 10.3390/cells12040574] [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: 11/30/2022] [Revised: 02/07/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
Emerging evidence from genomics, post-mortem, and preclinical studies point to a potential dysregulation of molecular signaling at postsynaptic density (PSD) in schizophrenia pathophysiology. The PSD that identifies the archetypal asymmetric synapse is a structure of approximately 300 nm in diameter, localized behind the neuronal membrane in the glutamatergic synapse, and constituted by more than 1000 proteins, including receptors, adaptors, kinases, and scaffold proteins. Furthermore, using FASS (fluorescence-activated synaptosome sorting) techniques, glutamatergic synaptosomes were isolated at around 70 nm, where the receptors anchored to the PSD proteins can diffuse laterally along the PSD and were stabilized by scaffold proteins in nanodomains of 50-80 nm at a distance of 20-40 nm creating "nanocolumns" within the synaptic button. In this context, PSD was envisioned as a multimodal hub integrating multiple signaling-related intracellular functions. Dysfunctions of glutamate signaling have been postulated in schizophrenia, starting from the glutamate receptor's interaction with scaffolding proteins involved in the N-methyl-D-aspartate receptor (NMDAR). Despite the emerging role of PSD proteins in behavioral disorders, there is currently no systematic review that integrates preclinical and clinical findings addressing dysregulated PSD signaling and translational implications for antipsychotic treatment in the aberrant postsynaptic function context. Here we reviewed a critical appraisal of the role of dysregulated PSD proteins signaling in the pathophysiology of schizophrenia, discussing how antipsychotics may affect PSD structures and synaptic plasticity in brain regions relevant to psychosis.
Collapse
|
17
|
Stickel KC, Mosley AL, Doud EH, Belecky-Adams TL, Baucum AJ. Mechanisms of spinophilin-dependent pancreas dysregulation underlying diabesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527495. [PMID: 36798361 PMCID: PMC9934546 DOI: 10.1101/2023.02.07.527495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Objective Spinophilin is an F-actin binding and protein phosphatase 1 (PP1) targeting protein that acts as a scaffold of PP1 to its substrates. Spinophilin knockout (Spino-/-) mice have decreased fat mass, increased lean mass, and improved glucose tolerance, with no difference in feeding behaviors. While spinophilin is enriched in neurons, its roles in non-neuronal tissues, such as beta cells of the pancreatic islets, are unclear. Methods & Results We have corroborated and expanded upon previous studies to determine that Spino-/- mice have decreased weight gain and improved glucose tolerance in two different models of obesity. Using proteomics and immunoblotting-based approaches we identified multiple putative spinophilin interacting proteins isolated from intact pancreas and observed increased interactions of spinophilin with exocrine, ribosomal, and cytoskeletal protein classes that mediate peptide hormone production, processing, and/or release in Leprdb/db and/or high fat-fed (HFF) models of obesity. Moreover, loss of spinophilin specifically in pancreatic beta cells improved glucose tolerance without impacting body weight. Conclusion Our data further support a role for spinophilin in mediating pathophysiological changes in body weight and whole-body metabolism associated with obesity and provide the first evidence that spinophilin mediates obesity-dependent pancreatic dysfunction that leads to deficits in glucose homeostasis or diabesity.
Collapse
Affiliation(s)
- Kaitlyn C. Stickel
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Amber L. Mosley
- Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
- Center for Proteome Analysis, Indiana University, Indianapolis, IN, USA
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine (IUSM), Indiana University, Indianapolis, IN, USA
| | - Emma H. Doud
- Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
- Center for Proteome Analysis, Indiana University, Indianapolis, IN, USA
| | - Teri L. Belecky-Adams
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
| | - Anthony J. Baucum
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, USA
- Department of Pharmacology and Toxicology, Indiana University, Indianapolis, IN, USA
- Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA
- Center for Diabetes and Metabolic Diseases Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
18
|
Saggu S, Chen Y, Cottingham C, Rehman H, Wang H, Zhang S, Augelli-Szafran C, Lu S, Lambert N, Jiao K, Lu XY, Wang Q. Activation of a novel α 2AAR-spinophilin-cofilin axis determines the effect of α 2 adrenergic drugs on fear memory reconsolidation. Mol Psychiatry 2023; 28:588-600. [PMID: 36357671 PMCID: PMC9647772 DOI: 10.1038/s41380-022-01851-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 11/11/2022]
Abstract
Posttraumatic stress disorder (PTSD) after the pandemic has emerged as a major neuropsychiatric component of post-acute COVID-19 syndrome, yet the current pharmacotherapy for PTSD is limited. The use of adrenergic drugs to treat PTSD has been suggested; however, it is hindered by conflicting clinical results and a lack of mechanistic understanding of drug actions. Our studies, using both genetically modified mice and human induced pluripotent stem cell-derived neurons, reveal a novel α2A adrenergic receptor (α2AAR)-spinophilin-cofilin axis in the hippocampus that is critical for regulation of contextual fear memory reconsolidation. In addition, we have found that two α2 ligands, clonidine and guanfacine, exhibit differential abilities in activating this signaling axis to disrupt fear memory reconsolidation. Stimulation of α2AAR with clonidine, but not guanfacine, promotes the interaction of the actin binding protein cofilin with the receptor and with the dendritic spine scaffolding protein spinophilin to induce cofilin activation at the synapse. Spinophilin-dependent regulation of cofilin is required for clonidine-induced disruption of contextual fear memory reconsolidation. Our results inform the interpretation of differential clinical observations of these two drugs on PTSD and suggest that clonidine could provide immediate treatment for PTSD symptoms related to the current pandemic. Furthermore, our study indicates that modulation of dendritic spine morphology may represent an effective strategy for the development of new pharmacotherapies for PTSD.
Collapse
Affiliation(s)
- Shalini Saggu
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Yunjia Chen
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Christopher Cottingham
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Department of Biology, University of North Alabama, Florence, AL, 35632, USA
| | - Hasibur Rehman
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Hongxia Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Sixue Zhang
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL, 35205, USA
| | - Corinne Augelli-Szafran
- Department of Chemistry, Scientific Platforms, Southern Research, Birmingham, AL, 35205, USA
- Scientific Platforms, Southern Research, Birmingham, AL, 35205, USA
| | - Sumin Lu
- Department of Pharmacology, Medical College of Georgia at Augusta University, Augusta, GA, GA30912, USA
| | - Nevin Lambert
- Department of Pharmacology, Medical College of Georgia at Augusta University, Augusta, GA, GA30912, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia at Augusta University, Augusta, GA, GA30912, USA
| | - Xin-Yun Lu
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA
| | - Qin Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
19
|
Feng Y, Yang L, Ma X, Huang Z, Zong X, Citadin CT, Lin HW, Zhang Q. Photobiomodulation treatment inhibits neurotoxic astrocytic polarization and protects neurons in in vitro and in vivo stroke models. Neurochem Int 2023; 162:105464. [PMID: 36539162 DOI: 10.1016/j.neuint.2022.105464] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
The beneficial effects of photobiomodulation (PBM) on function recovery after stroke have been well-established, while its molecular and cellular mechanisms remain to be elucidated. The current study was designed to investigate the effect of PBM on synaptic proteins and astrocyte polarization of photothrombotic (PT)-stroke induced rats in vivo, and explore the possible effect of PBM treatment on oxygen-glucose deprivation (OGD)-induced neurotoxic astrocytic polarization in vitro. We reported that 2-min PBM treatment (808 nm) for 7 days significantly increased synaptic proteins and neuroprotective astrocytic marker S100 Calcium Binding Protein A10 (S100A10) and inhibited neurotoxic astrocytic marker C3d in the peri-infarct region after ischemic stroke. Cell culture studies of primary cortical neurons and N2a cells showed that single-dose PBM treatment could increase cellular viability, regulate the apoptotic proteins (Caspase 9, Bcl-xL and BAX) and preserve synaptic proteins following OGD exposure. Additionly, PBM decreased the levels of C3d, inducible nitric oxide synthase (iNOS) and interleukin 1β (IL-1β) on astrocytes exposed to OGD. In summary, we demonstrated that PBM could inhibit neurotoxic astrocytic polarization, preserve synaptic integrity and protect neurons against stroke injury both in vitro and in vivo.
Collapse
Affiliation(s)
- Yu Feng
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA
| | - Luodan Yang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA
| | - Xiaohui Ma
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA
| | - Zhihai Huang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA
| | - Xuemei Zong
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA
| | - Cristiane Teresinha Citadin
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA; Department of Cellular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA
| | - Hung Wen Lin
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA
| | - Quanguang Zhang
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, 1501 Kings Highway, LA, 71103, USA.
| |
Collapse
|
20
|
Kaur H, Sarmah D, Datta A, Borah A, Yavagal DR, Bhattacharya P. Endovascular Stem Cell Therapy Promotes Neuronal Remodeling to Enhance Post Stroke Recovery by Alleviating Endoplasmic Reticulum Stress Modulated by BDNF Signaling. Stem Cell Rev Rep 2023; 19:264-274. [PMID: 36251114 DOI: 10.1007/s12015-022-10461-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2022] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND PURPOSE The impact of increased BDNF expression in brain by endovascular delivered mesenchymal stem cells (MSCs) post stroke towards modulating endoplasmic reticulum (ER) stress mediated neuronal remodeling has not been directly studied. Therefore, the present study investigates ER stress mediated neuronal remodeling following IA MSCs infusion in rodent model of ischemic stroke. METHODS Ovariectomized Sprague Dawley rats were subjected to MCAO followed by 1 × 105 IA MSCs administration at 6 h. Infarct and functional outcomes at different time points post-stroke were evaluated. Further, various genes and protein expression studies were performed to determine the underlying mechanisms of the effect of IA MSCs towards ER stress mediated neuronal remodeling. RESULTS Post-stroke IA MSCs administration significantly increased BDNF expression and decreased ER stress markers expression at day 1 post-stroke. A gradual rise in the expression of growth associate protein-43 (GAP 43) and spinophilin were observed at 7, 14- and 28-days post-stroke indicating an increase in neuronal remodeling towards functional restoration. CONCLUSIONS The results suggest that IA MSCs post-stroke can modulate neuronal remodeling by BDNF-mediated reduction in ER stress that contribute towards functional recovery.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gandhinagar, Gujarat, 382355, India
| | - Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gandhinagar, Gujarat, 382355, India
| | - Aishika Datta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gandhinagar, Gujarat, 382355, India
| | - Anupom Borah
- Cellular and Molecular Neurobiology Laboratory, Department of Life Science and Bioinformatics, Assam University, Silchar, Assam, 788011, India
| | - Dileep R Yavagal
- Department of Neurology and Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
21
|
Huang J, Wu Y, Chai X, Wang S, Zhao Y, Hou Y, Ma Y, Chen S, Zhao S, Zhu X. β-Hydroxybutyric acid improves cognitive function in a model of heat stress by promoting adult hippocampal neurogenesis. STRESS BIOLOGY 2022; 2:57. [PMID: 37676574 PMCID: PMC10441921 DOI: 10.1007/s44154-022-00079-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/15/2022] [Indexed: 09/08/2023]
Abstract
Heat stress has multiple potential effects on the brain, such as neuroinflammation, neurogenesis defects, and cognitive impairment. β-hydroxybutyric acid (BHBA) has been demonstrated to play neuroprotective roles in various models of neurological diseases. In the present study, we investigated the efficacy of BHBA in alleviating heat stress-induced impairments of adult hippocampal neurogenesis and cognitive function, as well as the underlying mechanisms. Mice were exposed to 43 ℃ for 15 min for 14 days after administration with saline, BHBA, or minocycline. Here, we showed for the first time that BHBA normalized memory ability in the heat stress-treated mice and attenuated heat stress-impaired hippocampal neurogenesis. Consistently, BHBA noticeably improved the synaptic plasticity in the heat stress-treated hippocampal neurons by inhibiting the decrease of synapse-associated proteins and the density of dendritic spines. Moreover, BHBA inhibited the expression of cleaved caspase-3 by suppressing endoplasmic reticulum (ER) stress, and increased the expression of brain-derived neurotrophic factor (BDNF) in the heat stress-treated hippocampus by activating the protein kinase B (Akt)/cAMP response element binding protein (CREB) and methyl-CpG binding protein 2 (MeCP2) pathways. These findings indicate that BHBA is a potential agent for improving cognitive functions in heat stress-treated mice. The action may be mediated by ER stress, and Akt-CREB-BDNF and MeCP2 pathways to improve adult hippocampal neurogenesis and synaptic plasticity.
Collapse
Affiliation(s)
- Jian Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Yongji Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Xuejun Chai
- Department of Basic Medicine, Xi'an Medical University, Xi'an, Shaanxi, 710021, People's Republic of China
| | - Shuai Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Yongkang Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Yan Hou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China.
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, 712100, People's Republic of China.
| |
Collapse
|
22
|
Karoussiotis C, Sotiriou A, Polissidis A, Symeonof A, Papavranoussi-Daponte D, Nikoletopoulou V, Georgoussi Z. The κ-opioid receptor-induced autophagy is implicated in stress-driven synaptic alterations. Front Mol Neurosci 2022; 15:1039135. [PMID: 36466809 PMCID: PMC9709411 DOI: 10.3389/fnmol.2022.1039135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/26/2022] [Indexed: 08/29/2023] Open
Abstract
Recent evidence has shown that G protein-coupled receptors (GPCRs) are direct sensors of the autophagic machinery and opioid receptors regulate neuronal plasticity and neurotransmission with an as yet unclarified mechanism. Using in vitro and in vivo experimental approaches, this study aims to clarify the potential role of autophagy and κ-opioid receptor (κ-OR) signaling in synaptic alterations. We hereby demonstrate that the selective κ-OR agonist U50,488H, induces autophagy in a time-and dose-dependent manner in Neuro-2A cells stably expressing the human κ-OR by upregulating microtubule-associated protein Light Chain 3-II (LC3-II), Beclin 1 and Autophagy Related Gene 5 (ATG5). Pretreatment of neuronal cells with pertussis toxin blocked the above κ-OR-mediated cellular responses. Our molecular analysis also revealed a κ-OR-driven upregulation of becn1 gene through ERK1,2-dependent activation of the transcription factor CREB in Neuro-2A cells. Moreover, our studies demonstrated that sub-chronic U50,488H administration in mice causes profound increases of specific autophagic markers in the hippocampus with a concomitant decrease of several pre-and post-synaptic proteins, such as spinophilin, postsynaptic density protein 95 (PSD-95) and synaptosomal associated protein 25 (SNAP25). Finally, using acute stress, a stimulus known to increase the levels of the endogenous κ-OR ligand dynorphin, we are demonstrating that administration of the κ-ΟR selective antagonist, nor-binaltorphimine (norBNI), blocks the induction of autophagy and the stress-evoked reduction of synaptic proteins in the hippocampus. These findings provide novel insights about the essential role of autophagic machinery into the mechanisms through which κ-OR signaling regulates brain plasticity.
Collapse
Affiliation(s)
- Christos Karoussiotis
- Laboratory of Cellular Signaling and Molecular Pharmacology, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece
| | - Aggeliki Sotiriou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | - Alexia Polissidis
- Center for Clinical Research, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Alexandra Symeonof
- Laboratory of Cellular Signaling and Molecular Pharmacology, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece
| | - Danae Papavranoussi-Daponte
- Laboratory of Cellular Signaling and Molecular Pharmacology, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece
| | | | - Zafiroula Georgoussi
- Laboratory of Cellular Signaling and Molecular Pharmacology, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece
| |
Collapse
|
23
|
Estévez-Silva HM, Cuesto G, Romero N, Brito-Armas JM, Acevedo-Arozena A, Acebes Á, Marcellino DJ. Pridopidine Promotes Synaptogenesis and Reduces Spatial Memory Deficits in the Alzheimer's Disease APP/PS1 Mouse Model. Neurotherapeutics 2022; 19:1566-1587. [PMID: 35917088 PMCID: PMC9606189 DOI: 10.1007/s13311-022-01280-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2022] [Indexed: 10/16/2022] Open
Abstract
Sigma-1 receptor agonists have recently gained a great deal of interest due to their anti-amnesic, neuroprotective, and neurorestorative properties. Compounds such as PRE-084 or pridopidine (ACR16) are being studied as a potential treatment against cognitive decline associated with neurodegenerative disease, also to include Alzheimer's disease. Here, we performed in vitro experiments using primary neuronal cell cultures from rats to evaluate the abilities of ACR16 and PRE-084 to induce new synapses and spines formation, analyzing the expression of the possible genes and proteins involved. We additionally examined their neuroprotective properties against neuronal death mediated by oxidative stress and excitotoxicity. Both ACR16 and PRE-084 exhibited a concentration-dependent neuroprotective effect against NMDA- and H2O2-related toxicity, in addition to promoting the formation of new synapses and dendritic spines. However, only ACR16 generated dendritic spines involved in new synapse establishment, maintaining a more expanded activation of MAPK/ERK and PI3K/Akt signaling cascades. Consequently, ACR16 was also evaluated in vivo, and a dose of 1.5 mg/kg/day was administered intraperitoneally in APP/PS1 mice before performing the Morris water maze. ACR16 diminished the spatial learning and memory deficits observed in APP/PS1 transgenic mice via PI3K/Akt pathway activation. These data point to ACR16 as a pharmacological tool to prevent synapse loss and memory deficits associated with Alzheimer's disease, due to its neuroprotective properties against oxidative stress and excitotoxicity, as well as the promotion of new synapses and spines through a mechanism that involves AKT and ERK signaling pathways.
Collapse
Affiliation(s)
- Héctor M Estévez-Silva
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| | - Germán Cuesto
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - Ninovska Romero
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain
| | - José Miguel Brito-Armas
- Unidad de Investigación, Hospital Universitario de Canarias, ITB-ULL/CIBERNED, Tenerife, Spain
| | - Abraham Acevedo-Arozena
- Unidad de Investigación, Hospital Universitario de Canarias, ITB-ULL/CIBERNED, Tenerife, Spain
| | - Ángel Acebes
- Departamento de Ciencias Médicas Básicas, Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna, Tenerife, Spain.
| | | |
Collapse
|
24
|
Gu Z, Cao H, Zuo C, Huang Y, Miao J, Song Y, Yang Y, Zhu L, Wang F. TFEB in Alzheimer's disease: From molecular mechanisms to therapeutic implications. Neurobiol Dis 2022; 173:105855. [PMID: 36031168 DOI: 10.1016/j.nbd.2022.105855] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022] Open
Abstract
Alzheimer's disease (AD), an age-dependent neurodegenerative disorder, is the most prevalent neurodegenerative disease worldwide. The primary pathological hallmarks of AD are the deposition of β-amyloid plaques and neurofibrillary tangles. Autophagy, a pathway of clearing damaged organelles, macromolecular aggregates, and long-lived proteins via lysosomal degradation, has emerged as critical for proteostasis in the central nervous system (CNS). Studies have demonstrated that defective autophagy is strongly implicated in AD pathogenesis. Transcription factor EB (TFEB), a master transcriptional regulator of autophagy, enhances the expression of related genes that control autophagosome formation, lysosome function, and autophagic flux. The study of TFEB has greatly increased over the last decade, and the dysfunction of TFEB has been reported to be strongly associated with the pathogenesis of many neurodegenerative disorders, including AD. Here, we delineate the basic understanding of TFEB dysregulation involved in AD pathogenesis, highlighting the existing work that has been conducted on TFEB-mediated autophagy in neurons and other nonneuronal cells in the CNS. Additionally, we summarize the small molecule compounds that target TFEB-regulated autophagy involved in AD therapy. Our review may yield new insights into therapeutic approaches by targeting TFEB and provide a broadly applicable basis for the clinical treatment of AD.
Collapse
Affiliation(s)
- Zhongya Gu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Huan Cao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Chengchao Zuo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yaqi Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Jinfeng Miao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yu Song
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yuyan Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Liudi Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Furong Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Road, Wuhan 430030, Hubei, China.
| |
Collapse
|
25
|
Kissoondoyal A, Crawford DA. Prostaglandin E2 Increases Neurite Length and the Formation of Axonal Loops, and Regulates Cone Turning in Differentiating NE4C Cells Via PKA. Cell Mol Neurobiol 2022; 42:1385-1397. [PMID: 33389417 PMCID: PMC11421704 DOI: 10.1007/s10571-020-01029-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/09/2020] [Indexed: 12/13/2022]
Abstract
Prostaglandin E2 (PGE2) is a membrane-derived lipid signaling molecule important in neuronal development. Abnormal levels of PGE2, due to environmental insults prenatal development, have been linked to brain pathologies. We have previously shown that the addition of PGE2 to neuroectodermal (NE4C) stem cells affects early stages of neuronal differentiation (day 0-8) including increased stem cell motility, accelerated formation of neurospheres, and elevated calcium levels in growth cones. In this study, we further examine whether PGE2 can influence actin-dependent neuronal morphology in later stages (day 8-12) of NE4C cell differentiation. We show that exposure to PGE2 from the initiation of differentiation increased neurite length and the proportion of neurites that formed axonal loops. We also observed changes in the proportion of turning growth cones as the differentiation progressed, with a reduced likelihood of observing turning (or asymmetrical) growth cones on day 8 and increased odds on days 10 and 12. Moreover, we showed for the first time that the observed changes in cytoskeletal morphology were PGE2/PKA dependent. Interestingly, we also found that PGE2 decreased the total protein levels of the actin-bound form of spinophilin and increased levels of unbound PKA-phosphorylated ser94-spinophilin. Hence, we propose that exposure to PGE2 can destabilize the actin cytoskeleton at various stages of neuronal differentiation due to dissociation of ser94-spinophilin causing changes in neuronal morphology.
Collapse
Affiliation(s)
- Ashby Kissoondoyal
- School of Kinesiology and Health Science, York University, Toronto, ON, M3J 1P3, Canada
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada
| | - Dorota A Crawford
- School of Kinesiology and Health Science, York University, Toronto, ON, M3J 1P3, Canada.
- Neuroscience Graduate Diploma Program, York University, Toronto, ON, M3J 1P3, Canada.
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
26
|
A high-content neuron imaging assay demonstrates inhibition of prion disease-associated neurotoxicity by an anti-prion protein antibody. Sci Rep 2022; 12:9493. [PMID: 35680944 PMCID: PMC9184462 DOI: 10.1038/s41598-022-13455-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/09/2022] [Indexed: 11/20/2022] Open
Abstract
There is an urgent need to develop disease-modifying therapies to treat neurodegenerative diseases which pose increasing challenges to global healthcare systems. Prion diseases, although rare, provide a paradigm to study neurodegenerative dementias as similar disease mechanisms involving propagation and spread of multichain assemblies of misfolded protein ("prion-like" mechanisms) are increasingly recognised in the commoner conditions such as Alzheimer's disease. However, studies of prion disease pathogenesis in mouse models showed that prion propagation and neurotoxicity can be mechanistically uncoupled and in vitro assays confirmed that highly purified prions are indeed not directly neurotoxic. To aid development of prion disease therapeutics we have therefore developed a cell-based assay for the specific neurotoxicity seen in prion diseases rather than to simply assess inhibition of prion propagation. We applied this assay to examine an anti-prion protein mouse monoclonal antibody (ICSM18) known to potently cure prion-infected cells and to delay onset of prion disease in prion-infected mice. We demonstrate that whilst ICSM18 itself lacks inherent neurotoxicity in this assay, it potently blocks prion disease-associated neurotoxicity.
Collapse
|
27
|
Mai Le N, Li J. Ras-related C3 botulinum toxin substrate 1 role in Pathophysiology of Neurological diseases. BRAIN HEMORRHAGES 2022. [DOI: 10.1016/j.hest.2022.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
28
|
Taniguchi K, Yamamoto F, Amamo A, Tamaoka A, Sanjo N, Yokota T, Kametani F, Araki W. Amyloid-β oligomers interact with NMDA receptors containing GluN2B subunits and metabotropic glutamate receptor 1 in primary cortical neurons: relevance to the synapse pathology of Alzheimer’s disease. Neurosci Res 2022; 180:90-98. [DOI: 10.1016/j.neures.2022.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 01/22/2023]
|
29
|
Verdugo-Sivianes EM, Carnero A. SPINOPHILIN: a multiplayer tumor suppressor. Genes Dis 2022; 10:187-198. [PMID: 37013033 PMCID: PMC10066247 DOI: 10.1016/j.gendis.2021.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/24/2021] [Indexed: 02/07/2023] Open
Abstract
SPINOPHILIN (SPN, PPP1R9B or NEURABIN-2) is a multifunctional protein that regulates protein-protein interactions in different cell signaling pathways. SPN is also one of the regulatory subunits of protein phosphatase 1 (PP1), implicated in the dephosphorylation of retinoblastoma protein (pRB) during cell cycle. The SPN gene has been described as a tumor suppressor in different human tumor contexts, in which low levels of SPN are correlated with a higher grade and worse prognosis. In addition, mutations of the SPN protein have been reported in human tumors. Recently, an oncogenic mutation of SPN, A566V, was described, which affects both the SPN-PP1 interaction and the phosphatase activity of the holoenzyme, and promotes p53-dependent tumorigenesis by increasing the cancer stem cell (CSC) pool in breast tumors. Thus, the loss or mutation of SPN could be late events that promotes tumor progression by increasing the CSC pool and, eventually, the malignant behavior of the tumor.
Collapse
|
30
|
Rivera A, Suárez-Boomgaard D, Miguelez C, Valderrama-Carvajal A, Baufreton J, Shumilov K, Taupignon A, Gago B, Real MÁ. Dopamine D 4 Receptor Is a Regulator of Morphine-Induced Plasticity in the Rat Dorsal Striatum. Cells 2021; 11:31. [PMID: 35011592 PMCID: PMC8750869 DOI: 10.3390/cells11010031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 02/06/2023] Open
Abstract
Long-term exposition to morphine elicits structural and synaptic plasticity in reward-related regions of the brain, playing a critical role in addiction. However, morphine-induced neuroadaptations in the dorsal striatum have been poorly studied despite its key function in drug-related habit learning. Here, we show that prolonged treatment with morphine triggered the retraction of the dendritic arbor and the loss of dendritic spines in the dorsal striatal projection neurons (MSNs). In an attempt to extend previous findings, we also explored whether the dopamine D4 receptor (D4R) could modulate striatal morphine-induced plasticity. The combined treatment of morphine with the D4R agonist PD168,077 produced an expansion of the MSNs dendritic arbors and restored dendritic spine density. At the electrophysiological level, PD168,077 in combination with morphine altered the electrical properties of the MSNs and decreased their excitability. Finally, results from the sustantia nigra showed that PD168,077 counteracted morphine-induced upregulation of μ opioid receptors (MOR) in striatonigral projections and downregulation of G protein-gated inward rectifier K+ channels (GIRK1 and GIRK2) in dopaminergic cells. The present results highlight the key function of D4R modulating morphine-induced plasticity in the dorsal striatum. Thus, D4R could represent a valuable pharmacological target for the safety use of morphine in pain management.
Collapse
Affiliation(s)
- Alicia Rivera
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
| | - Diana Suárez-Boomgaard
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
| | - Cristina Miguelez
- Department of Pharmacology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Alejandra Valderrama-Carvajal
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
| | - Jérôme Baufreton
- Institut des Maladies Neurodegeneratives, Université de Bordeaux, UMR 5293, 33000 Bordeaux, France; (J.B.); (A.T.)
- Institut des Maladies Neurodegeneratives, CNRS, UMR 5293, 33000 Bordeaux, France
| | - Kirill Shumilov
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
- School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Anne Taupignon
- Institut des Maladies Neurodegeneratives, Université de Bordeaux, UMR 5293, 33000 Bordeaux, France; (J.B.); (A.T.)
- Institut des Maladies Neurodegeneratives, CNRS, UMR 5293, 33000 Bordeaux, France
| | - Belén Gago
- Facultad de Medicina, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain;
| | - M. Ángeles Real
- Facultad de Ciencias, Instituto de Investigación Biomédica, Universidad de Málaga, 29071 Málaga, Spain; (D.S.-B.); (A.V.-C.); (K.S.); (M.Á.R.)
| |
Collapse
|
31
|
Li DC, Hinton EA, Gourley SL. Persistent behavioral and neurobiological consequences of social isolation during adolescence. Semin Cell Dev Biol 2021; 118:73-82. [PMID: 34112579 PMCID: PMC8434983 DOI: 10.1016/j.semcdb.2021.05.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/05/2021] [Accepted: 05/14/2021] [Indexed: 12/15/2022]
Abstract
Meaningful social interactions are a fundamental human need, the lack of which can pose serious risks to an individual's physical and mental health. Across species, peer-oriented social behaviors are dramatically reshaped during adolescence, a developmental period characterized by dynamic changes in brain structure and function as individuals transition into adulthood. Thus, the experience of social isolation during this critical developmental stage may be especially pernicious, as it could permanently derail typical neurobiological processes that are necessary for establishing adaptive adult behaviors. The purpose of this review is to summarize investigations in which rodents were isolated during adolescence, then re-housed in typical social groups prior to testing, thus allowing the investigators to resolve the long-term consequences of social adversity experienced during adolescent sensitive periods, despite subsequent normalization of the social environment. Here, we discuss alterations in social, anxiety-like, cognitive, and decision-making behaviors in previously isolated adult rodents. We then explore corresponding neurobiological findings, focusing on the prefrontal cortex, including changes in synaptic densities and protein levels, white matter and oligodendrocyte function, and neuronal physiology. Made more urgent by the recent wave of social deprivation resulting from the COVID-19 pandemic, especially amongst school-aged adolescents, understanding the mechanisms by which even transient social adversity can negatively impact brain function across the lifespan is of paramount importance.
Collapse
Affiliation(s)
- Dan C Li
- Medical Scientist Training Program, Emory University School of Medicine, USA; Graduate Training Program in Neuroscience, Emory University, USA; Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, USA; Yerkes National Primate Research Center, Emory University, USA.
| | - Elizabeth A Hinton
- Graduate Training Program in Neuroscience, Emory University, USA; Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, USA; Yerkes National Primate Research Center, Emory University, USA
| | - Shannon L Gourley
- Graduate Training Program in Neuroscience, Emory University, USA; Department of Pediatrics, Children's Healthcare of Atlanta, Emory University School of Medicine, USA; Yerkes National Primate Research Center, Emory University, USA.
| |
Collapse
|
32
|
Kissoondoyal A, Rai-Bhogal R, Crawford DA. Abnormal dendritic morphology in the cerebellum of cyclooxygenase-2 - knockin mice. Eur J Neurosci 2021; 54:6355-6373. [PMID: 34510613 DOI: 10.1111/ejn.15454] [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: 06/05/2021] [Accepted: 09/02/2021] [Indexed: 11/28/2022]
Abstract
Prostaglandin E2 (PGE2) is a bioactive signalling molecule metabolized from the phospholipid membranes by the enzymatic activity of cycloxygenase-2 (COX-2). In the developing brain, COX-2 constitutively regulates the production of PGE2, which is important in neuronal development. However, abnormal COX-2/PGE2 signalling has been linked to neurodevelopmental disorders including autism spectrum disorders (ASDs). We have previously demonstrated that COX-2- -KI mice show autism-related behaviours including social deficits, repetitive behaviours and anxious behaviours. COX-2-deficient mice also have deficits in pathways involved in synaptic transmission and dendritic spine formation. In this study, we use a Golgi-COX staining method to examine sex-dependent differences in dendritic and dendritic spine morphology in neurons of COX-2- -KI mice cerebellum compared with wild-type (WT) matched controls at postnatal day 25 (P25). We show that COX-2- -KI mice have increased dendritic arborization closer to the cell soma and increased dendritic looping. We also observed a sex-dependent effect of the COX-2- -KI on dendritic thickness, dendritic spine density, dendritic spine morphology, and the expression of β-actin and the actin-binding protein spinophilin. Our findings show that changes in COX-2/PGE2 signalling lead to impaired morphology of dendrites and dendritic spines in a sex-dependant manner and may contribute the pathology of the cerebellum seen in individuals with ASD. This study provides further evidence that the COX-2- -KI mouse model can be used to study a subset of ASD pathologies.
Collapse
Affiliation(s)
- Ashby Kissoondoyal
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,Neuroscience Graduate Diploma Program, York University, Toronto, Ontario, Canada
| | - Ravneet Rai-Bhogal
- Neuroscience Graduate Diploma Program, York University, Toronto, Ontario, Canada.,Department of Biology, York University, Toronto, Ontario, Canada
| | - Dorota A Crawford
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada.,Neuroscience Graduate Diploma Program, York University, Toronto, Ontario, Canada.,Department of Biology, York University, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Urbinati C, Cosentino L, Germinario EAP, Valenti D, Vigli D, Ricceri L, Laviola G, Fiorentini C, Vacca RA, Fabbri A, De Filippis B. Treatment with the Bacterial Toxin CNF1 Selectively Rescues Cognitive and Brain Mitochondrial Deficits in a Female Mouse Model of Rett Syndrome Carrying a MeCP2-Null Mutation. Int J Mol Sci 2021; 22:6739. [PMID: 34201747 PMCID: PMC8269120 DOI: 10.3390/ijms22136739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/25/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
Rett syndrome (RTT) is a rare neurological disorder caused by mutations in the X-linked MECP2 gene and a major cause of intellectual disability in females. No cure exists for RTT. We previously reported that the behavioural phenotype and brain mitochondria dysfunction are widely rescued by a single intracerebroventricular injection of the bacterial toxin CNF1 in a RTT mouse model carrying a truncating mutation of the MeCP2 gene (MeCP2-308 mice). Given the heterogeneity of MECP2 mutations in RTT patients, we tested the CNF1 therapeutic efficacy in a mouse model carrying a null mutation (MeCP2-Bird mice). CNF1 selectively rescued cognitive defects, without improving other RTT-related behavioural alterations, and restored brain mitochondrial respiratory chain complex activity in MeCP2-Bird mice. To shed light on the molecular mechanisms underlying the differential CNF1 effects on the behavioural phenotype, we compared treatment effects on relevant signalling cascades in the brain of the two RTT models. CNF1 provided a significant boost of the mTOR activation in MeCP2-308 hippocampus, which was not observed in the MeCP2-Bird model, possibly explaining the differential effects of CNF1. These results demonstrate that CNF1 efficacy depends on the mutation beared by MeCP2-mutated mice, stressing the need of testing potential therapeutic approaches across RTT models.
Collapse
Affiliation(s)
- Chiara Urbinati
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Livia Cosentino
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Elena Angela Pia Germinario
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.A.P.G.); (A.F.)
| | - Daniela Valenti
- Bioenergetics and Molecular Biotechnologies, Institute of Biomembranes, National Council of Research, 70126 Bari, Italy; (D.V.); (R.A.V.)
| | - Daniele Vigli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Laura Ricceri
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Giovanni Laviola
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| | - Carla Fiorentini
- Association for Research on Integrative Oncology Therapies (ARTOI), 00165 Rome, Italy;
| | - Rosa Anna Vacca
- Bioenergetics and Molecular Biotechnologies, Institute of Biomembranes, National Council of Research, 70126 Bari, Italy; (D.V.); (R.A.V.)
| | - Alessia Fabbri
- Department of Cardiovascular, Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (E.A.P.G.); (A.F.)
| | - Bianca De Filippis
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (C.U.); (L.C.); (D.V.); (L.R.); (G.L.)
| |
Collapse
|
34
|
Liaci C, Camera M, Caslini G, Rando S, Contino S, Romano V, Merlo GR. Neuronal Cytoskeleton in Intellectual Disability: From Systems Biology and Modeling to Therapeutic Opportunities. Int J Mol Sci 2021; 22:ijms22116167. [PMID: 34200511 PMCID: PMC8201358 DOI: 10.3390/ijms22116167] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/25/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Intellectual disability (ID) is a pathological condition characterized by limited intellectual functioning and adaptive behaviors. It affects 1–3% of the worldwide population, and no pharmacological therapies are currently available. More than 1000 genes have been found mutated in ID patients pointing out that, despite the common phenotype, the genetic bases are highly heterogeneous and apparently unrelated. Bibliomic analysis reveals that ID genes converge onto a few biological modules, including cytoskeleton dynamics, whose regulation depends on Rho GTPases transduction. Genetic variants exert their effects at different levels in a hierarchical arrangement, starting from the molecular level and moving toward higher levels of organization, i.e., cell compartment and functions, circuits, cognition, and behavior. Thus, cytoskeleton alterations that have an impact on cell processes such as neuronal migration, neuritogenesis, and synaptic plasticity rebound on the overall establishment of an effective network and consequently on the cognitive phenotype. Systems biology (SB) approaches are more focused on the overall interconnected network rather than on individual genes, thus encouraging the design of therapies that aim to correct common dysregulated biological processes. This review summarizes current knowledge about cytoskeleton control in neurons and its relevance for the ID pathogenesis, exploiting in silico modeling and translating the implications of those findings into biomedical research.
Collapse
Affiliation(s)
- Carla Liaci
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Mattia Camera
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Giovanni Caslini
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Simona Rando
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
| | - Salvatore Contino
- Department of Engineering, University of Palermo, Viale delle Scienze Ed. 8, 90128 Palermo, Italy;
| | - Valentino Romano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze Ed. 16, 90128 Palermo, Italy;
| | - Giorgio R. Merlo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (C.L.); (M.C.); (G.C.); (S.R.)
- Correspondence: ; Tel.: +39-0116706449; Fax: +39-0116706432
| |
Collapse
|
35
|
Lo LHY, Dong R, Lyu Q, Lai KO. The Protein Arginine Methyltransferase PRMT8 and Substrate G3BP1 Control Rac1-PAK1 Signaling and Actin Cytoskeleton for Dendritic Spine Maturation. Cell Rep 2021; 31:107744. [PMID: 32521269 DOI: 10.1016/j.celrep.2020.107744] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/01/2020] [Accepted: 05/18/2020] [Indexed: 01/25/2023] Open
Abstract
Excitatory synapses of neurons are located on dendritic spines. Spine maturation is essential for the stability of synapses and memory consolidation, and overproduction of the immature filopodia is associated with brain disorders. The structure and function of synapses can be modulated by protein post-translational modification (PTM). Arginine methylation is a major PTM that regulates chromatin structure, transcription, and splicing within the nucleus. Here we find that the protein arginine methyltransferase PRMT8 is present at neuronal synapses and its expression is upregulated in the hippocampus when dendritic spine maturation occurs. Depletion of PRMT8 leads to overabundance of filopodia and mis-localization of excitatory synapses. Mechanistically, PRMT8 promotes dendritic spine morphology through methylation of the dendritic RNA-binding protein G3BP1 and suppression of the Rac1-PAK1 signaling pathway to control synaptic actin dynamics. Our findings unravel arginine methylation as a crucial regulatory mechanism for actin cytoskeleton during synapse development.
Collapse
Affiliation(s)
- Louisa Hoi-Ying Lo
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Rui Dong
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Quanwei Lyu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Kwok-On Lai
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
36
|
Verdugo-Sivianes EM, Carnero A. Role of the Holoenzyme PP1-SPN in the Dephosphorylation of the RB Family of Tumor Suppressors During Cell Cycle. Cancers (Basel) 2021; 13:cancers13092226. [PMID: 34066428 PMCID: PMC8124259 DOI: 10.3390/cancers13092226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cell cycle progression is highly regulated by modulating the phosphorylation status of retinoblastoma (RB) family proteins. This process is controlled by a balance in the action of kinases, such as the complexes formed by cyclin-dependent kinases (CDKs) and cyclins, and phosphatases, mainly the protein phosphatase 1 (PP1). However, while the phosphorylation of the RB family has been largely studied, its dephosphorylation is less known. Recently, the PP1-Spinophilin (SPN) holoenzyme has been described as the main phosphatase responsible for the dephosphorylation of RB proteins during the G0/G1 transition and at the end of G1. Here, we describe the regulation of the phosphorylation status of RB family proteins, giving importance not only to their inactivation by phosphorylation but also to their dephosphorylation to restore the cell cycle. Abstract Cell cycle progression is highly regulated by modulating the phosphorylation status of the retinoblastoma protein (pRB) and the other two members of the RB family, p107 and p130. This process is controlled by a balance in the action of kinases, such as the complexes formed by cyclin-dependent kinases (CDKs) and cyclins, and phosphatases, mainly the protein phosphatase 1 (PP1). However, while the phosphorylation of the RB family has been largely studied, its dephosphorylation is less known. Phosphatases are holoenzymes formed by a catalytic subunit and a regulatory protein with substrate specificity. Recently, the PP1-Spinophilin (SPN) holoenzyme has been described as the main phosphatase responsible for the dephosphorylation of RB proteins during the G0/G1 transition and at the end of G1. Moreover, SPN has been described as a tumor suppressor dependent on PP1 in lung and breast tumors, where it promotes tumorigenesis by increasing the cancer stem cell pool. Therefore, a connection between the cell cycle and stem cell biology has also been proposed via SPN/PP1/RB proteins.
Collapse
Affiliation(s)
- Eva M. Verdugo-Sivianes
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013 Seville, Spain;
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocio, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Avda. Manuel Siurot s/n, 41013 Seville, Spain;
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-955-92-31-11
| |
Collapse
|
37
|
Atrophy of lacunosum moleculare layer is important for learning and memory in APP/PS1 transgenic mice. Neuroreport 2021; 32:596-602. [PMID: 33850085 DOI: 10.1097/wnr.0000000000001639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Changes in the hippocampus are closely associated with learning and memory in Alzheimer's disease; however, it is not clear which morphological and cellular and subcellular changes are essential for learning and memory. Here, we accurately quantitatively studied the hippocampal microstructure changes in Alzheimer's disease model mice and analyzed the relationship between the hippocampal microstructure changes and learning and memory. Ten-month-old male APP/PS1 transgenic mice and age-matched nontransgenic littermate mice were randomly selected. The spatial learning and memory abilities were assessed using the Morris water maze. The volumes of each layer and numbers of neurons, dendritic spines and oligodendrocytes in the hippocampal subregions were investigated using unbiased stereological techniques. The APP/PS1 transgenic mice showed a decline in hippocampus-dependent spatial learning and memory abilities, smaller volumes of each layer (other than stratum radiatum) and fewer numbers of neurons, dendritic spine synapses and mature oligodendrocytes in the hippocampal subregions than nontransgenic mice. In particular, the decline of spatial learning ability was significantly correlated with the atrophy of lacunosum moleculare layer (LMol) and the decrease of hippocampal neurons and mature oligodendrocytes rather than dendritic spines. The CA1-3 fields (including LMol) atrophy was significantly correlated with the decrease both of neurons, dendritic spines and mature oligodendrocytes. However, the dentate gyrus atrophy was significantly correlated with the decrease of neurons and mature oligodendrocytes rather than dendritic spines. The loss of neurons, dendritic spines synapses and mature oligodendrocytes together caused the LMol atrophy and then led to a decline in hippocampus-dependent spatial learning ability in mice with Alzheimer's disease.
Collapse
|
38
|
Xie YH, Zhou CN, Liang X, Tang J, Yang CM, Luo YM, Chao FL, Jiang L, Wang J, Qi YQ, Zhu PL, Li Y, Xiao K, Tang Y. Anti-Lingo-1 antibody ameliorates spatial memory and synapse loss induced by chronic stress. J Comp Neurol 2021; 529:1571-1583. [PMID: 32965038 DOI: 10.1002/cne.25038] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 07/22/2020] [Accepted: 09/08/2020] [Indexed: 11/10/2022]
Abstract
Chronic stress can induce cognitive impairment, and synapse number was significantly decreased in the hippocampus of rats suffering from chronic stress. Lingo-1 is a potent negative regulator of axonal outgrowth and synaptic plasticity. In the current study, the effects of anti-Lingo-1 antibody on the spatial learning and memory abilities and hippocampal synapses of stressed rats were investigated. After 4 weeks of stress exposure, the model group was randomly divided into a chronic stress group and an anti-Lingo-1 group. Then, the anti-Lingo-1 group rats were treated with anti-Lingo-1 antibody (8 mg/kg) for 3 weeks. The effects of anti-Lingo-1 antibody on the spatial learning and memory abilities were investigated with the Morris water maze test. Immunohistological staining and an unbiased stereological method were used to estimate the total number of dendritic spine synapses in the hippocampus. At the behavioral level, after 3 weeks of treatment, the anti-Lingo-1 group rats displayed significantly more platform location crossings in the Morris water maze test than the chronic stress group rats. Anti-Lingo-1 significantly prevented the declines in dendritic spine synapses and postsynaptic density protein-95 (PSD-95) expression in the dentate gyrus and the CA1 and CA3 regions of the hippocampus. The present results indicated that anti-Lingo-1 antibody may be a safe and effective drug for alleviating memory impairment in rats after chronic stress and protecting synapses in the hippocampus of stressed rats.
Collapse
Affiliation(s)
- Yu-Han Xie
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Chun-Ni Zhou
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Xin Liang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Jing Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Chun-Mao Yang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Yan-Min Luo
- Department of Physiology, Chongqing Medical University, Chongqing, P.R. China
| | - Feng-Lei Chao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Lin Jiang
- Lab Teaching & Management Center, Chongqing Medical University, Chongqing, P.R. China
| | - Jin Wang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Ying-Qiang Qi
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Pei-Lin Zhu
- Department of Physiology, Chongqing Medical University, Chongqing, P.R. China
| | - Yue Li
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Kai Xiao
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| | - Yong Tang
- Department of Histology and Embryology, Chongqing Medical University, Chongqing, P.R. China.,Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
39
|
Regulation of Synaptic Transmission and Plasticity by Protein Phosphatase 1. J Neurosci 2021; 41:3040-3050. [PMID: 33827970 DOI: 10.1523/jneurosci.2026-20.2021] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 01/22/2023] Open
Abstract
Protein phosphatases, by counteracting protein kinases, regulate the reversible phosphorylation of many substrates involved in synaptic plasticity, a cellular model for learning and memory. A prominent phosphatase regulating synaptic plasticity and neurologic disorders is the serine/threonine protein phosphatase 1 (PP1). PP1 has three isoforms (α, β, and γ, encoded by three different genes), which are regulated by a vast number of interacting subunits that define their enzymatic substrate specificity. In this review, we discuss evidence showing that PP1 regulates synaptic transmission and plasticity, as well as presenting novel models of PP1 regulation suggested by recent experimental evidence. We also outline the required targeting of PP1 by neurabin and spinophilin to achieve substrate specificity at the synapse to regulate AMPAR and NMDAR function. We then highlight the role of inhibitor-2 in regulating PP1 function in plasticity, including its positive regulation of PP1 function in vivo in memory formation. We also discuss the distinct function of the three PP1 isoforms in synaptic plasticity and brain function, as well as briefly discuss the role of inhibitory phosphorylation of PP1, which has received recent emphasis in the regulation of PP1 activity in neurons.
Collapse
|
40
|
Martínez-Torres N, González-Tapia D, Flores-Soto M, Vázquez-Hernández N, Salgado-Ceballos H, González-Burgos I. Spinogenesis in spinal cord motor neurons following pharmacological lesions to the rat motor cortex. NEUROLOGÍA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.nrleng.2017.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
41
|
Wang JL, Wang Y, Sun W, Yu Y, Wei N, Du R, Yang Y, Liang T, Wang XL, Ou CH, Chen J. Spinophilin modulates pain through suppressing dendritic spine morphogenesis via negative control of Rac1-ERK signaling in rat spinal dorsal horn. Neurobiol Dis 2021; 152:105302. [PMID: 33609640 DOI: 10.1016/j.nbd.2021.105302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/07/2021] [Accepted: 02/14/2021] [Indexed: 01/07/2023] Open
Abstract
Both spinophilin (SPN, also known as neurabin 2) and Rac1 (a member of Rho GTPase family) are believed to play key roles in dendritic spine (DS) remodeling and spinal nociception. However, how SPN interacts with Rac1 in the above process is unknown. Here, we first demonstrated natural existence of SPN-protein phosphatase 1-Rac1 complex in the spinal dorsal horn (DH) neurons by both double immunofluorescent labeling and co-immunoprecipitation, then the effects of SPN over-expression and down-regulation on mechanical and thermal pain sensitivity, GTP-bound Rac1-ERK signaling activity, and spinal DS density were studied. Over-expression of SPN in spinal neurons by intra-DH pAAV-CMV-SPN-3FLAG could block both mechanical and thermal pain hypersensitivity induced by intraplantar bee venom injection, however it had no effect on the basal pain sensitivity. Over-expression of SPN also resulted in a significant decrease in GTP-Rac1-ERK activities, relative to naive and irrelevant control (pAAV-MCS). In sharp contrast, knockdown of SPN in spinal neurons by intra-DH pAAV-CAG-eGFP-U6-shRNA[SPN] produced both pain hypersensitivity and dramatic elevation of GTP-Rac1-ERK activities, relative to naive and irrelevant control (pAAV-shRNA [NC]). Moreover, knockdown of SPN resulted in increase in DS density while over-expression of it had no such effect. Collectively, SPN is likely to serve as a regulator of Rac1 signaling to suppress DS morphogenesis via negative control of GTP-bound Rac1-ERK activities at postsynaptic component in rat DH neurons wherein both mechanical and thermal pain sensitivity are controlled.
Collapse
Affiliation(s)
- Jiang-Lin Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Yan Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Wei Sun
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Yang Yu
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Na Wei
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Rui Du
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Yan Yang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Ting Liang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Xiao-Liang Wang
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China
| | - Ce-Hua Ou
- Department of Pain Management, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, PR China
| | - Jun Chen
- Institute for Biomedical Sciences of Pain, Tangdu Hospital, The Fourth Military Medical University, Xi'an 710038, Shaanxi Province, PR China; Key Laboratory of Brain Stress and Behavior, People's Liberation Army, Xi'an 710038, PR China.
| |
Collapse
|
42
|
Brocos-Mosquera I, Gabilondo AM, Meana JJ, Callado LF, Erdozain AM. Spinophilin expression in postmortem prefrontal cortex of schizophrenic subjects: Effects of antipsychotic treatment. Eur Neuropsychopharmacol 2021; 42:12-21. [PMID: 33257116 DOI: 10.1016/j.euroneuro.2020.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 10/22/2022]
Abstract
Schizophrenia has been associated with alterations in neurotransmission and synaptic dysfunction. Spinophilin is a multifunctional scaffold protein that modulates excitatory synaptic transmission and dendritic spine morphology. Spinophilin can also directly interact with and regulate several receptors for neurotransmitters, such as dopamine D2 receptors, which play a role in the pathophysiology of schizophrenia and are targets of antipsychotics. Several studies have thus suggested an implication of spinophilin in schizophrenia. In the present study spinophilin protein expression was determined by western blot in the postmortem dorsolateral prefrontal cortex of 24 subjects with schizophrenia (12 antipsychotic-free and 12 antipsychotic-treated subjects) and 24 matched controls. Experiments were performed in synaptosomal membranes (SPM) and in postsynaptic density fractions (PSD). As previously reported, two specific bands for this protein were observed: an upper 120-130 kDa band and a lower 80-95 kDa band. The spinophilin lower band showed a significant decrease in schizophrenia subjects compared to matched controls, both in SPM and PSD fractions (-15%, p = 0.007 and -15%, p = 0.039, respectively). When schizophrenia subjects were divided by the presence or absence of antipsychotics in blood at death, the lower band showed a significant decrease in antipsychotic-treated schizophrenia subjects (-24%, p = 0.003 for SPM and -26%, p = 0.014 for PSD), but not in antipsychotic-free subjects, compared to their matched controls. These results suggest that antipsychotics could produce alterations in spinophilin expression that do not seem to be related to schizophrenia per se. These changes may underlie some of the side effects of antipsychotics.
Collapse
Affiliation(s)
- Iria Brocos-Mosquera
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain
| | - Ane M Gabilondo
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - J Javier Meana
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Luis F Callado
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain; Biocruces Bizkaia Health Research Institute, Barakaldo, Bizkaia, Spain
| | - Amaia M Erdozain
- Department of Pharmacology, University of the Basque Country, UPV/EHU, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Spain.
| |
Collapse
|
43
|
Glausier JR, Datta D, Fish KN, Chung DW, Melchitzky DS, Lewis DA. Laminar Differences in the Targeting of Dendritic Spines by Cortical Pyramidal Neurons and Interneurons in Human Dorsolateral Prefrontal Cortex. Neuroscience 2021; 452:181-191. [PMID: 33212224 PMCID: PMC7770119 DOI: 10.1016/j.neuroscience.2020.10.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/05/2020] [Accepted: 10/21/2020] [Indexed: 01/05/2023]
Abstract
Activation of specific neural circuits in different layers of the primate dorsolateral prefrontal cortex (DLPFC) is essential for working memory, a core cognitive function. Recurrent excitation between pyramidal neurons in middle and deep layers of the DLPFC contributes to the laminar-specific activity associated with different working memory subprocesses. Excitation between cortical pyramidal neurons is mediated by glutamatergic synapses on dendritic spines, but whether the relative abundance of spines receiving cortical inputs differs between middle and deep cortical layers in human DLPFC is unknown. Additionally, GABAergic inputs to spines sculpt pyramidal neuron activity. Whether dendritic spines that receive a glutamatergic input from a cortical pyramidal neuron are targeted by GABAergic interneurons in the human DLPFC is unknown. Using triple-label fluorescence confocal microscopy, we found that 1) the density of spines receiving an input from a cortical pyramidal neuron is greater in the middle than in the deep laminar zone, 2) dendritic spines dually innervated by a cortical pyramidal neuron and an interneuron are present in the human DLPFC, and 3) the density of spines dually innervated by a cortical pyramidal neuron and an interneuron is also greater in the middle than in the deep laminar zone. Ultrastructural analyses support the presence of spines that receive a cortical pyramidal neuron synapse and an interneuron synapse in human and monkey DLPFC. These data support the notion that the DLPFC middle laminar zone is particularly endowed with a microcircuit structure that supports the gating, integrating and fine-tuning of synaptic information in recurrent excitatory microcircuits.
Collapse
Affiliation(s)
- Jill R Glausier
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA
| | - Dibyadeep Datta
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA; Department of Neuroscience, Yale University, Sterling Hall of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Kenneth N Fish
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA
| | - Daniel W Chung
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA
| | - Darlene S Melchitzky
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA
| | - David A Lewis
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Biomedical Science Tower W1654, 3811 O'Hara Street, Pittsburgh, PA 15213, USA; Department of Neuroscience, University of Pittsburgh, A210 Langley Hall, Pittsburgh, PA 15260, USA.
| |
Collapse
|
44
|
Platholi J, Hemmings HC. Modulation of dendritic spines by protein phosphatase-1. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2020; 90:117-144. [PMID: 33706930 DOI: 10.1016/bs.apha.2020.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Protein phosphatase-1 (PP-1), a highly conserved multifunctional serine/threonine phosphatase, is enriched in dendritic spines where it plays a major role in modulating excitatory synaptic activity. In addition to established functions in spine maturation and development, multi-subunit holoenzyme forms of PP-1 modulate higher-order cognitive functions such learning and memory. Mechanisms involved in regulating PP-1 activity and localization in spines include interactions with neurabin and spinophilin, structurally related synaptic scaffolding proteins associated with the actin cytoskeleton. Since PP-1 is a critical element in synaptic development, signaling, and plasticity, alterations in PP-1 signaling in dendritic spines are implicated in various neurological and psychiatric disorders. The effects of PP-1 depend on its isoform-specific association with regulatory proteins and activation of downstream signaling pathways. Here we review the role of PP-1 and its binding proteins neurabin and spinophilin in both developing and established dendritic spines, as well as some of the disorders that result from its dysregulation.
Collapse
Affiliation(s)
- Jimcy Platholi
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, United States
| | - Hugh C Hemmings
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, United States; Department of Pharmacology, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
45
|
Sbai O, Soussi R, Bole A, Khrestchatisky M, Esclapez M, Ferhat L. The actin binding protein α-actinin-2 expression is associated with dendritic spine plasticity and migrating granule cells in the rat dentate gyrus following pilocarpine-induced seizures. Exp Neurol 2020; 335:113512. [PMID: 33098872 DOI: 10.1016/j.expneurol.2020.113512] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
α-actinin-2 (α-actn-2) is an F-actin-crosslinking protein, localized in dendritic spines. In vitro studies suggested that it is involved in spinogenesis, morphogenesis, actin organization, cell migration and anchoring of the NR1 subunit of the N-methyl-D-aspartate (NMDA) receptors in dendritic spines. However, little is known regarding its function in vivo. We examined the levels of α-actn-2 expression within the dentate gyrus (DG) during the development of chronic limbic seizures (epileptogenesis) induced by pilocarpine in rats. In this model, plasticity of the DG glutamatergic granule cells including spine loss, spinogenesis, morphogenesis, neo-synaptogenesis, aberrant migration, and alterations of NMDA receptors have been well characterized. We showed that α-actn-2 immunolabeling was reduced in the inner molecular layer at 1-2 weeks post-status epilepticus (SE), when granule cell spinogenesis and morphogenesis occur. This low level persisted at the chronic stage when new functional synapses are established. This decreased of α-actn-2 protein is concomitant with the recovery of drebrin A (DA), another actin-binding protein, at the chronic stage. Indeed, we demonstrated in cultured cells that in contrast to DA, α-actn-2 did not protect F-actin destabilization and DA inhibited α-actn-2 binding to F-actin. Such alteration could affect the anchoring of NR1 in dendritic spines. Furthermore, we showed that the expression of α-actn-2 and NR1 are co-down-regulated in membrane fractions of pilocarpine animals at chronic stage. Last, we showed that α-actn-2 is expressed in migrating newly born granule cells observed within the hilus of pilocarpine-treated rats. Altogether, our results suggest that α-actn-2 is not critical for the structural integrity and stabilization of granule cell dendritic spines. Instead, its expression is regulated when spinogenesis and morphogenesis occur and within migrating granule cells. Our data also suggest that the balance between α-actn-2 and DA expression levels may modulate NR1 anchoring within dendritic spines.
Collapse
Affiliation(s)
- Oualid Sbai
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Rabia Soussi
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | - Angélique Bole
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France
| | | | - Monique Esclapez
- Aix-Marseille Univ, INSERM, INS, Inst Neurosci Syst, Marseille, France
| | - Lotfi Ferhat
- Aix-Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.
| |
Collapse
|
46
|
Gao S, Zhang T, Jin L, Liang D, Fan G, Song Y, Lucassen PJ, Yu R, Swaab DF. CAPON Is a Critical Protein in Synaptic Molecular Networks in the Prefrontal Cortex of Mood Disorder Patients and Contributes to Depression-Like Behavior in a Mouse Model. Cereb Cortex 2020; 29:3752-3765. [PMID: 30307500 DOI: 10.1093/cercor/bhy254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 08/16/2018] [Indexed: 12/16/2022] Open
Abstract
Aberrant regulation and activity of synaptic proteins may cause synaptic pathology in the prefrontal cortex (PFC) of mood disorder patients. Carboxy-terminal PDZ ligand of NOS1 (CAPON) is a critical scaffold protein linked to synaptic proteins like nitric oxide synthase 1, synapsins. We hypothesized that CAPON is altered together with its interacting synaptic proteins in the PFC in mood disorder patients and may contribute to depression-like behaviors in mice subjected to chronic unpredictable mild stress (CUMS). Here, we found that CAPON-immunoreactivity (ir) was significantly increased in the dorsolateral PFC (DLPFC) and anterior cingulate cortex in major depressive disorder (MDD), which was accompanied by an upregulation of spinophilin-ir and a downregulation of synapsin-ir. The increases in CAPON and spinophilin and the decrease in synapsin in the DLPFC of MDD patients were also seen in the PFC of CUMS mice. CAPON-ir positively correlated with spinophilin-ir (but not with synapsin-ir) in mood disorder patients. CAPON colocalized with spinophilin in the DLPFC of MDD patients and interacted with spinophilin in human brain. Viral-mediated CAPON downregulation in the medial PFC notably reversed the depression-like behaviors in the CUMS mice. These data suggest that CAPON may contribute to aspects of depressive behavior, possibly as an interacting protein for spinophilin in the PFC.
Collapse
Affiliation(s)
- Shangfeng Gao
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Tong Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Lei Jin
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Dong Liang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Guangwei Fan
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Yunnong Song
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, XH, Amsterdam, The Netherlands
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Dick F Swaab
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Okino E, Morita S, Hoshikawa Y, Tsukahara S. The glutamatergic system in the preoptic area is involved in the retention of maternal behavior in maternally experienced female rats. Psychoneuroendocrinology 2020; 120:104792. [PMID: 32653768 DOI: 10.1016/j.psyneuen.2020.104792] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/27/2022]
Abstract
Maternally experienced female rats show high maternal behavior performance for a long time after acquisition of maternal experience, although the mechanisms responsible for the retention of maternal behavior are not well understood. The medial preoptic area (MPOA) plays an important role in the onset and maintenance of maternal behavior in female rats. We aimed to determine whether maternal experience affects the glutamatergic system in the MPOA for the retention of maternal behavior in female rats. First, to determine the effects of maternal experience in the postpartum period on dendritic spines, which are the postsynaptic component of excitatory glutamatergic neurotransmission, we examined the number of dendritic spines on MPOA neurons of primiparous mothers that had experienced mothering until weaning (sufficiently experienced mothers) and of primiparous mothers that were separated from their pups on the day of parturition (insufficiently experienced mothers). The number of mushroom spines, but not other types of spine, was significantly greater in the sufficiently experienced mothers compared with that in the insufficiently experienced mothers. Next, to determine the effects of maternal experience in the postpartum period on the expression of ionotropic glutamate receptors, we measured the mRNA levels of AMPA receptor subunits (GluA1-A4) and NMDA receptor subunits (GluN1, GluN2A-2D) in the MPOA of primiparous female rats that were kept with pups until brain sampling. As a result, we found that the mRNA levels of GluA3 and GluN2B were significantly higher in primiparous females on the day of weaning compared with those in primiparous females on the day of parturition. Additionally, we examined the effects of CNQX, an AMPA receptor antagonist, and MK-801, an NMDA receptor antagonist, injected into the MPOA on maternal behavior in maternally experienced primiparous female rats. Maternal behavioral activity was significantly reduced when CNQX or MK-801 was injected into the MPOA. These findings indicate that long-term maternal experience in the postpartum period up-regulates glutamatergic neurotransmission by increasing the number of mushroom spines and glutamate receptor expression, which may be involved in the retention of maternal behavior in maternally experienced female rats.
Collapse
Affiliation(s)
- Eri Okino
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, 338-8570, Japan
| | - Sayaka Morita
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, 338-8570, Japan
| | - Yumi Hoshikawa
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, 338-8570, Japan
| | - Shinji Tsukahara
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama, 338-8570, Japan.
| |
Collapse
|
48
|
Zhang Y, Song L, Dong H, Kim DS, Sun Z, Boger H, Wang Q, Wang H. Spinophilin-deficient mice are protected from diet-induced obesity and insulin resistance. Am J Physiol Endocrinol Metab 2020; 319:E354-E362. [PMID: 32603260 PMCID: PMC7473908 DOI: 10.1152/ajpendo.00114.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/27/2020] [Accepted: 06/16/2020] [Indexed: 02/06/2023]
Abstract
Browning of white adipose tissue (WAT) has been shown to reduce obesity and obesity-related complications, suggesting that factors that promote WAT browning may have applications in the development of therapeutic strategies for treating obesity. Here, we show that ablation of spinophilin (SPL), a ubiquitously expressed, multidomain scaffolding protein, increases metabolism and improves energy balance. Male and female SPL knockout (KO) and wild-type (WT) littermate controls were fed a chow diet or a high-fat diet (HFD). Body weight, hepatic steatosis, glucose and insulin tolerance, physical activity, and expression of browning genes in adipose tissues were measured and compared. Male SPL knockout (KO) mice fed a chow diet were significantly leaner, had lower body weights, and exhibited better glucose tolerance and insulin sensitivity than wild-type (WT) littermate controls. When fed an HFD, SPL KO mice were protected from increased body fat, weight gain, hepatic steatosis, hyperinsulinemia, and insulin resistance. Physical activity of SPL KO mice was markedly increased compared with WT controls. Furthermore, expression of the brown adipocyte marker, uncoupling protein-1 (UCP-1), and the mitochondrial activity markers, cd137 and c-idea, were significantly increased in visceral WAT (vWAT) of SPL KO mice, suggesting that SPL knockout protected the mice from HFD-induced obesity and its metabolic complications, at least in part, by promoting the browning of white adipocytes in vWAT. Our data identify a critical role of SPL in regulating glucose homeostasis, obesity, and adipocyte browning. These results suggest SPL may serve as a drug target for obesity and diabetes.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
- College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Lili Song
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Huansheng Dong
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
- College of Life Sciences, Qingdao Agricultural University, Qingdao, People's Republic of China
| | - Do-Sung Kim
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Zhen Sun
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
| | - Heather Boger
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Qin Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina
| |
Collapse
|
49
|
Altermann Torre V, Machado AG, de Sá Couto-Pereira N, Mar Arcego D, Dos Santos Vieira A, Salerno PSV, Dos Santos Garcia E, Lazzaretti C, Toniazzo AP, Nedel F, Noschang C, Schmitz F, Wyse ATS, Dalmaz C, Krolow R. Consumption of a palatable diet rich in simple sugars during development impairs memory of different degrees of emotionality and changes hippocampal plasticity according to the age of the rats. Int J Dev Neurosci 2020; 80:354-368. [PMID: 32299124 DOI: 10.1002/jdn.10032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/18/2020] [Accepted: 03/23/2020] [Indexed: 01/01/2023] Open
Abstract
We investigated the effect of a chronic palatable diet rich in simple sugars on memory of different degrees of emotionality in male adult rats, and on hippocampal plasticity markers in different stages of development. On postnatal day (PND) 21, 45 male Wistar rats were divided in two groups, according to their diet: (1-Control) receiving standard lab chow or (2-Palatable Diet) receiving both standard chow plus palatable diet ad libitum. At PND 60, behavioral tests were performed to investigate memory in distinct tasks. Hippocampal plasticity markers were investigated at PND 28 in half of the animals, and after the behavioral tests. Palatable diet consumption induced an impairment in memory, aversive or not, and increased Na+ , K+ -ATPase activity, both at PND 28, and in the adulthood. Synaptophysin, brain-derived neurotrophic factor (BDNF), and protein kinase B (AKT), and phosphorylated AKT were reduced in the hippocampus at PND 28. However, at PND 75, this diet consumption led to increased hippocampal levels of synaptophysin, spinophilin/neurabin-II, and decreased BDNF and neuronal nitric oxide synthase. These results showed a strongly association of simple sugars-rich diet consumption during the development with memory impairments. Plasticity markers are changed, with results that depend on the stage of development evaluated.
Collapse
Affiliation(s)
- Viviane Altermann Torre
- Pós-Graduação em Saúde e Comportamento, Universidade Católica de Pelotas (UCPel), Pelotas, Brazil
| | - Alessandra Gonçalves Machado
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Natividade de Sá Couto-Pereira
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Danusa Mar Arcego
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Aline Dos Santos Vieira
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | | | - Emily Dos Santos Garcia
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Camilla Lazzaretti
- Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Ana Paula Toniazzo
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Fernanda Nedel
- Pós-Graduação em Saúde e Comportamento, Universidade Católica de Pelotas (UCPel), Pelotas, Brazil
| | - Cristie Noschang
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Felipe Schmitz
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Angela T S Wyse
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Carla Dalmaz
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
- Pós-Graduação em Neurociências, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| | - Rachel Krolow
- Pós-Graduação em Saúde e Comportamento, Universidade Católica de Pelotas (UCPel), Pelotas, Brazil
- Pós-Graduação em Ciências Biológicas: Bioquímica/Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, Brazil
| |
Collapse
|
50
|
Camacho-Arroyo I, Piña-Medina AG, Bello-Alvarez C, Zamora-Sánchez CJ. Sex hormones and proteins involved in brain plasticity. VITAMINS AND HORMONES 2020; 114:145-165. [PMID: 32723542 DOI: 10.1016/bs.vh.2020.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
It is well known that peripheral sex steroid hormones cross the blood-brain barrier and control a broad spectrum of reproductive behaviors. However, their role in other essential brain functions was investigated since the 1980s, when the accumulation of pregnenolone and dehydroepiandrosterone in the brain of mammalian species was determined. Since then, numerous studies have demonstrated the participation of sex hormones in brain plasticity processes. Sex hormones through both genomic and non-genomic mechanisms of action are capable of inducing gene transcription or activating signaling cascades that result in the promotion of different physiological and pathological events of brain plasticity, such as remodeling or formation of dendritic spines, neurogenesis, synaptogenesis or myelination. In this chapter, we will present the effects of sex hormones and proteins involved in brain plasticity.
Collapse
Affiliation(s)
- Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico.
| | - Ana Gabriela Piña-Medina
- Facultad de Química, Departamento de Biología, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| | - Claudia Bello-Alvarez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Carmen J Zamora-Sánchez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|