1
|
Angel D, Tsutiya A, Hayani H, Madencioglu D, Kul E, Caliskan G, Demiray YE, Dityatev A, Stork O. The Serine/Threonine Kinase NDR2 Regulates Integrin Signaling, Synapse Formation, and Synaptic Plasticity in the Hippocampus. J Neurochem 2025; 169:e70094. [PMID: 40439020 PMCID: PMC12120816 DOI: 10.1111/jnc.70094] [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: 09/02/2024] [Revised: 04/03/2025] [Accepted: 05/11/2025] [Indexed: 06/02/2025]
Abstract
Nuclear Dbf2-related (NDR) kinases are core components of the Hippo pathway, which controls neuronal polarity and neurite growth in the central nervous system (CNS). NDR2 is the principal NDR kinase in the mouse CNS, where it has been shown to regulate integrin-dependent dendritic branching as well as growth and plasticity in hippocampal mossy fibers. Given the well-established involvement of integrins in plasticity, we hypothesized that NDR2 might regulate synapse formation and plasticity through integrin-mediated mechanisms. In this study, using constitutive NDR2 null mutant mice, we demonstrate that Ndr2 deficiency leads to a reduction of T788/789 phosphorylated β1 integrin expression at synaptic sites both in the hippocampal area CA1 and in primary hippocampal neurons in vitro. This reduction is associated with decreased synaptic density in both conditions and accompanied by reduced long-term potentiation in the synapses between Schaffer collaterals/commissural fibers and CA1 pyramidal cells, which could be restored by activation of integrins with an arginine-glycine-aspartate-containing peptide, as well as with mild spatial memory deficits. Together, our results suggest that NDR2 is involved in integrin-dependent synapse formation and plasticity in the mouse hippocampus.
Collapse
Affiliation(s)
- Del Angel
- Department of Genetics & Molecular NeurobiologyInstitute of Biology, Otto‐Von‐Guericke UniversityMagdeburgGermany
| | - Atsuhiro Tsutiya
- Department of Genetics & Molecular NeurobiologyInstitute of Biology, Otto‐Von‐Guericke UniversityMagdeburgGermany
| | - Hussam Hayani
- Molecular Neuroplasticity GroupGerman Center for Neurodegenerative DiseasesMagdeburgGermany
| | - Deniz Madencioglu
- Department of Genetics & Molecular NeurobiologyInstitute of Biology, Otto‐Von‐Guericke UniversityMagdeburgGermany
| | - Emre Kul
- Department of Genetics & Molecular NeurobiologyInstitute of Biology, Otto‐Von‐Guericke UniversityMagdeburgGermany
| | - Gürsel Caliskan
- Department of Genetics & Molecular NeurobiologyInstitute of Biology, Otto‐Von‐Guericke UniversityMagdeburgGermany
- Center for Behavioural Brain SciencesMagdeburgGermany
| | - Yunus Emre Demiray
- Department of Genetics & Molecular NeurobiologyInstitute of Biology, Otto‐Von‐Guericke UniversityMagdeburgGermany
| | - Alexander Dityatev
- Molecular Neuroplasticity GroupGerman Center for Neurodegenerative DiseasesMagdeburgGermany
- Center for Behavioural Brain SciencesMagdeburgGermany
- Medical FacultyOtto‐Von‐Guericke UniversityMagdeburgGermany
| | - Oliver Stork
- Department of Genetics & Molecular NeurobiologyInstitute of Biology, Otto‐Von‐Guericke UniversityMagdeburgGermany
- Center for Behavioural Brain SciencesMagdeburgGermany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C‐I‐R‐C)Jena‐Magdeburg‐HalleGermany
- German Center for Mental Health (DZPG)Site Jena‐Magdeburg‐HalleGermany
| |
Collapse
|
2
|
Su W, Liu Y, Lam A, Hao X, Baudry M, Bi X. Contextual fear memory impairment in Angelman syndrome model mice is associated with altered transcriptional responses. Sci Rep 2023; 13:18647. [PMID: 37903805 PMCID: PMC10616231 DOI: 10.1038/s41598-023-45769-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 10/24/2023] [Indexed: 11/01/2023] Open
Abstract
Angelman syndrome (AS) is a rare neurogenetic disorder caused by UBE3A deficiency and characterized by severe developmental delay, cognitive impairment, and motor dysfunction. In the present study, we performed RNA-seq on hippocampal samples from both wildtype (WT) and AS male mice, with or without contextual fear memory recall. There were 281 recall-associated differentially expressed genes (DEGs) in WT mice and 268 DEGs in AS mice, with 129 shared by the two genotypes. Gene ontology analysis showed that extracellular matrix and stimulation-induced response genes were prominently enriched in recall-associated DEGs in WT mice, while nuclear acid metabolism and tissue development genes were highly enriched in those from AS mice. Further analyses showed that the 129 shared DEGs belonged to nuclear acid metabolism and tissue development genes. Unique recall DEGs in WT mice were enriched in biological processes critical for synaptic plasticity and learning and memory, including the extracellular matrix network clustered around fibronectin 1 and collagens. In contrast, AS-specific DEGs were not enriched in any known pathways. These results suggest that memory recall in AS mice, while altering the transcriptome, fails to recruit memory-associated transcriptional programs, which could be responsible for the memory impairment in AS mice.
Collapse
Affiliation(s)
- Wenyue Su
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Yan Liu
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Aileen Lam
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA
| | - Xiaoning Hao
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA
| | - Michel Baudry
- College of Dental Medicine, Western University of Health Sciences, Pomona, CA, 91766, USA
| | - Xiaoning Bi
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 701 E. 2nd St., Pomona, CA, 91766-1854, USA.
| |
Collapse
|
3
|
Biose IJ, Ismael S, Ouvrier B, White AL, Bix GJ. The Potential Role of Integrin Signaling in Memory and Cognitive Impairment. Biomolecules 2023; 13:biom13010108. [PMID: 36671492 PMCID: PMC9855855 DOI: 10.3390/biom13010108] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/29/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
Dementia currently has no cure and, due to the increased prevalence and associated economic and personal burden of this condition, current research efforts for the development of potential therapies have intensified. Recently, targeting integrins as a strategy to ameliorate dementia and other forms of cognitive impairment has begun to gain traction. Integrins are major bidirectional signaling receptors in mammalian cells, mediating various physiological processes such as cell-cell interaction and cell adhesion, and are also known to bind to the extracellular matrix. In particular, integrins play a critical role in the synaptic transmission of signals, hence their potential contribution to memory formation and significance in cognitive impairment. In this review, we describe the physiological roles that integrins play in the blood-brain barrier (BBB) and in the formation of memories. We also provide a clear overview of how integrins are implicated in BBB disruption following cerebral pathology. Given that vascular contributions to cognitive impairment and dementia and Alzheimer's' disease are prominent forms of dementia that involve BBB disruption, as well as chronic inflammation, we present current approaches shown to improve dementia-like conditions with integrins as a central focus. We conclude that integrins are vital in memory formation and that their disruption could lead to various forms of cognitive impairment. While further research to understand the relationships between integrins and memory is needed, we propose that the translational relevance of research efforts in this area could be improved through the use of appropriately aged, comorbid, male and female animals.
Collapse
Affiliation(s)
- Ifechukwude Joachim Biose
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Saifudeen Ismael
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Blake Ouvrier
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Amanda Louise White
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Brain Institute, Tulane University, New Orleans, LA 70112, USA
| | - Gregory Jaye Bix
- Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- School of Medicine, Tulane University, New Orleans, LA 70112, USA
- Department of Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70122, USA
- Correspondence: ; Tel.: +1-504-988-3564
| |
Collapse
|
4
|
Shiosaka S. Kallikrein 8: A key sheddase to strengthen and stabilize neural plasticity. Neurosci Biobehav Rev 2022; 140:104774. [PMID: 35820483 DOI: 10.1016/j.neubiorev.2022.104774] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/01/2022] [Accepted: 07/06/2022] [Indexed: 11/19/2022]
Abstract
Neural networks are modified and reorganized throughout life, even in the matured brain. Synapses in the networks form, change, or disappear dynamically in the plasticity state. The pre- and postsynaptic signaling, transmission, and structural dynamics have been studied considerably well. However, not many studies have shed light on the events in the synaptic cleft and intercellular space. Neural activity-dependent protein shedding is a phenomenon in which (1) presynaptic excitation evokes secretion or activation of sheddases, (2) sheddases are involved not only in cleavage of membrane- or matrix-bound proteins but also in mechanical modulation of cell-to-cell connectivity, and (3) freed activity domains of protein factors play a role in receptor-mediated or non-mediated biological actions. Kallikrein 8/neuropsin (KLK8) is a kallikrein family serine protease rich in the mammalian limbic brain. Accumulated evidence has suggested that KLK8 is an important modulator of neural plasticity and consequently, cognition. Insufficiency, as well as excess of KLK8 may have detrimental effects on limbic functions.
Collapse
Affiliation(s)
- Sadao Shiosaka
- Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka Prefectural Hospital Organization, Miyanosaka 3-16-21, Hirakata-shi, Osaka 573-0022, Japan.
| |
Collapse
|
5
|
Kietzman HW, Shapiro LP, Trinoskey-Rice G, Gourley SL. Cell adhesion presence during adolescence controls the architecture of projection-defined prefrontal cortical neurons and reward-related action strategies later in life. Dev Cogn Neurosci 2022; 54:101097. [PMID: 35325840 PMCID: PMC8938620 DOI: 10.1016/j.dcn.2022.101097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/13/2022] [Accepted: 03/11/2022] [Indexed: 01/17/2023] Open
Abstract
Adolescent brain development is characterized by neuronal remodeling in the prefrontal cortex; relationships with behavior are largely undefined. Integrins are cell adhesion factors that link the extracellular matrix with intracellular actin cytoskeleton. We find that β1-integrin presence in the prelimbic prefrontal cortex (PL) during adolescence, but not adulthood, is necessary for mice to select actions based on reward likelihood and value. As such, adult mice that lacked β1-integrin during adolescence failed to modify response strategies when rewards lost value or failed to be delivered. This pattern suggests that β1-integrin-mediated neuronal development is necessary for PL function in adulthood. We next visualized adolescent PL neurons, including those receiving input from the basolateral amygdala (BLA) - thought to signal salience - and projecting to the dorsomedial striatum (DMS) - the striatal output by which the PL controls goal-seeking behavior. Firstly, we found that these projection-defined neurons had a distinct morphology relative to general layer V PL neurons. Secondly, β1-integrin loss triggered the overexpression of stubby-type dendritic spines at the expense of mature spines, including on projection-defined neurons. This phenotype was not observed when β1-integrins were silenced before or after adolescence. Altogether, our experiments localize β1-integrin-mediated cell adhesion within a developing di-synaptic circuit coordinating adaptive action.
Collapse
Affiliation(s)
- Henry W Kietzman
- Medical Scientist Training Program, Emory University School of Medicine, United States; Departments of Pediatrics and Psychiatry, Emory University School of Medicine, United States; Graduate Program in Neuroscience, Emory University, United States; Yerkes National Primate Research Center, Emory University, United States
| | - Lauren P Shapiro
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, United States; Yerkes National Primate Research Center, Emory University, United States; Graduate Program in Molecular and Systems Pharmacology, Emory University, United States
| | - Gracy Trinoskey-Rice
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, United States; Yerkes National Primate Research Center, Emory University, United States
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, United States; Graduate Program in Neuroscience, Emory University, United States; Yerkes National Primate Research Center, Emory University, United States; Graduate Program in Molecular and Systems Pharmacology, Emory University, United States; Children's Healthcare of Atlanta, United States.
| |
Collapse
|
6
|
Harris RM, Kao HY, Alarcon JM, Hofmann HA, Fenton AA. Hippocampal transcriptomic responses to enzyme-mediated cellular dissociation. Hippocampus 2019; 29:876-882. [PMID: 31087609 DOI: 10.1002/hipo.23095] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 03/08/2019] [Accepted: 03/15/2019] [Indexed: 11/08/2022]
Abstract
Single-neuron gene expression studies may be especially important for understanding nervous system structure and function because of the neuron-specific functionality and plasticity that defines functional neural circuits. Cellular dissociation is a prerequisite technical manipulation for single-cell and single cell-population studies, but the extent to which the cellular dissociation process affects neural gene expression has not been determined. This information is necessary for interpreting the results of experimental manipulations that affect neural function such as learning and memory. The goal of this research was to determine the impact of cellular dissociation on brain transcriptomes. We compared gene expression of microdissected samples from the dentate gyrus (DG), CA3, and CA1 subfields of the mouse hippocampus either prepared by a standard tissue homogenization protocol or subjected to enzymatic digestion used to dissociate cells within tissues. We report that compared to homogenization, enzymatic dissociation alters about 350 genes or 2% of the hippocampal transcriptome. While only a few genes canonically implicated in long-term potentiation and fear memory change expression levels in response to the dissociation procedure, these data indicate that sample preparation can affect gene expression profiles, which might confound interpretation of results depending on the research question. This study is important for the investigation of any complex tissues as research effort moves from subfield level analysis to single cell analysis of gene expression.
Collapse
Affiliation(s)
- Rayna M Harris
- Department of Integrative Biology, Center for Computational Biology and Bioinformatics, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas.,Neural Systems and Behavior Course, Marine Biological Laboratory, Woods Hole, Massachusetts
| | - Hsin-Yi Kao
- Neural Systems and Behavior Course, Marine Biological Laboratory, Woods Hole, Massachusetts.,Center for Neural Science, New York University, New York, New York
| | - Juan Marcos Alarcon
- Neural Systems and Behavior Course, Marine Biological Laboratory, Woods Hole, Massachusetts.,Department of Pathology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA.,The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Medical Center, Brooklyn, New York
| | - Hans A Hofmann
- Department of Integrative Biology, Center for Computational Biology and Bioinformatics, Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, Texas.,Neural Systems and Behavior Course, Marine Biological Laboratory, Woods Hole, Massachusetts
| | - André A Fenton
- Neural Systems and Behavior Course, Marine Biological Laboratory, Woods Hole, Massachusetts.,Center for Neural Science, New York University, New York, New York.,The Robert F. Furchgott Center for Neural and Behavioral Science, State University of New York, Downstate Medical Center, Brooklyn, New York.,Department of Physiology and Pharmacology, State University of New York, Downstate Medical Center, Brooklyn, New York, USA.,Neuroscience Institute at the New York University Langone Medical Center, New York University, New York, New York
| |
Collapse
|
7
|
Farizatto KLG, Almeida MF, Long RT, Bahr BA. Early Synaptic Alterations and Selective Adhesion Signaling in Hippocampal Dendritic Zones Following Organophosphate Exposure. Sci Rep 2019; 9:6532. [PMID: 31024077 PMCID: PMC6484076 DOI: 10.1038/s41598-019-42934-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/11/2019] [Indexed: 11/09/2022] Open
Abstract
Organophosphates account for many of the world's deadliest poisons. They inhibit acetylcholinesterase causing cholinergic crises that lead to seizures and death, while survivors commonly experience long-term neurological problems. Here, we treated brain explants with the organophosphate compound paraoxon and uncovered a unique mechanism of neurotoxicity. Paraoxon-exposed hippocampal slice cultures exhibited progressive declines in synaptophysin, synapsin II, and PSD-95, whereas reduction in GluR1 was slower and NeuN and Nissl staining showed no indications of neuronal damage. The distinctive synaptotoxicity was observed in dendritic zones of CA1 and dentate gyrus. Interestingly, declines in synapsin II dendritic labeling correlated with increased staining for β1 integrin, a component of adhesion receptors that regulate synapse maintenance and plasticity. The paraoxon-induced β1 integrin response was targeted to synapses, and the two-fold increase in β1 integrin was selective as other synaptic adhesion molecules were unchanged. Additionally, β1 integrin-cofilin signaling was triggered by the exposure and correlations were found between the extent of synaptic decline and the level of β1 integrin responses. These findings identified organophosphate-mediated early and lasting synaptotoxicity which can explain delayed neurological dysfunction later in life. They also suggest that the interplay between synaptotoxic events and compensatory adhesion responses influences neuronal fate in exposed individuals.
Collapse
Affiliation(s)
- Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Michael F Almeida
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Ronald T Long
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, North Carolina, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, North Carolina, USA. .,Department of Biology, University of North Carolina-Pembroke, Pembroke, North Carolina, USA. .,Department of Chemistry and Physics, University of North Carolina-Pembroke, Pembroke, North Carolina, USA.
| |
Collapse
|
8
|
Hunter DD, Manglapus MK, Bachay G, Claudepierre T, Dolan MW, Gesuelli KA, Brunken WJ. CNS synapses are stabilized trans-synaptically by laminins and laminin-interacting proteins. J Comp Neurol 2017; 527:67-86. [PMID: 29023785 DOI: 10.1002/cne.24338] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/21/2017] [Accepted: 09/29/2017] [Indexed: 01/05/2023]
Abstract
The retina expresses several laminins in the outer plexiform layer (OPL), where they may provide an extracellular scaffold for synapse stabilization. Mice with a targeted deletion of the laminin β2 gene (Lamb2) exhibit retinal disruptions: photoreceptor synapses in the OPL are disorganized and the retinal physiological response is attenuated. We hypothesize that laminins are required for proper trans-synaptic alignment. To test this, we compared the distribution, expression, association and modification of several pre- and post-synaptic elements in wild-type and Lamb2-null retinae. A potential laminin receptor, integrin α3, is at the presynaptic side of the wild-type OPL. Another potential laminin receptor, dystroglycan, is at the post-synaptic side of the wild-type OPL. Integrin α3 and dystroglycan can be co-immunoprecipitated with the laminin β2 chain, demonstrating that they may bind laminins. In the absence of the laminin β2 chain, the expression of many pre-synaptic components (bassoon, kinesin, among others) is relatively undisturbed although their spatial organization and anchoring to the membrane is disrupted. In contrast, in the Lamb2-null, β-dystroglycan (β-DG) expression is altered, co-localization of β-DG with dystrophin and the glutamate receptor mGluR6 is disrupted, and the post-synaptic bipolar cell components mGluR6 and GPR179 become dissociated, suggesting that laminins mediate scaffolding of post-synaptic components. In addition, although pikachurin remains associated with β-DG, pikachurin is no longer closely associated with mGluR6 or α-DG in the Lamb2-null. These data suggest that laminins act as links among pre- and post-synaptic laminin receptors and α-DG and pikachurin in the synaptic space to maintain proper trans-synaptic alignment.
Collapse
Affiliation(s)
- Dale D Hunter
- Department of Anatomy and Cellular Biology, Tufts University and Tufts Center for Vision Research, Boston, Massachusetts.,Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| | - Mary K Manglapus
- Department of Anatomy and Cellular Biology, Tufts University and Tufts Center for Vision Research, Boston, Massachusetts
| | - Galina Bachay
- Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| | - Thomas Claudepierre
- Department of Anatomy and Cellular Biology, Tufts University and Tufts Center for Vision Research, Boston, Massachusetts
| | - Michael W Dolan
- Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| | - Kelly-Ann Gesuelli
- Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| | - William J Brunken
- Department of Anatomy and Cellular Biology, Tufts University and Tufts Center for Vision Research, Boston, Massachusetts.,Department of Ophthalmology and the SUNY Eye Institute, Upstate Medical University, Syracuse, New York
| |
Collapse
|
9
|
Abstract
Integrins are a large family of extracellular matrix (ECM) receptors. In the developing and adult brain, many integrins are present at high levels at synapses. The tetrapartite structure of synapses - which comprises presynaptic and postsynaptic neurons, the ECM and glial processes - places synaptic integrins in an excellent position to sense dynamic changes in the synaptic environment and use this information to coordinate further changes in synapse structure and function that will shape neural circuit properties. Recent developments in our understanding of the cellular and physiological roles of integrins, which range from control of neural process outgrowth and synapse formation to regulation of synaptic plasticity and memory, enable us to attempt a synthesis of synaptic integrin function.
Collapse
|
10
|
Affiliation(s)
- R Douglas Fields
- Laboratory of Developmental Neurobiology, National Institutes of Health, NICHD, Bethesda, Maryland
| |
Collapse
|
11
|
Varney S, Polston KF, Jessen T, Carneiro AMD. Mice lacking integrin β3 expression exhibit altered response to chronic stress. Neurobiol Stress 2015; 2:51-58. [PMID: 26634222 PMCID: PMC4664197 DOI: 10.1016/j.ynstr.2015.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Recent studies indicate multiple roles for integrin αvβ3 in adult neurons, including response to pharmacological agents such as cocaine and selective serotonin reuptake inhibitors. In this study, we examined the role of the integrin β3 gene (Itgb3) in the response to environmental stimuli by subjecting Itgb3+/+ and Itgb3-/- mice to unpredictable chronic mild stressors. We found that genetic abrogation of integrin β3 expression elicits an exaggerated vulnerability to chronic unpredictable stress in the open field test. In this test, chronic stress elicited significant decreases in stereotypic behavior and horizontal locomotor activity, including increases in anxiety behaviors. Mild chronic stress led to reductions in dopamine turnover in midbrains of Itgb3+/+, but not Itgb3-/- mice, suggesting a disruption of stress-dependent regulation of DA homeostasis. Chronic stress elicited altered synaptic expression of syntaxin and synaptophysin in midbrains of Itgb3-/- mice, when compared to Itgb3+/+. Semi-quantitative Western blot studies revealed that the synaptic expression, but not total tissue expression, of multiple signaling proteins is correlated with integrin αv levels in the midbrain. Moreover, loss of integrin β3 expression modifies this correlation network. Together, these findings demonstrate that Itgb3-/- mice display a pattern of changes indicating disrupted regulation of midbrain synaptic systems involved in conferring resilience to mild stressors.
Collapse
Affiliation(s)
- Seth Varney
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| | - Keith F Polston
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| | - Tammy Jessen
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| | - Ana M D Carneiro
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232-6600, USA
| |
Collapse
|
12
|
Levy AD, Omar MH, Koleske AJ. Extracellular matrix control of dendritic spine and synapse structure and plasticity in adulthood. Front Neuroanat 2014; 8:116. [PMID: 25368556 PMCID: PMC4202714 DOI: 10.3389/fnana.2014.00116] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/29/2014] [Indexed: 12/20/2022] Open
Abstract
Dendritic spines are the receptive contacts at most excitatory synapses in the central nervous system. Spines are dynamic in the developing brain, changing shape as they mature as well as appearing and disappearing as they make and break connections. Spines become much more stable in adulthood, and spine structure must be actively maintained to support established circuit function. At the same time, adult spines must retain some plasticity so their structure can be modified by activity and experience. As such, the regulation of spine stability and remodeling in the adult animal is critical for normal function, and disruption of these processes is associated with a variety of late onset diseases including schizophrenia and Alzheimer's disease. The extracellular matrix (ECM), composed of a meshwork of proteins and proteoglycans, is a critical regulator of spine and synapse stability and plasticity. While the role of ECM receptors in spine regulation has been extensively studied, considerably less research has focused directly on the role of specific ECM ligands. Here, we review the evidence for a role of several brain ECM ligands and remodeling proteases in the regulation of dendritic spine and synapse formation, plasticity, and stability in adults.
Collapse
Affiliation(s)
- Aaron D Levy
- Interdepartmental Neuroscience Program, Yale University New Haven, CT, USA ; Department of Molecular Biophysics and Biochemistry, Yale University New Haven, CT, USA
| | - Mitchell H Omar
- Interdepartmental Neuroscience Program, Yale University New Haven, CT, USA ; Department of Molecular Biophysics and Biochemistry, Yale University New Haven, CT, USA
| | - Anthony J Koleske
- Interdepartmental Neuroscience Program, Yale University New Haven, CT, USA ; Department of Molecular Biophysics and Biochemistry, Yale University New Haven, CT, USA ; Department of Neurobiology, Yale University New Haven, CT, USA
| |
Collapse
|
13
|
Venkatasubramaniam A, Drude A, Good T. Role of N-terminal residues in Aβ interactions with integrin receptor and cell surface. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:2568-77. [DOI: 10.1016/j.bbamem.2014.06.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 05/26/2014] [Accepted: 06/13/2014] [Indexed: 01/12/2023]
|
14
|
Ojelade SA, Acevedo SF, Rothenfluh A. The role of the actin cytoskeleton in regulating Drosophila behavior. Rev Neurosci 2014; 24:471-84. [PMID: 24077615 DOI: 10.1515/revneuro-2013-0017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 08/21/2013] [Indexed: 10/26/2022]
Abstract
Over the past decade, the function of the cytoskeleton has been studied extensively in developing and mature neurons. Actin, a major cytoskeletal protein, is indispensable for the structural integrity and plasticity of neurons and their synapses. Disruption of actin dynamics has significant consequence for neurons, neuronal circuits, and the functions they govern. In particular, cell adhesion molecules, members of the Rho family of GTPases, and actin-binding proteins are important modulators of actin dynamics and neuronal as well as behavioral plasticity. In this review, we discuss recent advances in Drosophila that highlight the importance of actin regulatory proteins in mediating fly behaviors such as circadian rhythm, courtship behavior, learning and memory, and the development of drug addiction.
Collapse
|
15
|
Babayan AH, Kramár EA. Rapid effects of oestrogen on synaptic plasticity: interactions with actin and its signalling proteins. J Neuroendocrinol 2013; 25:1163-72. [PMID: 24112361 PMCID: PMC3989941 DOI: 10.1111/jne.12108] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 09/09/2013] [Accepted: 09/16/2013] [Indexed: 01/02/2023]
Abstract
Oestrogen rapidly enhances fast excitatory postsynaptic potentials, facilitates long-term potentiation (LTP) and increases spine numbers. Each effect likely contributes to the influence of the steroid on cognition and memory. In the present review, we first describe a model for the substrates of LTP that includes an outline of the synaptic events occurring during induction, expression and consolidation. Briefly, critical signalling pathways involving the small GTPases RhoA and Rac/Cdc42 are activated by theta burst-induced calcium influx and initiate actin filament assembly via phosphorylation (inactivation) of cofilin. Reorganisation of the actin cytoskeleton changes spine and synapse morphology, resulting in increased concentrations of AMPA receptors at stimulated contacts. We then use the synaptic model to develop a specific hypothesis about how oestrogen affects both baseline transmission and plasticity. Brief infusions of 17β-oestradiol (E2 ) reversibly stimulate the RhoA, cofilin phosphorylation and actin polymerisation cascade of the LTP machinery; blocking this eliminates the effects of the steroid on transmission. We accordingly propose that E2 induces a weak form of LTP and thereby increases synaptic responses, a hypothesis that also accounts for how it markedly enhances theta burst induced potentiation. Although the effects of E2 on the cytoskeleton could be a result of the direct activation of small GTPases by oestrogen receptors on the synaptic membrane, the hormone also activates tropomyosin-related kinase B receptors for brain-derived neurotrophic factor, a neurotrophin that engages the RhoA-cofilin sequence and promotes LTP. The latter observations raise the possibility that E2 produces its effects on synaptic physiology via transactivation of neighbouring receptors that have prominent roles in the management of spine actin, synaptic physiology and plasticity.
Collapse
Affiliation(s)
- A H Babayan
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | | |
Collapse
|
16
|
Tanigami H, Okamoto T, Yasue Y, Shimaoka M. Astroglial integrins in the development and regulation of neurovascular units. PAIN RESEARCH AND TREATMENT 2012; 2012:964652. [PMID: 23304493 PMCID: PMC3529429 DOI: 10.1155/2012/964652] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 11/13/2012] [Indexed: 11/18/2022]
Abstract
In the neurovascular units of the central nervous system, astrocytes form extensive networks that physically and functionally connect the neuronal synapses and the cerebral vascular vessels. This astrocytic network is thought to be critically important for coupling neuronal signaling activity and energy demand with cerebral vascular tone and blood flow. To establish and maintain this elaborate network, astrocytes must precisely calibrate their perisynaptic and perivascular processes in order to sense and regulate neuronal and vascular activities, respectively. Integrins, a prominent family of cell-adhesion molecules that support astrocytic migration in the brain during developmental and normal adult stages, have been implicated in regulating the integrity of the blood brain barrier and the tripartite synapse to facilitate the formation of a functionally integrated neurovascular unit. This paper describes the significant roles that integrins and connexins play not only in regulating astrocyte migration during the developmental and adult stages of the neurovascular unit, but also in general health and in such diseases as hepatic encephalopathy.
Collapse
Affiliation(s)
- Hironobu Tanigami
- Department of Anesthesiology, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka 537-8511, Japan
| | - Takayuki Okamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Mie, Tsu City, Japan
| | - Yuichi Yasue
- Department of Anesthesiology, Osaka Medical Center for Cancer and Cardiovascular Diseases, 1-3-3 Nakamichi, Higashinari-ku, Osaka 537-8511, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Mie, Tsu City, Japan
| |
Collapse
|
17
|
Almonte AG, Qadri LH, Sultan FA, Watson JA, Mount DJ, Rumbaugh G, Sweatt JD. Protease-activated receptor-1 modulates hippocampal memory formation and synaptic plasticity. J Neurochem 2012; 124:109-22. [PMID: 23113835 DOI: 10.1111/jnc.12075] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Revised: 09/21/2012] [Accepted: 10/22/2012] [Indexed: 11/28/2022]
Abstract
Protease-activated receptor-1 (PAR1) is an unusual G-protein coupled receptor (GPCR) that is activated through proteolytic cleavage by extracellular serine proteases. Although previous work has shown that inhibiting PAR1 activation is neuroprotective in models of ischemia, traumatic injury, and neurotoxicity, surprisingly little is known about PAR1's contribution to normal brain function. Here, we used PAR1-/- mice to investigate the contribution of PAR1 function to memory formation and synaptic function. We demonstrate that PAR1-/- mice have deficits in hippocampus-dependent memory. We also show that while PAR1-/- mice have normal baseline synaptic transmission at Schaffer collateral-CA1 synapses, they exhibit severe deficits in N-methyl-d-aspartate receptor (NMDAR)-dependent long-term potentiation (LTP). Mounting evidence indicates that activation of PAR1 leads to potentiation of NMDAR-mediated responses in CA1 pyramidal cells. Taken together, this evidence and our data suggest an important role for PAR1 function in NMDAR-dependent processes subserving memory formation and synaptic plasticity.
Collapse
Affiliation(s)
- Antoine G Almonte
- Department of Neurobiology and Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Ning L, Tian L, Smirnov S, Vihinen H, Llano O, Vick K, Davis RL, Rivera C, Gahmberg CG. Interactions between ICAM-5 and β1 integrins regulate neuronal synapse formation. J Cell Sci 2012; 126:77-89. [PMID: 23015592 DOI: 10.1242/jcs.106674] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Intercellular adhesion molecule-5 (ICAM-5) is a dendrite-specific adhesion molecule, which functions in both the immune and nervous systems. ICAM-5 is the only negative regulator that has been identified for maturation of dendritic spines so far. Shedding of the ICAM-5 ectodomain promotes spine maturation and enhances synaptic activity. However, the mechanism by which ICAM-5 regulates spine development remains poorly understood. In this study, we found that ablation of ICAM5 expression resulted in a significant increase in the formation of synaptic contacts and the frequency of miniature excitatory post-synaptic currents, an indicator of pre-synaptic release probability. Antibodies against ICAM-5 and β1 integrins altered spine maturation. Furthermore, we found that β1 integrins serve as binding partners for ICAM-5. β1 integrins were immunoprecipitated with ICAM-5 from mouse brain and the binding region in ICAM-5 was localized to the two first Ig domains. β1 integrins were juxtaposed to filopodia tips at the early stage of synaptic formation, but as synapses matured, β1 integrins covered the mushroom spines. Loss of β1 integrins from the pre-synaptic sites affected the morphology of the post-synaptic structures. ICAM-5 ectodomain cleavage decreased or increased when the interaction between ICAM-5 and β1 integrins was potentiated or weakened, respectively, using antibodies. These results suggest that the interaction between ICAM-5 and β1 integrins is important in formation of functional synapses.
Collapse
Affiliation(s)
- Lin Ning
- Division of Biochemistry and Biotechnology, Faculty of Biological and Environmental Sciences, University of Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wu X, Reddy DS. Integrins as receptor targets for neurological disorders. Pharmacol Ther 2011; 134:68-81. [PMID: 22233753 DOI: 10.1016/j.pharmthera.2011.12.008] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 12/15/2011] [Indexed: 12/18/2022]
Abstract
This review focuses on the neurobiology of integrins, pathophysiological roles of integrins in neuroplasticity and nervous system disorders, and therapeutic implications of integrins as potential drug targets and possible delivery pathways. Neuroplasticity is a central phenomenon in many neurological conditions such as seizures, trauma, and traumatic brain injury. During the course of many brain diseases, in addition to intracellular compartment changes, alterations in non-cell compartments such as extracellular matrix (ECM) are recognized as an essential process in forming and reorganizing neural connections. Integrins are heterodimeric transmembrane receptors that mediate cell-ECM and cell-cell adhesion events. Although the mechanisms of neuroplasticity remain unclear, it has been suggested that integrins undergo plasticity including clustering through interactions with ECM proteins, modulating ion channels, intracellular Ca(2+) and protein kinase signaling, and reorganization of cytoskeletal filaments. As cell surface receptors, integrins are central to the pathophysiology of many brain diseases, such as epilepsy, and are potential targets for the development of new drugs for neurological disorders.
Collapse
Affiliation(s)
- Xin Wu
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center College of Medicine, Bryan, TX 77807, USA
| | | |
Collapse
|
20
|
Monje FJ, Kim EJ, Pollak DD, Cabatic M, Li L, Baston A, Lubec G. Focal adhesion kinase regulates neuronal growth, synaptic plasticity and hippocampus-dependent spatial learning and memory. Neurosignals 2011; 20:1-14. [PMID: 21952616 DOI: 10.1159/000330193] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Accepted: 06/20/2011] [Indexed: 01/07/2023] Open
Abstract
The focal adhesion kinase (FAK) is a non-receptor tyrosine kinase abundantly expressed in the mammalian brain and highly enriched in neuronal growth cones. Inhibitory and facilitatory activities of FAK on neuronal growth have been reported and its role in neuritic outgrowth remains controversial. Unlike other tyrosine kinases, such as the neurotrophin receptors regulating neuronal growth and plasticity, the relevance of FAK for learning and memory in vivo has not been clearly defined yet. A comprehensive study aimed at determining the role of FAK in neuronal growth, neurotransmitter release and synaptic plasticity in hippocampal neurons and in hippocampus-dependent learning and memory was therefore undertaken using the mouse model. Gain- and loss-of-function experiments indicated that FAK is a critical regulator of hippocampal cell morphology. FAK mediated neurotrophin-induced neuritic outgrowth and FAK inhibition affected both miniature excitatory postsynaptic potentials and activity-dependent hippocampal long-term potentiation prompting us to explore the possible role of FAK in spatial learning and memory in vivo. Our data indicate that FAK has a growth-promoting effect, is importantly involved in the regulation of the synaptic function and mediates in vivo hippocampus-dependent spatial learning and memory.
Collapse
Affiliation(s)
- Francisco J Monje
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
21
|
McGeachie AB, Cingolani LA, Goda Y. Stabilising influence: integrins in regulation of synaptic plasticity. Neurosci Res 2011; 70:24-9. [PMID: 21352859 PMCID: PMC3242036 DOI: 10.1016/j.neures.2011.02.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2010] [Revised: 02/03/2011] [Accepted: 02/03/2011] [Indexed: 12/28/2022]
Abstract
Hebbian synaptic plasticity, such as hippocampal long-term potentiation (LTP), is thought to be important for particular types of learning and memory. It involves changes in the expression and activity of a large array of proteins, including cell adhesion molecules. The integrin class of cell adhesion molecules has been extensively studied in this respect, and appear to have a defined role in consolidating both structural and functional changes brought about by LTP. With the use of integrin inhibitors, it has been possible to identify a critical time window of several minutes after LTP induction for the participation of integrins in LTP. Altering the interactions of integrins with their ligands during this time compromises structural changes involving actin polymerisation and spine enlargement that could be required for accommodating new AMPA receptors (AMPARs). After this critical window of structural remodelling and plasticity, integrins "lock-in" and stabilise the morphological changes, conferring the requisite longevity for LTP. Genetic manipulations targeting integrin subtypes have helped identify the specific integrin subunits involved in LTP and correlate alterations in plasticity with behavioural deficits. Moreover, recent studies have implicated integrins in AMPAR trafficking and glycine receptor lateral diffusion, highlighting their multifaceted functions at the synapse.
Collapse
Affiliation(s)
- Andrew B. McGeachie
- Medical Research Council Laboratory for Molecular Cell Biology and Cell Biology Unit, University College London, Gower Street, London WC1E 6BT, UK
| | - Lorenzo A. Cingolani
- Medical Research Council Laboratory for Molecular Cell Biology and Cell Biology Unit, University College London, Gower Street, London WC1E 6BT, UK
| | - Yukiko Goda
- Medical Research Council Laboratory for Molecular Cell Biology and Cell Biology Unit, University College London, Gower Street, London WC1E 6BT, UK
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London WC1E 6BT, UK
| |
Collapse
|
22
|
Ziehn MO, Avedisian AA, Tiwari-Woodruff S, Voskuhl RR. Hippocampal CA1 atrophy and synaptic loss during experimental autoimmune encephalomyelitis, EAE. J Transl Med 2010; 90:774-86. [PMID: 20157291 PMCID: PMC3033772 DOI: 10.1038/labinvest.2010.6] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Over half of multiple sclerosis (MS) patients experience cognitive deficits, including learning and memory dysfunction, and the mechanisms underlying these deficits remain poorly understood. Neuronal injury and synaptic loss have been shown to occur within the hippocampus in other neurodegenerative disease models, and these pathologies have been correlated with cognitive impairment. Whether hippocampal abnormalities occur in MS models is unknown. Using experimental autoimmune encephalomyelitis (EAE), we evaluated hippocampal neurodegeneration and inflammation during disease. Hippocampal pathology began early in EAE disease course, and included decreases in CA1 pyramidal layer volume, loss of inhibitory interneurons and increased cell death of neurons and glia. It is interesting to note that these effects occurred in the presence of chronic microglial activation, with a relative paucity of infiltrating blood-borne immune cells. Widespread diffuse demyelination occurred in the hippocampus, but there was no significant decrease in axonal density. Furthermore, there was a significant reduction in pre-synaptic puncta and synaptic protein expression within the hippocampus, as well as impaired performance on a hippocampal-dependent spatial learning task. Our results demonstrate that neurodegenerative changes occur in the hippocampus during autoimmune-mediated demyelinating disease. This work establishes a preclinical model for assessing treatments targeted toward preventing hippocampal neuropathology and dysfunction in MS.
Collapse
MESH Headings
- Animals
- Apoptosis/immunology
- Astrocytes/metabolism
- Astrocytes/pathology
- Atrophy
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Female
- Glial Fibrillary Acidic Protein/metabolism
- Hippocampus/immunology
- Hippocampus/pathology
- Hippocampus/physiopathology
- In Situ Nick-End Labeling
- Learning Disabilities/immunology
- Learning Disabilities/physiopathology
- Male
- Memory Disorders/immunology
- Memory Disorders/physiopathology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Microglia/immunology
- Microglia/pathology
- Nerve Degeneration/immunology
- Nerve Degeneration/pathology
- Nerve Degeneration/physiopathology
- Neurons/metabolism
- Neurons/pathology
- Synapses/immunology
- Synapses/pathology
Collapse
Affiliation(s)
- Marina O Ziehn
- Interdepartmental Program of Neuroscience, University of California, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
23
|
Rushton E, Rohrbough J, Broadie K. Presynaptic secretion of mind-the-gap organizes the synaptic extracellular matrix-integrin interface and postsynaptic environments. Dev Dyn 2009; 238:554-71. [PMID: 19235718 PMCID: PMC2677818 DOI: 10.1002/dvdy.21864] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Mind-the-Gap (MTG) is required during synaptogenesis of the Drosophila glutamatergic neuromuscular junction (NMJ) to organize the postsynaptic domain. Here, we generate MTG::GFP transgenic animals to demonstrate MTG is synaptically targeted, secreted, and localized to punctate domains in the synaptic extracellular matrix (ECM). Drosophila NMJs form specialized ECM carbohydrate domains, with carbohydrate moieties and integrin ECM receptors occupying overlapping territories. Presynaptically secreted MTG recruits and reorganizes secreted carbohydrates, and acts to recruit synaptic integrins and ECM glycans. Transgenic MTG::GFP expression rescues hatching, movement, and synaptogenic defects in embryonic-lethal mtg null mutants. Targeted neuronal MTG expression rescues mutant synaptogenesis defects, and increases rescue of adult viability, supporting an essential neuronal function. These results indicate that presynaptically secreted MTG regulates the ECM-integrin interface, and drives an inductive mechanism for the functional differentiation of the postsynaptic domain of glutamatergic synapses. We suggest that MTG pioneers a novel protein family involved in ECM-dependent synaptic differentiation.
Collapse
Affiliation(s)
| | | | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt Brain Institute, Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
24
|
Juhász G, Vass G, Bozsó Z, Budai D, Penke B, Szegedi V. Integrin activation modulates NMDA and AMPA receptor function of CA1 cells in a dose-related fashion in vivo. Brain Res 2008; 1233:20-6. [DOI: 10.1016/j.brainres.2008.05.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2008] [Revised: 04/12/2008] [Accepted: 05/10/2008] [Indexed: 10/22/2022]
|
25
|
Surface trafficking of N-methyl-D-aspartate receptors: physiological and pathological perspectives. Neuroscience 2008; 158:4-18. [PMID: 18583064 DOI: 10.1016/j.neuroscience.2008.05.029] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 05/06/2008] [Accepted: 05/17/2008] [Indexed: 11/20/2022]
Abstract
The N-methyl-D-aspartate receptor (NMDAR) plays a crucial role in shaping the strength of synaptic connections. Over the last decades, extensive studies have defined the cellular and molecular mechanisms by which synaptic NMDARs control the maturation and plasticity of synaptic transmission, and how altered synaptic NMDAR signaling is implicated in neurodegenerative and psychiatric disorders. It is now clear that activation of synaptic or extrasynaptic NMDARs produces different signaling cascades and thus neuronal functions. Our current understanding of NMDAR surface distribution and trafficking is only emerging. Exchange of NMDARs between synaptic and extrasynaptic areas through surface diffusion is a highly dynamic and regulated process. The aim of this review is to describe the identified mechanisms that regulate surface NMDAR behaviors and discuss the impact of this new trafficking pathway on the well-established NMDAR-dependent physiological and pathophysiological processes.
Collapse
|
26
|
Tamura H, Ng DC, Tokuda T, Naoki H, Nakagawa T, Mizuno T, Hatanaka Y, Ishikawa Y, Ohta J, Shiosaka S. One-chip sensing device (biomedical photonic LSI) enabled to assess hippocampal steep and gradual up-regulated proteolytic activities. J Neurosci Methods 2008; 173:114-20. [PMID: 18601951 DOI: 10.1016/j.jneumeth.2008.06.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 05/29/2008] [Accepted: 06/04/2008] [Indexed: 11/17/2022]
Abstract
We developed an implantable one-chip biofluoroimaging device (termed biomedical photonic LSI; BpLSI) which enabled real-time molecular imaging with conventional electrophysiology in vivo in deep brain areas. The multimodal LSI enabled long-term sequential imaging of the fluorescence emitted by proteolysis-linked fluorogenic substrate. Using the BpLSI, we observed a process of stimulation-dependent modulation at synapse with multi-site (16 x 19 pixel) in widespread area and a high-speed video rate, and found that the gradual up-regulated proteolytic activity in a wide range of hippocampal CA1 area and the steep activity in local area, indicating that the proteolysis system is a basis for the fixation of long-term potentiation in post-excited synapses in the hippocampus. Mathematical data analysis confirmed the direct involvement of functional proteolysis for neural plasticity.
Collapse
Affiliation(s)
- Hideki Tamura
- Division of Structural Cell Biology, Graduate School of Biological Sciences, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Bevan S, Vakharia V, Parker D. Changes in gene expression and integrin-mediated structural changes are associated with long-term plasticity of a spinal cord locomotor network. Neuroscience 2008; 152:160-8. [DOI: 10.1016/j.neuroscience.2007.12.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2007] [Revised: 12/06/2007] [Accepted: 12/06/2007] [Indexed: 12/27/2022]
|
28
|
Influence of Integrin-blocking Peptide on Gadolinium- and Hypertonic Shrinking-induced Neurotransmitter Release in Rat Brain Synaptosomes. Neurochem Res 2008; 33:1316-24. [DOI: 10.1007/s11064-007-9585-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2007] [Accepted: 12/27/2007] [Indexed: 11/26/2022]
|
29
|
Ethell IM, Ethell DW. Matrix metalloproteinases in brain development and remodeling: synaptic functions and targets. J Neurosci Res 2008; 85:2813-23. [PMID: 17387691 DOI: 10.1002/jnr.21273] [Citation(s) in RCA: 307] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Matrix metalloproteinases (MMPs) play critical roles in egg fertilization, embryonic development, wound repair, cancer, and inflammatory and neurologic diseases. This subfamily of metzincin peptidases can cleave extracellular matrix (ECM) and pericellular proteins that have profound effects on cell behavior. Among known MMP substrates are several proteins that play important roles in synaptogenesis, synaptic plasticity, and long-term potentiation (LTP). In this Mini-Review we discuss how MMP-directed cleavage of these proteins can impact the formation and function of synapses within the brain. Pyramidal neurons in the hippocampus, and other large neurons, are surrounded by perineuronal nets that are composed of brevican, tenascin-R, and laminin, each of which is subject to proteolytic cleavage by MMPs. Tenascin-R knockout mice show deficits in learning and memory and LTP, as do at least two MMP knockouts. Impaired LTP is also seen in brain-derived neurotrophic factor (BDNF) knockout mice, which is interesting in that pro-BDNF can be processed into mature BDNF by several MMPs and thereby regulate activation of the high-affinity BDNF receptor TrkB. At the synaptic level, MMP substrates also include ephrins, Eph receptors, and cadherins, which are also involved in synapse development and plasticity. MMPs can also process membrane-bound tumor necrosis factor-alpha into a potent soluble cytokine that is increasingly implicated in neuron-glial signaling, particularly in neurologic disease. Finally, we discuss how the development of therapeutics to attenuate MMP activity in neurodegenerative disorders may become powerful tools for future studies of synaptic formation and function within the developing and mature brain.
Collapse
Affiliation(s)
- Iryna M Ethell
- Division of Biomedical Sciences, University of California Riverside, Riverside, California 92521-0121, USA.
| | | |
Collapse
|
30
|
Kanakry CG, Li Z, Nakai Y, Sei Y, Weinberger DR. Neuregulin-1 regulates cell adhesion via an ErbB2/phosphoinositide-3 kinase/Akt-dependent pathway: potential implications for schizophrenia and cancer. PLoS One 2007; 2:e1369. [PMID: 18159252 PMCID: PMC2147048 DOI: 10.1371/journal.pone.0001369] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2007] [Accepted: 12/05/2007] [Indexed: 12/19/2022] Open
Abstract
Background Neuregulin-1 (NRG1) is a putative schizophrenia susceptibility gene involved extensively in central nervous system development as well as cancer invasion and metastasis. Using a B lymphoblast cell model, we previously demonstrated impairment in NRG1α-mediated migration in cells derived from patients with schizophrenia as well as effects of risk alleles in NRG1 and catechol-O-methyltransferase (COMT), a second gene implicated both in schizophrenia susceptibility and in cancer. Methodology/Principal Findings Here, we examine cell adhesion, an essential component process of cell motility, using an integrin-mediated cell adhesion assay based on an interaction between ICAM-1 and the CD11a/CD18 integrin heterodimer expressed on lymphoblasts. In our assay, NRG1α induces lymphoblasts to assume varying levels of adhesion characterized by time-dependent fluctuations in the firmness of attachment. The maximum range of variation in adhesion over sixty minutes correlates strongly with NRG1α-induced migration (r2 = 0.61). NRG1α-induced adhesion variation is blocked by erbB2, PI3K, and Akt inhibitors, but not by PLC, ROCK, MLCK, or MEK inhibitors, implicating the erbB2/PI3K/Akt1 signaling pathway in NRG1-stimulated, integrin-mediated cell adhesion. In cell lines from 20 patients with schizophrenia and 20 normal controls, cells from patients show a significant deficiency in the range of NRG1α-induced adhesion (p = 0.0002). In contrast, the response of patient-derived cells to phorbol myristate acetate is unimpaired. The COMT Val108/158Met genotype demonstrates a strong trend towards predicting the range of the NRG1α-induced adhesion response with risk homozygotes having decreased variation in cell adhesion even in normal subjects (p = 0.063). Conclusion/Significance Our findings suggest that a mechanism of the NRG1 genetic association with schizophrenia may involve the molecular biology of cell adhesion.
Collapse
Affiliation(s)
- Christopher G. Kanakry
- Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
- Howard Hughes Medical Institute (HHMI)–National Institutes of Health (NIH) Research Scholars Program, Chevy Chase, Maryland, United States of America
| | - Zhen Li
- Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Yoko Nakai
- Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Yoshitatsu Sei
- Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
| | - Daniel R. Weinberger
- Genes, Cognition and Psychosis Program, Intramural Research Program, National Institute of Mental Health, National Institutes of Health (NIH), Bethesda, Maryland, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
31
|
Jones S, Pfister-Genskow M, Benca RM, Cirelli C. Molecular correlates of sleep and wakefulness in the brain of the white-crowned sparrow. J Neurochem 2007; 105:46-62. [PMID: 18028333 DOI: 10.1111/j.1471-4159.2007.05089.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
In the mammalian brain, sleep and wakefulness are associated with widespread changes in gene expression. The extent to which the molecular correlates of vigilance state are conserved across phylogeny, however, is only beginning to be explored. The goal of this study was to determine whether sleep and wakefulness affect gene expression in the avian brain. To achieve this end we performed an extensive microarray analysis of gene expression during sleep, wakefulness, and short-term sleep deprivation in the telencephalon of the white-crowned sparrow (Zonotrichia leucophrys gambelii). We found that, as in the rodent cerebral cortex, behavioral state, independent of time of day, has widespread effects on avian brain gene expression, affecting the transcript levels of 255 genes (1.4% of all tested transcripts). Wakefulness-related transcripts (n = 114) code for proteins involved in energy metabolism and oxidative phosphorylation, immediate early genes and transcription factors associated with activity-dependent neural plasticity, as well as heat-shock proteins and molecular chaperones associated with the unfolded protein response. Sleep-related transcripts (n = 141) code for proteins involved in membrane trafficking, lipid/cholesterol synthesis, translational regulation, cellular adhesion, and cytoskeletal organization. Remarkably, despite the considerable differences in morphology and cytology between the mammalian neocortex and the avian telencephalon, the functional categories of transcripts identified in this study exhibit a significant degree of overlap with those identified in the rodent cortex.
Collapse
Affiliation(s)
- Stephany Jones
- Neuroscience Training Program, and Department of Psychiatry, University of Wisconsin, Madison, Wisconsin 53719, USA
| | | | | | | |
Collapse
|
32
|
Rohrbough J, Rushton E, Woodruff E, Fergestad T, Vigneswaran K, Broadie K. Presynaptic establishment of the synaptic cleft extracellular matrix is required for post-synaptic differentiation. Genes Dev 2007; 21:2607-28. [PMID: 17901219 PMCID: PMC2000325 DOI: 10.1101/gad.1574107] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Formation and regulation of excitatory glutamatergic synapses is essential for shaping neural circuits throughout development. In a Drosophila genetic screen for synaptogenesis mutants, we identified mind the gap (mtg), which encodes a secreted, extracellular N-glycosaminoglycan-binding protein. MTG is expressed neuronally and detected in the synaptic cleft, and is required to form the specialized transsynaptic matrix that links the presynaptic active zone with the post-synaptic glutamate receptor (GluR) domain. Null mtg embryonic mutant synapses exhibit greatly reduced GluR function, and a corresponding loss of localized GluR domains. All known post-synaptic signaling/scaffold proteins functioning upstream of GluR localization are also grossly reduced or mislocalized in mtg mutants, including the dPix-dPak-Dock cascade and the Dlg/PSD-95 scaffold. Ubiquitous or neuronally targeted mtg RNA interference (RNAi) similarly reduce post-synaptic assembly, whereas post-synaptically targeted RNAi has no effect, indicating that presynaptic MTG induces and maintains the post-synaptic pathways driving GluR domain formation. These findings suggest that MTG is secreted from the presynaptic terminal to shape the extracellular synaptic cleft domain, and that the cleft domain functions to mediate transsynaptic signals required for post-synaptic development.
Collapse
Affiliation(s)
- Jeffrey Rohrbough
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Emma Rushton
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Elvin Woodruff
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Tim Fergestad
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Krishanthan Vigneswaran
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Kendal Broadie
- Department of Biological Sciences, Kennedy Center for Research on Human Development, Vanderbilt University, Nashville, Tennessee 37235, USA
- Corresponding author.E-MAIL ; FAX (615) 936-0129
| |
Collapse
|
33
|
Nagy V, Bozdagi O, Huntley GW. The extracellular protease matrix metalloproteinase-9 is activated by inhibitory avoidance learning and required for long-term memory. Learn Mem 2007; 14:655-64. [PMID: 17909100 PMCID: PMC2044557 DOI: 10.1101/lm.678307] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Matrix metalloproteinases (MMPs) are a family of extracellularly acting proteolytic enzymes with well-recognized roles in plasticity and remodeling of synaptic circuits during brain development and following brain injury. However, it is now becoming increasingly apparent that MMPs also function in normal, nonpathological synaptic plasticity of the kind that may underlie learning and memory. Here, we extend this idea by investigating the role and regulation of MMP-9 in an inhibitory avoidance (IA) learning and memory task. We demonstrate that following IA training, protein levels and proteolytic activity of MMP-9 become elevated in hippocampus by 6 h, peak at 12-24 h, then decline to baseline values by approximately 72 h. When MMP function is abrogated by intrahippocampal infusion of a potent gelatinase (MMP-2 and MMP-9) inhibitor 3.5 h following IA training, a time prior to the onset of training-induced elevation in levels, IA memory retention is significantly diminished when tested 1-3 d later. Animals impaired at 3 d exhibit robust IA memory when retrained, suggesting that such impairment is not likely attributed to toxic or other deleterious effects that permanently disrupt hippocampal function. In anesthetized adult rats, the effective distance over which synaptic plasticity is impaired by a single intrahippocampal infusion of the MMP inhibitor of the kind that blocks IA memory is approximately 1200 microm. Taken together, these data suggest that IA training induces a slowly emerging, but subsequently protracted period of MMP-mediated proteolysis critical for enabling long-lasting synaptic modification that underlies long-term memory consolidation.
Collapse
Affiliation(s)
- Vanja Nagy
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, New York 10029-6574, USA
| | - Ozlem Bozdagi
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, New York 10029-6574, USA
| | - George W. Huntley
- Fishberg Department of Neuroscience, The Mount Sinai School of Medicine, New York, New York 10029-6574, USA
- Corresponding author.E-mail ; fax (212) 659-5979
| |
Collapse
|
34
|
O'Connor DH, Wittenberg GM, Wang SSH. Timing and contributions of pre-synaptic and post-synaptic parameter changes during unitary plasticity events at CA3-CA1 synapses. Synapse 2007; 61:664-78. [PMID: 17503487 DOI: 10.1002/syn.20403] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
At individual synapses, post-synaptic responses include a mixture of "successes" and "failures" in which transmitter is released or not released, respectively. Previously we measured synaptic strength at CA3-CA1 synapses averaged over all trials, including both successes and failures, using an induction protocol that allowed us to observe potentiation and depression events as step-like changes. Here we report quantal properties of 15 of the earlier experiments, including 14 potentiation events and eight depression events. In five experiments both potentiation events and depression events were evoked at the same synapse. During potentiation, success rate increased from 0.56 +/- 0.14 (mean +/- SD) to 0.69 +/- 0.12, and during depression, success rate decreased from 0.70 +/- 0.09 to 0.51 +/- 0.10. During potentiation potency increased from 10 +/- 5 to 19 +/- 9 pA, and during depression, potency decreased from 18 +/- 12 to 12 +/- 7 pA. On average, changes in potency accounted for 76% of the change in response size in potentiation events and 60% of the change in depression events. A reduced-assumption spectral analysis method showed evidence for multiple quantal peaks in distributions of post-synaptic current amplitudes. Consistent with the observed changes in potency, estimated quantal size (Q) increased with potentiation and decreased with depression. A change in potency, which is thought to reflect post-synaptic expression mechanisms, was followed within seconds to minutes by a change in success rate, which is thought to reflect pre-synaptic expression mechanisms. Synaptic plasticity events may therefore consist of changes that occur on both sides of a synapse in a temporally coordinated fashion.
Collapse
Affiliation(s)
- Daniel H O'Connor
- Department of Molecular Biology and Program in Neuroscience, Princeton University, Princeton, New Jersey 08544, USA.
| | | | | |
Collapse
|
35
|
Chan CS, Levenson JM, Mukhopadhyay PS, Zong L, Bradley A, Sweatt JD, Davis RL. Alpha3-integrins are required for hippocampal long-term potentiation and working memory. Learn Mem 2007; 14:606-15. [PMID: 17848500 PMCID: PMC1994082 DOI: 10.1101/lm.648607] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Integrins comprise a large family of heterodimeric, transmembrane cell adhesion receptors that mediate diverse neuronal functions in the developing and adult CNS. Recent pharmacological and genetic studies have suggested that beta1-integrins are critical in synaptic plasticity and memory formation. To further define the role of integrins in these processes, we generated a postnatal forebrain and excitatory neuron-specific knockout of alpha3-integrin, one of several binding partners for beta1 subunit. At hippocampal Schaffer collateral-CA1 synapses, deletion of alpha3-integrin resulted in impaired long-term potentiation (LTP). Basal synaptic transmission and paired-pulse facilitation were normal in the absence of alpha3-integrin. Behavioral studies demonstrated that the mutant mice were selectively defective in a hippocampus-dependent, nonmatch-to-place working memory task, but were normal in other hippocampus-dependent spatial tasks. The impairment in LTP and working memory is similar to that observed in beta1-integrin conditional knockout mice, suggesting that alpha3-integrin is the functional binding partner for beta1 for these processes in the forebrain.
Collapse
Affiliation(s)
- Chi-Shing Chan
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jonathan M. Levenson
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Pharmacology and The Waisman Center, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin 53706, USA
| | - Partha S. Mukhopadhyay
- Department of Pharmacology and The Waisman Center, University of Wisconsin School of Medicine & Public Health, Madison, Wisconsin 53706, USA
| | - Lin Zong
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Allan Bradley
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - J. David Sweatt
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Ronald L. Davis
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, USA
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas 77030, USA
- Corresponding author.E-mail ; fax (713) 798-8005
| |
Collapse
|
36
|
Bozdagi O, Nagy V, Kwei KT, Huntley GW. In vivo roles for matrix metalloproteinase-9 in mature hippocampal synaptic physiology and plasticity. J Neurophysiol 2007; 98:334-44. [PMID: 17493927 PMCID: PMC4415272 DOI: 10.1152/jn.00202.2007] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Extracellular proteolysis is an important regulatory nexus for coordinating synaptic functional and structural plasticity, but the identity of such proteases is incompletely understood. Matrix metalloproteinases (MMPs) have well-known, mostly deleterious roles in remodeling after injury or stroke, but their role in nonpathological synaptic plasticity and function in intact adult brains has not been extensively investigated. Here we address the role of MMP-9 in hippocampal synaptic plasticity using both gain- and loss-of-function approaches in urethane-anesthetized adult rats. Acute blockade of MMP-9 proteolytic activity with inhibitors or neutralizing antibodies impairs maintenance, but not induction, of long-term potentiation (LTP) at synapses formed between Schaffer-collaterals and area CA1 dendrites. LTP is associated with significant increases in levels of MMP-9 and proteolytic activity within the potentiated neuropil. By introducing a novel application of gelatin-substrate zymography in vivo, we find that LTP is associated with significantly elevated numbers of gelatinolytic puncta in the potentiated neuropil that codistribute with immunolabeling for MMP-9 and for markers of synapses and dendrites. Such increases in proteolytic activity require NMDA receptor activation. Exposing intact area CA1 neurons to recombinant-active MMP-9 induces a slow synaptic potentiation that mutually occludes, and is occluded by, tetanically evoked potentiation. Taken together, our data reveal novel roles for MMP-mediated proteolysis in regulating nonpathological synaptic function and plasticity in mature hippocampus.
Collapse
Affiliation(s)
- Ozlem Bozdagi
- Fishberg Dept of Neuroscience, The Mount Sinai School of Medicine, New York, NY 10029-6574, USA
| | | | | | | |
Collapse
|
37
|
Wang Q, Klyubin I, Wright S, Griswold-Prenner I, Rowan MJ, Anwyl R. Alpha v integrins mediate beta-amyloid induced inhibition of long-term potentiation. Neurobiol Aging 2007; 29:1485-93. [PMID: 17442458 DOI: 10.1016/j.neurobiolaging.2007.03.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2007] [Revised: 03/13/2007] [Accepted: 03/17/2007] [Indexed: 11/22/2022]
Abstract
Beta-amyloid (Abeta) is the principal component of the extracellular plaques present in patients with Alzheimer's disease. Several studies have recently shown that acutely applied Abeta inhibits the induction of LTP in the hippocampus. In the present studies, we have investigated the role of integrins in such Abeta-mediated block of LTP in the dentate gyrus in vitro and in the CA1 in vivo. Selective antibodies to the alpha v integrin subunit were found to prevent the Abeta inhibition of LTP, both in the dentate gyrus in vitro and in the CA1 in vivo. In contrast, two control antibodies did not prevent such action of Abeta. In addition, a small molecule nonpeptide antagonist of alpha v-containing integrins and two other antagonistic ligands of integrins, superfibronectin and the disintegrin echistatin, also prevented the Abeta inhibition of LTP. These studies indicate that alpha v integrins may be important mediators of synaptic dysfunction prior to neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Qinwen Wang
- Department of Physiology, Trinity College, Dublin 2, Ireland
| | | | | | | | | | | |
Collapse
|
38
|
Watson PMD, Humphries MJ, Relton J, Rothwell NJ, Verkhratsky A, Gibson RM. Integrin-binding RGD peptides induce rapid intracellular calcium increases and MAPK signaling in cortical neurons. Mol Cell Neurosci 2007; 34:147-54. [PMID: 17150373 DOI: 10.1016/j.mcn.2006.10.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2006] [Revised: 10/12/2006] [Accepted: 10/17/2006] [Indexed: 10/23/2022] Open
Abstract
Integrins mediate cell adhesion to the extracellular matrix and initiate intracellular signaling. They play key roles in the central nervous system (CNS), participating in synaptogenesis, synaptic transmission and memory formation, but their precise mechanism of action remains unknown. Here we show that the integrin ligand-mimetic peptide GRGDSP induced NMDA receptor-dependent increases in intracellular calcium levels within seconds of presentation to primary cortical neurons. These were followed by transient activation and nuclear translocation of the ERK1/2 mitogen-activated protein kinase. RGD-induced effects were reduced by the NMDA receptor antagonist MK801, and ERK1/2 signaling was specifically inhibited by ifenprodil and PP2, indicating a functional connection between integrins, Src and NR2B-containing NMDA receptors. GRGDSP peptides were not significantly neuroprotective against excitotoxic insults. These results demonstrate a previously undescribed, extremely rapid effect of RGD peptide binding to integrins on cortical neurons that implies a close, functionally relevant connection between adhesion receptors and synaptic transmission.
Collapse
Affiliation(s)
- P Marc D Watson
- Faculty of Life Sciences, Michael Smith Building, University of Manchester, Manchester, M13 9PT, UK
| | | | | | | | | | | |
Collapse
|
39
|
Lynch G, Rex CS, Gall CM. LTP consolidation: Substrates, explanatory power, and functional significance. Neuropharmacology 2007; 52:12-23. [PMID: 16949110 DOI: 10.1016/j.neuropharm.2006.07.027] [Citation(s) in RCA: 146] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Revised: 07/05/2006] [Accepted: 07/17/2006] [Indexed: 12/18/2022]
Abstract
Long-term potentiation (LTP) resembles memory in that it is initially unstable and then, over about 30 min, becomes increasingly resistant to disruption. Here we present an hypothesis to account for this initial consolidation effect and consider implications that follow from it. Anatomical studies indicate that LTP is accompanied by changes in spine morphology and therefore likely involves cytoskeletal changes. Accordingly, theta bursts initiate calpain-mediated proteolysis of the actin cross-linking protein spectrin and trigger actin polymerization in spine heads, two effects indicative of cytoskeletal reorganization. Polymerization occurs within 2 min, has the same threshold as LTP, is dependent on integrins, and becomes resistant to disruption over 30 min. We propose that the stabilization of the new cytoskeletal organization, and thus of a new spine morphology, underlies the initial phase of LTP consolidation. This hypothesis helps explain the diverse array of proteins and signaling cascades implicated in LTP, as well as the often-contradictory results about contributions of particular molecules. It also provides a novel explanation for why LTP is potently modulated by factors likely to be released during theta trains (e.g., BDNF). Finally, building on evidence that normal patterns of activity reverse LTP, we suggest that consolidation provides a delay that allows brain networks to sculpt newly formed memories.
Collapse
Affiliation(s)
- Gary Lynch
- Department of Psychiatry and Human Behavior, Gillespie Neuroscience Research Facility, University of California, Irvine, CA 92697-4292, USA.
| | | | | |
Collapse
|
40
|
Bramham CR. Control of synaptic consolidation in the dentate gyrus: mechanisms, functions, and therapeutic implications. PROGRESS IN BRAIN RESEARCH 2007; 163:453-71. [PMID: 17765733 DOI: 10.1016/s0079-6123(07)63025-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Synaptic consolidation refers to the development and stabilization of protein synthesis-dependent modifications of synaptic strength as observed during long-term potentiation (LTP) and long-term depression (LTD). Activity-dependent changes in synaptic strength are thought to underlie memory storage and other adaptive responses of the nervous systems of importance in mood stability, reward behavior, and pain control. This chapter focuses on the mechanisms and functions of synaptic consolidation in the dentate gyrus, a critical structure not only in hippocampal memory function, but also in regulation of stress responses and cognitive aspects of depression. Recent evidence suggests that synaptic consolidation at excitatory medial perforant path-granule cell synapses requires brain-derived neurotrophic factor (BDNF) signaling and induction of the immediate early gene activity-regulated cytoskeleton-associated protein (Arc). Arc mRNA is strongly induced and transported to dendritic processes following high-frequency stimulation (HFS) that induces LTP in the rat dentate gyrus in vivo. Sustained synthesis of Arc during a surprisingly protracted time-window is required for hyperphosphorylation of actin depolymerizing factor/cofilin and local expansion of the actin cytoskeleton in vivo. Furthermore, this process of Arc-dependent synaptic consolidation is activated in response to brief infusion of BDNF. Microarray expression profiling has revealed a panel of BDNF-regulated genes that may cooperate with Arc during synaptic consolidation. In addition to regulating gene expression, BDNF signaling modulates the fine localization and biochemical activation of the translation machinery. By modulating the spatial and temporal translation of newly induced (Arc) and constitutively-expressed mRNA in dendrites, BDNF may effectively control the window of synaptic consolidation. Dysregulation of BDNF synthesis and Arc function, specifically within the dentate gyrus, is linked to behavioral symptoms and cognitive deficits in animal models of depression and Alzheimer's disease. Therapeutics strategies targeting synaptic consolidation hold promise for the future.
Collapse
Affiliation(s)
- Clive R Bramham
- Department of Biomedicine and Bergen Mental Health Research Center, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway.
| |
Collapse
|
41
|
Jovanova-Nesic K, Shoenfeld Y. MMP-2, VCAM-1 and NCAM-1 expression in the brain of rats with experimental autoimmune encephalomyelitis as a trigger mechanism for synaptic plasticity and pathology. J Neuroimmunol 2006; 181:112-21. [PMID: 17064783 DOI: 10.1016/j.jneuroim.2006.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2006] [Revised: 08/16/2006] [Accepted: 08/28/2006] [Indexed: 11/30/2022]
Abstract
The neural cell adhesion molecules (NCAMs), and vascular cell adhesion molecules (VCAMs) that regulate cell-to-extracellular matrix adhesion, and matrix metalloproteinases (MMPs), modulating the extracellular matrix (ECM), are considered to play an important role in the pathogenesis of experimental autoimmune encephalomyelitis (EAE). Clinical signs appearance and significant increases of MMP-2 expression in CA1 and CA3 subdomains of the hippocampus and around the central canal of the cervical spinal cord, with the clusters of VCAM-1(+) immunoreactive cells localized in the choroid plexus epithelium and hypothalamo-hypophyses portal vessel system indicate an inflammation in acute EAE. Decreased NCAM-1 expression in CA1 and CA3 fields of the hippocampus, and in a lesser degree in the basal ganglia, limbic structure and cervical spinal cord, support the concept that the demyelinating neuroinflammatory damage in an autoimmune brain affect synaptic organization of the brain, altering the balance between extracellular proteases and cell adhesion molecules which appears to be critical for both the brain plasticity and autoimmune processes.
Collapse
Affiliation(s)
- Katica Jovanova-Nesic
- Immunology Research Center, Department of Neuroimmunology, Belgrade, Serbia and Montenegro
| | | |
Collapse
|
42
|
Gui P, Wu X, Ling S, Stotz SC, Winkfein RJ, Wilson E, Davis GE, Braun AP, Zamponi GW, Davis MJ. Integrin Receptor Activation Triggers Converging Regulation of Cav1.2 Calcium Channels by c-Src and Protein Kinase A Pathways. J Biol Chem 2006; 281:14015-25. [PMID: 16554304 DOI: 10.1074/jbc.m600433200] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
L-type, voltage-gated Ca2+ channels (CaL) play critical roles in brain and muscle cell excitability. Here we show that currents through heterologously expressed neuronal and smooth muscle CaL channel isoforms are acutely potentiated following alpha5beta1 integrin activation. Only the alpha1C pore-forming channel subunit is critical for this process. Truncation and site-directed mutagenesis strategies reveal that regulation of Cav1.2 by alpha5beta1 integrin requires phosphorylation of alpha1C C-terminal residues Ser1901 and Tyr2122. These sites are known to be phosphorylated by protein kinase A (PKA) and c-Src, respectively, and are conserved between rat neuronal (Cav1.2c) and smooth muscle (Cav1.2b) isoforms. Kinase assays are consistent with phosphorylation of these two residues by PKA and c-Src. Following alpha5beta1 integrin activation, native CaL channels in rat arteriolar smooth muscle exhibit potentiation that is completely blocked by combined PKA and Src inhibition. Our results demonstrate that integrin-ECM interactions are a common mechanism for the acute regulation of CaL channels in brain and muscle. These findings are consistent with the growing recognition of the importance of integrin-channel interactions in cellular responses to injury and the acute control of synaptic and blood vessel function.
Collapse
Affiliation(s)
- Peichun Gui
- Department of Medical Pharmacology & Physiology, University of Missouri School of Medicine, Columbia, Missouri 65212, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Bronson NW, Hamilton JS, Han M, Li PA, Hornstra I, Horowitz JM, Horwitz BA. LOXL null mice demonstrate selective dentate structural changes but maintain dentate granule cell and CA1 pyramidal cell potentiation in the hippocampus. Neurosci Lett 2005; 390:118-22. [PMID: 16157454 DOI: 10.1016/j.neulet.2005.08.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 07/26/2005] [Accepted: 08/04/2005] [Indexed: 11/29/2022]
Abstract
Lysyl oxidase-like protein (LOXL), part of the lysyl oxidase copper-dependent amine oxidase family, is expressed in the extracellular matrix and in the nucleus. It likely plays a role in cross-linking collagen and elastin, possibly modulating cellular functions. Immunohistochemical studies show the presence of LOXL in the pyramidal cell layer of the hippocampus; and in this study, we report that cells in the granule cell layer have significantly smaller somas in LOXL -/- compared to LOXL +/+ mice. In addition we tested the hypothesis that these structural alterations in the dentate granule layer were associated with synaptic efficacy and thus muted long-term potentiation in mice lacking the protein. Electrical recordings were obtained in 300-mum hippocampal slices in dentate and CA1 pyramidal cell layers in age-matched wild type and LOXL null mice. Potentiation in the CA1 cell layer of 10 LOXL -/- and 8 LOXL +/+ mice was 191.0+/-9.3% and 181.6+/-9.1%, respectively (mean+/-S.E.M.). Dentate potentiation was 120.8+/-7.0% and 121.0+/-3.4% in 11 LOXL -/- and 11 LOXL +/+ mice, respectively. No phenotypic difference in potentiation of population spike amplitude (or in EPSP slope) in either layer was observed. Thus, contrary to expectation, structural changes in the hippocampus of LOXL -/- mice did not affect synaptic remodeling in a manner that impaired the establishment of LTP.
Collapse
Affiliation(s)
- Nathan W Bronson
- Section of Neurobiology, Physiology, and Behavior, University of California, Davis, Davis, CA 95616, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Tamura H, Ishikawa Y, Hino N, Maeda M, Yoshida S, Kaku S, Shiosaka S. Neuropsin is essential for early processes of memory acquisition and Schaffer collateral long-term potentiation in adult mouse hippocampus in vivo. J Physiol 2005; 570:541-51. [PMID: 16308352 PMCID: PMC1479887 DOI: 10.1113/jphysiol.2005.098715] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Long-term potentiation (LTP) is thought to be particularly important in the acquisition of hippocampus-associated memory, in part because it develops quickly and persists for indefinite periods. Extracellular proteolysis has been hypothesized to contribute to LTP by modifying adhesive relations of synapses and thus the morphology of excitatory synapses. Here we report that neuropsin (NP), an extracellular serine protease, is critically involved in the formation of both the potentiation effect and hippocampus-dependent forms of memory. NP-knockout mice were significantly impaired in the Morris water maze and Y-mazes and failed to exhibit early phase LTP induced by a single tetanus. Potentiation was also impaired or completely blocked by in vivo application of a specific inhibitor or a neutralizing monoclonal antibody for NP. Intriguingly, recombinant (r-) NP alone, without tetanic stimulation, elicited either long-lasting potentiation or depression, depending on the applied dose. The r-NP-elicited potentiation was occluded by prior induction of LTP, while theta-burst-elicited LTP was occluded by application of r-NP alone, suggesting that the two forms of plasticity have a common signalling pathway. r-NP-elicited potentiation and depression increased phosphorylation at different sites on the GluR1 subunit of the AMPA receptor that had previously been associated with LTP or long-term depression. Thus, we conclude that NP is necessary for establishment of LTP and has a significant role in memory acquisition.
Collapse
Affiliation(s)
- Hideki Tamura
- Division of Structural Cell Biology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | | | | | | | | | | | | |
Collapse
|
45
|
Zhang TA, Hendricson AW, Wilkemeyer MF, Lippmann MJ, Charness ME, Morrisett RA. Synergistic effects of the peptide fragment D-NAPVSIPQ on ethanol inhibition of synaptic plasticity and NMDA receptors in rat hippocampus. Neuroscience 2005; 134:583-93. [PMID: 15963648 DOI: 10.1016/j.neuroscience.2005.04.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2004] [Revised: 04/14/2005] [Accepted: 04/16/2005] [Indexed: 11/22/2022]
Abstract
The L1 cell adhesion molecule has been implicated in ethanol teratogenesis as well as NMDAR-dependent long-term potentiation (LTP) of synaptic transmission, a process thought to be critical for neural development. Ethanol inhibits LTP at least in part by interacting with NMDA receptors. Ethanol also inhibits L1-mediated cell adhesion in a manner that is prevented by an octapeptide, D-NAPVSIPQ (D-NAP), as well as long chain alcohols such as 1-octanol. Here we analyzed the effects of D-NAP and 1-octanol on ethanol modulation of LTP induced by theta burst stimulation in two subfields of the rat hippocampus, the dentate gyrus and area CA1. When theta burst stimulation was delivered in ethanol (50 mM), LTP was inhibited by about 50%. Surprisingly, when D-NAP (10(-7) M) and ethanol were co-applied or applied sequentially, LTP was completely absent. The effects of D-NAP were persistent, since delivery of a second theta burst stimulation following washout of D-NAP and ethanol elicited minimal plasticity. Application of D-NAP alone had no effect on LTP induction or expression. The synergistic effect of D-NAP on ethanol inhibition of LTP was concentration-dependent since D-NAP (10(-10) M) had an intermediate effect, while D-NAP (10(-13) M) had no effect on ethanol suppression of LTP. These observations were also replicated with a different ethanol antagonist, 1-octanol, in area CA1. To address the mechanisms underlying this long-lasting suppression of LTP, the sensitivity of pharmacologically isolated NMDAR extracellular field potentials to combinations of D-NAP and ethanol was determined. D-NAP (10(-7)M) alone had no effect on NMDA extracellular field potentials; however, the peptide significantly increased the inhibitory action of ethanol on NMDA extracellular field potential. The findings suggest that D-NAP and 1-octanol selectively interact with NMDA receptors in an ethanol-dependent manner, further implicating the L1 cell adhesion molecule in alcohol-related brain disorders.
Collapse
Affiliation(s)
- T A Zhang
- The College of Pharmacy and The Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712-1074, USA
| | | | | | | | | | | |
Collapse
|
46
|
Gibson RM, Craig SE, Heenan L, Tournier C, Humphries MJ. Activation of integrin alpha5beta1 delays apoptosis of Ntera2 neuronal cells. Mol Cell Neurosci 2005; 28:588-98. [PMID: 15737747 DOI: 10.1016/j.mcn.2004.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2004] [Revised: 10/28/2004] [Accepted: 11/03/2004] [Indexed: 10/25/2022] Open
Abstract
Integrins are dynamic membrane proteins that mediate adhesion of cells to the extracellular matrix. Integrins initiate signal transduction, alone and cooperatively with growth factor receptors, and regulate many aspects of cell behavior. We report here that alpha5beta1-mediated adhesion of Ntera2 neuronal cells to fibronectin decreased apoptosis in response to serum withdrawal. Adhesion induced phosphorylation of FAK, and strongly increased the AKT phosphorylation induced by growth factors, demonstrating for the first time in neuronal cells that integrin-mediated adhesion and growth factors cooperate to regulate AKT activity. Integrins exist on cells in different activation states, and cell survival on fibronectin was enhanced by the antibody 12G10, that modulates the conformation of beta1 in favor of its active form. The antibody 12G10 specifically delayed loss of phosphorylation of AKT on serine 473, and GSK-3beta on serine 9, induced by serum withdrawal, suggesting that these kinases are critical sensors of integrin activation on neuronal cells.
Collapse
Affiliation(s)
- Rosemary M Gibson
- Faculty of Life Sciences, University of Manchester, 1.124 Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| | | | | | | | | |
Collapse
|
47
|
Bernard-Trifilo JA, Kramár EA, Torp R, Lin CY, Pineda EA, Lynch G, Gall CM. Integrin signaling cascades are operational in adult hippocampal synapses and modulate NMDA receptor physiology. J Neurochem 2005; 93:834-49. [PMID: 15857387 DOI: 10.1111/j.1471-4159.2005.03062.x] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Integrin class adhesion proteins are concentrated at adult brain synapses. Whether synaptic integrins engage kinase signaling cascades has not been determined, but is a question of importance to ideas about integrin involvement in functional synaptic plasticity. Accordingly, synaptoneurosomes from adult rat brain were used to test if matrix ligands activate integrin-associated tyrosine kinases, and if integrin signaling targets include NMDA-class glutamate neurotransmitter receptors. The integrin ligand peptide Gly-Arg-Gly-Asp-Ser-Pro (GRGDSP) induced rapid (within 5 min) and robust increases in tyrosine phosphorylation of focal adhesion kinase, proline-rich tyrosine kinase 2 and Src family kinases. Increases were similarly induced by the native ligand fibronectin, blocked with neutralizing antibodies to beta1 integrin, and not obtained with control peptides, indicating that kinase activation was integrin-mediated. Both GRGDSP and fibronectin caused rapid Src kinase-dependent increases in tyrosine phosphorylation of NMDA receptor subunits NR2A and NR2B in synaptoneurosomes and acute hippocampal slices. Tests of the physiological significance of the latter result showed that ligand treatment caused a rapid and beta1 integrin-dependent increase in NMDA receptor-mediated synaptic responses. These results provide the first evidence that, in adult brain, synaptic integrins activate local kinase cascades with potent effects on the operation of nearby neurotransmitter receptors implicated in synaptic plasticity.
Collapse
|
48
|
Lin CY, Lynch G, Gall CM. AMPA receptor stimulation increases alpha5beta1 integrin surface expression, adhesive function and signaling. J Neurochem 2005; 94:531-46. [PMID: 16000124 PMCID: PMC2366053 DOI: 10.1111/j.1471-4159.2005.03203.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Integrin proteins are critical for stabilization of hippocampal long-term potentiation but the mechanisms by which integrin activities are involved in synaptic transmission are not known. The present study tested whether activation of alpha-amino-3-hydroxy-5-methylisoxazole-4-proprionate (AMPA) class glutamate receptors increases surface expression of alpha5beta1 integrin implicated in synaptic potentiation. Surface protein biotinylation assays demonstrated that AMPA treatment of COS7 cells expressing GluR1 homomeric AMPA receptors increased membrane insertion and steady-state surface levels of alpha5 and beta1 subunits. Treated cells exhibited increased adhesion to fibronectin- and anti-alpha5-coated substrates and tyrosine kinase signaling elicited by fibronectin-substrate adhesion, as expected if new surface receptors are functional. Increased surface expression did not occur in calcium-free medium and was blocked by the protein kinase C inhibitor chelerythrine chloride and the exocytosis inhibitor brefeldin A. AMPA treatment similarly increased alpha5 and beta1 surface expression in dissociated neurons and cultured hippocampal slices. In both neuronal preparations AMPA-induced integrin trafficking was blocked by combined antagonism of NMDA receptor and L-type voltage-sensitive calcium channel activities but was not induced by NMDA treatment alone. These results provide the first evidence that glutamate receptor activation increases integrin surface expression and function, and suggest a novel mechanism by which synaptic activity can engage a volley of new integrin signaling in coordination with, and probably involved in, stabilization of synaptic potentiation.
Collapse
Affiliation(s)
- Ching-Yi Lin
- Department of Anatomy and Neurobiology, University of California, Irvine, California 92697-4292, USA
| | | | | |
Collapse
|
49
|
Bramham CR, Messaoudi E. BDNF function in adult synaptic plasticity: the synaptic consolidation hypothesis. Prog Neurobiol 2005; 76:99-125. [PMID: 16099088 DOI: 10.1016/j.pneurobio.2005.06.003] [Citation(s) in RCA: 878] [Impact Index Per Article: 43.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2005] [Revised: 05/09/2005] [Accepted: 06/16/2005] [Indexed: 12/19/2022]
Abstract
Interest in BDNF as an activity-dependent modulator of neuronal structure and function in the adult brain has intensified in recent years. Localization of BDNF-TrkB to glutamate synapses makes this system attractive as a dynamic, activity-dependent regulator of excitatory transmission and plasticity. Despite individual breakthroughs, an integrated understanding of BDNF function in synaptic plasticity is lacking. Here, we attempt to distill current knowledge of the molecular mechanisms and function of BDNF in LTP. BDNF activates distinct mechanisms to regulate the induction, early maintenance, and late maintenance phases of LTP. Evidence from genetic and pharmacological approaches is reviewed and tabulated. The specific contribution of BDNF depends on the stimulus pattern used to induce LTP, which impacts the duration and perhaps the subcellular site of BDNF release. Particular attention is given to the role of BDNF as a trigger for protein synthesis-dependent late phase LTP--a process referred to as synaptic consolidation. Recent experiments suggest that BDNF activates synaptic consolidation through transcription and rapid dendritic trafficking of mRNA encoded by the immediate early gene, Arc. A model is proposed in which BDNF signaling at glutamate synapses drives the translation of newly transported (Arc) and locally stored (i.e., alphaCaMKII) mRNA in dendrites. In this model BDNF tags synapses for mRNA capture, while Arc translation defines a critical window for synaptic consolidation. The biochemical mechanisms by which BDNF regulates local translation are also discussed. Elucidation of these mechanisms should shed light on a range of adaptive brain responses including memory and mood resilience.
Collapse
Affiliation(s)
- Clive R Bramham
- Department of Biomedicine, Bergen Mental Health Research Center, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.
| | | |
Collapse
|
50
|
Hernandez RV, Navarro MM, Rodriguez WA, Martinez JL, LeBaron RG. Differences in the magnitude of long-term potentiation produced by theta burst and high frequency stimulation protocols matched in stimulus number. ACTA ACUST UNITED AC 2005; 15:6-13. [PMID: 15878145 DOI: 10.1016/j.brainresprot.2005.02.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2004] [Revised: 01/31/2005] [Accepted: 02/08/2005] [Indexed: 10/25/2022]
Abstract
Theta-burst stimulation (TBS: four pulses at 100 Hz repeated with 200 ms inter-burst-intervals) and another commonly used high-frequency stimulation protocol (HFS: 1 s burst of equally spaced pulses at 100 Hz) were compared for the magnitude of LTP produced in rat hippocampal slices. The total number of pulses applied during tetanus (TET) was either 40, 100, 200, or 300. In a conventional analysis of the last 10 min of the post-TET period, a two-way ANOVA revealed no difference either in LTP of the field excitatory post-synaptic potential (fEPSP) between TBS and HFS or differences across pulse number at 40, 100, or 200 pulses. At 300 pulses, there was a significant main effect by pulse number but not by protocol. A linear regression analysis showed that stimulation protocol accounted for only about 10% of the change in magnitude while pulse number contributed to 30% of the change. However, when an extended analysis of the same data was performed across the entire post-TET period with a repeated-measure ANOVA, a small but persistent increase in TBS over HFS at 200 pulses was significant. A difference between TBS and HFS at 300 pulses that occurred only during the early phase of LTP was also significant. These results suggest that, over a range of stimuli, the number of pulses in an induction protocol, rather than the pattern of stimulation, determines the magnitude of late phase LTP, while TBS produces greater potentiation than HFS in the early phase of LTP with higher TET number.
Collapse
Affiliation(s)
- Ruben V Hernandez
- Department of Biology and the Cajal Neuroscience Institute, The University of Texas at San Antonio, 6900 North Loop 1604 West, San Antonio, TX 78249, USA.
| | | | | | | | | |
Collapse
|