1
|
Grubisha MJ, Sun T, Eisenman L, Erickson SL, Chou S, Helmer CD, Trudgen MT, Ding Y, Homanics GE, Penzes P, Wills ZP, Sweet RA. A Kalirin missense mutation enhances dendritic RhoA signaling and leads to regression of cortical dendritic arbors across development. Proc Natl Acad Sci U S A 2021; 118:e2022546118. [PMID: 34848542 PMCID: PMC8694055 DOI: 10.1073/pnas.2022546118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 09/15/2021] [Indexed: 11/26/2022] Open
Abstract
Normally, dendritic size is established prior to adolescence and then remains relatively constant into adulthood due to a homeostatic balance between growth and retraction pathways. However, schizophrenia is characterized by accelerated reductions of cerebral cortex gray matter volume and onset of clinical symptoms during adolescence, with reductions in layer 3 pyramidal neuron dendritic length, complexity, and spine density identified in multiple cortical regions postmortem. Nogo receptor 1 (NGR1) activation of the GTPase RhoA is a major pathway restricting dendritic growth in the cerebral cortex. We show that the NGR1 pathway is stimulated by OMGp and requires the Rho guanine nucleotide exchange factor Kalirin-9 (KAL9). Using a genetically encoded RhoA sensor, we demonstrate that a naturally occurring missense mutation in Kalrn, KAL-PT, that was identified in a schizophrenia cohort, confers enhanced RhoA activitation in neuronal dendrites compared to wild-type KAL. In mice containing this missense mutation at the endogenous locus, there is an adolescent-onset reduction in dendritic length and complexity of layer 3 pyramidal neurons in the primary auditory cortex. Spine density per unit length of dendrite is unaffected. Early adult mice with these structural deficits exhibited impaired detection of short gap durations. These findings provide a neuropsychiatric model of disease capturing how a mild genetic vulnerability may interact with normal developmental processes such that pathology only emerges around adolescence. This interplay between genetic susceptibility and normal adolescent development, both of which possess inherent individual variability, may contribute to heterogeneity seen in phenotypes in human neuropsychiatric disease.
Collapse
Affiliation(s)
- Melanie J Grubisha
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Tao Sun
- Department of Biostatistics, University of Pittsburgh, PA 15261
| | - Leanna Eisenman
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Susan L Erickson
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Shinnyi Chou
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Cassandra D Helmer
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Melody T Trudgen
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| | - Ying Ding
- Department of Biostatistics, University of Pittsburgh, PA 15261
| | - Gregg E Homanics
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15261
- Department of Anesthesiology and Perioperative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | - Peter Penzes
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Zachary P Wills
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15261
| | - Robert A Sweet
- Department of Psychiatry, Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213;
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213
| |
Collapse
|
2
|
Scott MC, Bedi SS, Olson SD, Sears CM, Cox CS. Microglia as therapeutic targets after neurological injury: strategy for cell therapy. Expert Opin Ther Targets 2021; 25:365-380. [PMID: 34029505 DOI: 10.1080/14728222.2021.1934447] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Microglia is the resident tissue macrophages of the central nervous system. Prolonged microglial activation often occurs after traumatic brain injury and is associated with deteriorating neurocognitive outcomes. Resolution of microglial activation is associated with limited tissue loss and improved neurocognitive outcomes. Limiting the prolonged pro-inflammatory response and the associated secondary tissue injury provides the rationale and scientific premise for considering microglia as a therapeutic target. AREAS COVERED In this review, we discuss markers of microglial activation, such as immunophenotype and microglial response to injury, including cytokine/chemokine release, free radical formation, morphology, phagocytosis, and metabolic shifts. We compare the origin and role in neuroinflammation of microglia and monocytes/macrophages. We review potential therapeutic targets to shift microglial polarization. Finally, we review the effect of cell therapy on microglia. EXPERT OPINION Dysregulated microglial activation after neurologic injury, such as traumatic brain injury, can worsen tissue damage and functional outcomes. There are potential targets in microglia to attenuate this activation, such as proteins and molecules that regulate microglia polarization. Cellular therapeutics that limit, but do not eliminate, the inflammatory response have improved outcomes in animal models by reducing pro-inflammatory microglial activation via secondary signaling. These findings have been replicated in early phase clinical trials.
Collapse
Affiliation(s)
- M Collins Scott
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston (Uthealth), USA
| | - Supinder S Bedi
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Candice M Sears
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Medical School at Houston, Houston, Texas, USA
| |
Collapse
|
3
|
Ernest James Phillips T, Maguire E. Phosphoinositides: Roles in the Development of Microglial-Mediated Neuroinflammation and Neurodegeneration. Front Cell Neurosci 2021; 15:652593. [PMID: 33841102 PMCID: PMC8032904 DOI: 10.3389/fncel.2021.652593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/08/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia are increasingly recognized as vital players in the pathology of a variety of neurodegenerative conditions including Alzheimer’s (AD) and Parkinson’s (PD) disease. While microglia have a protective role in the brain, their dysfunction can lead to neuroinflammation and contributes to disease progression. Also, a growing body of literature highlights the seven phosphoinositides, or PIPs, as key players in the regulation of microglial-mediated neuroinflammation. These small signaling lipids are phosphorylated derivates of phosphatidylinositol, are enriched in the brain, and have well-established roles in both homeostasis and disease.Disrupted PIP levels and signaling has been detected in a variety of dementias. Moreover, many known AD disease modifiers identified via genetic studies are expressed in microglia and are involved in phospholipid metabolism. One of these, the enzyme PLCγ2 that hydrolyzes the PIP species PI(4,5)P2, displays altered expression in AD and PD and is currently being investigated as a potential therapeutic target.Perhaps unsurprisingly, neurodegenerative conditions exhibiting PIP dyshomeostasis also tend to show alterations in aspects of microglial function regulated by these lipids. In particular, phosphoinositides regulate the activities of proteins and enzymes required for endocytosis, toll-like receptor signaling, purinergic signaling, chemotaxis, and migration, all of which are affected in a variety of neurodegenerative conditions. These functions are crucial to allow microglia to adequately survey the brain and respond appropriately to invading pathogens and other abnormalities, including misfolded proteins. AD and PD therapies are being developed to target many of the above pathways, and although not yet investigated, simultaneous PIP manipulation might enhance the beneficial effects observed. Currently, only limited therapeutics are available for dementia, and although these show some benefits for symptom severity and progression, they are far from curative. Given the importance of microglia and PIPs in dementia development, this review summarizes current research and asks whether we can exploit this information to design more targeted, or perhaps combined, dementia therapeutics. More work is needed to fully characterize the pathways discussed in this review, but given the strength of the current literature, insights in this area could be invaluable for the future of neurodegenerative disease research.
Collapse
Affiliation(s)
| | - Emily Maguire
- UK Dementia Research Institute at Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
4
|
Density of small dendritic spines and microtubule-associated-protein-2 immunoreactivity in the primary auditory cortex of subjects with schizophrenia. Neuropsychopharmacology 2019; 44:1055-1061. [PMID: 30795003 PMCID: PMC6461932 DOI: 10.1038/s41386-019-0350-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 02/15/2019] [Accepted: 02/16/2019] [Indexed: 12/22/2022]
Abstract
Previously, we demonstrated that dendritic spine density (DSD) in deep layer 3 of the primary auditory cortex (A1) is lower, due to having fewer small spines, in subjects with schizophrenia (SZ) than non-psychiatric control (NPC) subjects. We also previously demonstrated that microtubule-associated-protein-2 immunoreactivity (MAP2-IR) in A1 deep layer 3 is lower, and positively correlated with DSD, in SZ subjects. Here, we first sought to confirm these findings in an independent cohort of 25 SZ-NPC subject pairs (cohort 1). We used immunohistochemistry and confocal microscopy to measure DSD and MAP2-IR in A1 deep layer 3. Consistent with previous studies, both DSD and MAP2-IR were lower in SZ subjects. We then tested the hypothesis that MAP2-IR mediates the effect of SZ on DSD in a cohort of 45 SZ-NPC subject pairs (combined cohort) that included all subjects from cohort 1 and two previously studied cohorts. Based on the distribution of MAP2-IR values in NPC subjects, we categorized each SZ subject as having either low MAP2-IR (SZ MAP2-IR(low)) or normal MAP2-IR (SZ MAP2-IR(normal)). Among SZ MAP-IR(low) subjects, mean DSD was significantly lower than in NPC subjects. However, mean DSD did not differ between SZ MAP2-IR(normal) and NPC subjects. Moreover, MAP2-IR statistically mediated small spine differences, with lower MAP2-IR values associated with fewer small spines. Our findings confirm that low density of small spines and low MAP2-IR are robust SZ phenotypes and suggest that MAP2-IR mediates the effect of SZ on DSD.
Collapse
|
5
|
Franco Bocanegra DK, Nicoll JAR, Boche D. Innate immunity in Alzheimer's disease: the relevance of animal models? J Neural Transm (Vienna) 2017; 125:827-846. [PMID: 28516241 PMCID: PMC5911273 DOI: 10.1007/s00702-017-1729-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/27/2017] [Indexed: 12/12/2022]
Abstract
The mouse is one of the organisms most widely used as an animal model in biomedical research, due to the particular ease with which it can be handled and reproduced in laboratory. As a member of the mammalian class, mice share with humans many features regarding metabolic pathways, cell morphology and anatomy. However, important biological differences between mice and humans exist and must be taken into consideration when interpreting research results, to properly translate evidence from experimental studies into information that can be useful for human disease prevention and/or treatment. With respect to Alzheimer’s disease (AD), much of the experimental information currently known about this disease has been gathered from studies using mainly mice as models. Therefore, it is notably important to fully characterise the differences between mice and humans regarding important aspects of the disease. It is now widely known that inflammation plays an important role in the development of AD, a role that is not only a response to the surrounding pathological environment, but rather seems to be strongly implicated in the aetiology of the disease as indicated by the genetic studies. This review highlights relevant differences in inflammation and in microglia, the innate immune cell of the brain, between mice and humans regarding genetics and morphology in normal ageing, and the relationship of microglia with AD-like pathology, the inflammatory profile, and cognition. We conclude that some noteworthy differences exist between mice and humans regarding microglial characteristics, in distribution, gene expression, and states of activation. This may have repercussions in the way that transgenic mice respond to, and influence, the AD-like pathology. However, despite these differences, human and mouse microglia also show similarities in morphology and behaviour, such that the mouse is a suitable model for studying the role of microglia, as long as these differences are taken into consideration when delineating new strategies to approach the study of neurodegenerative diseases.
Collapse
Affiliation(s)
- Diana K Franco Bocanegra
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Mailpoint 806, Southampton, SO16 6YD, UK
| | - James A R Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Mailpoint 806, Southampton, SO16 6YD, UK.,Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton, Southampton, SO16 6YD, UK
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Mailpoint 806, Southampton, SO16 6YD, UK.
| |
Collapse
|
6
|
Avery AW, Crain J, Thomas DD, Hays TS. A human β-III-spectrin spinocerebellar ataxia type 5 mutation causes high-affinity F-actin binding. Sci Rep 2016; 6:21375. [PMID: 26883385 PMCID: PMC4756369 DOI: 10.1038/srep21375] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 01/21/2016] [Indexed: 01/06/2023] Open
Abstract
Spinocerebellar ataxia type 5 (SCA5) is a human neurodegenerative disease that stems from mutations in the SPTBN2 gene encoding the protein β-III-spectrin. Here we investigated the molecular consequence of a SCA5 missense mutation that results in a L253P substitution in the actin-binding domain (ABD) of β-III-spectrin. We report that the L253P substitution in the isolated β-III-spectrin ABD causes strikingly high F-actin binding affinity (Kd = 75.5 nM) compared to the weak F-actin binding affinity of the wild-type ABD (Kd = 75.8 μM). The mutation also causes decreased thermal stability (Tm = 44.6 °C vs 59.5 °C). Structural analyses indicate that leucine 253 is in a loop at the interface of the tandem calponin homology (CH) domains comprising the ABD. Leucine 253 is predicted to form hydrophobic contacts that bridge the CH domains. The decreased stability of the mutant indicates that these bridging interactions are probably disrupted, suggesting that the high F-actin binding affinity of the mutant is due to opening of the CH domain interface. These results support a fundamental role for leucine 253 in regulating opening of the CH domain interface and binding of the ABD to F-actin. This study indicates that high-affinity actin binding of L253P β-III-spectrin is a likely driver of neurodegeneration.
Collapse
Affiliation(s)
- Adam W Avery
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455
| | - Jonathan Crain
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - David D Thomas
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN 55455
| | - Thomas S Hays
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
7
|
Shelton MA, Newman JT, Gu H, Sampson AR, Fish KN, MacDonald ML, Moyer CE, DiBitetto JV, Dorph-Petersen KA, Penzes P, Lewis DA, Sweet RA. Loss of Microtubule-Associated Protein 2 Immunoreactivity Linked to Dendritic Spine Loss in Schizophrenia. Biol Psychiatry 2015; 78:374-85. [PMID: 25818630 PMCID: PMC4520801 DOI: 10.1016/j.biopsych.2014.12.029] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 11/25/2014] [Accepted: 12/19/2014] [Indexed: 02/05/2023]
Abstract
BACKGROUND Microtubule-associated protein 2 (MAP2) is a neuronal protein that plays a role in maintaining dendritic structure through its interaction with microtubules. In schizophrenia (Sz), numerous studies have revealed that the typically robust immunoreactivity (IR) of MAP2 is significantly reduced across several cortical regions. The relationship between MAP2-IR reduction and lower dendritic spine density, which is frequently reported in Sz, has not been explored in previous studies, and MAP2-IR loss has not been investigated in the primary auditory cortex (Brodmann area 41), a site of conserved pathology in Sz. METHODS Using quantitative spinning disk confocal microscopy in two cohorts of subjects with Sz and matched control subjects (Sz subjects, n = 20; control subjects, n = 20), we measured MAP2-IR and dendritic spine density and spine number in deep layer 3 of BA41. RESULTS Subjects with Sz exhibited a significant reduction in MAP2-IR. The reductions in MAP2-IR were not associated with neuron loss, loss of MAP2 protein, clinical confounders, or technical factors. Dendritic spine density and number also were reduced in Sz and correlated with MAP2-IR. In 12 (60%) subjects with Sz, MAP2-IR values were lower than the lowest values in control subjects; only in this group were spine density and number significantly reduced. CONCLUSIONS These findings demonstrate that MAP2-IR loss is closely linked to dendritic spine pathology in Sz. Because MAP2 shares substantial sequence, regulatory, and functional homology with MAP tau, the wealth of knowledge regarding tau biology and the rapidly expanding field of tau therapeutics provide resources for identifying how MAP2 is altered in Sz and possible leads to novel therapeutics.
Collapse
Affiliation(s)
- Micah A Shelton
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Jason T Newman
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hong Gu
- Department of Statistics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Allan R Sampson
- Department of Statistics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kenneth N Fish
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Matthew L MacDonald
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Caitlin E Moyer
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - James V DiBitetto
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Karl-Anton Dorph-Petersen
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - David A Lewis
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Robert A Sweet
- Translational Neuroscience Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Mental Illness Research, Education, and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania.
| |
Collapse
|
8
|
Abstract
Myelination of axons in the nervous system of vertebrates enables fast, saltatory impulse propagation, one of the best-understood concepts in neurophysiology. However, it took a long while to recognize the mechanistic complexity both of myelination by oligodendrocytes and Schwann cells and of their cellular interactions. In this review, we highlight recent advances in our understanding of myelin biogenesis, its lifelong plasticity, and the reciprocal interactions of myelinating glia with the axons they ensheath. In the central nervous system, myelination is also stimulated by axonal activity and astrocytes, whereas myelin clearance involves microglia/macrophages. Once myelinated, the long-term integrity of axons depends on glial supply of metabolites and neurotrophic factors. The relevance of this axoglial symbiosis is illustrated in normal brain aging and human myelin diseases, which can be studied in corresponding mouse models. Thus, myelinating cells serve a key role in preserving the connectivity and functions of a healthy nervous system.
Collapse
Affiliation(s)
- Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, D-37075 Göttingen, Germany; ,
| | | |
Collapse
|
9
|
Madry C, Attwell D. Receptors, ion channels, and signaling mechanisms underlying microglial dynamics. J Biol Chem 2015; 290:12443-50. [PMID: 25855789 DOI: 10.1074/jbc.r115.637157] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microglia, the innate immune cells of the CNS, play a pivotal role in brain injury and disease. Microglia are extremely motile; their highly ramified processes constantly survey the brain parenchyma, and they respond promptly to brain damage with targeted process movement toward the injury site. Microglia play a key role in brain development and function by pruning synapses during development, phagocytosing apoptotic newborn neurons, and regulating neuronal activity by direct microglia-neuron or indirect microglia-astrocyte-neuron interactions, which all depend on their process motility. This review highlights recent discoveries about microglial dynamics, focusing on the receptors, ion channels, and signaling pathways involved.
Collapse
Affiliation(s)
- Christian Madry
- From the Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - David Attwell
- From the Department of Neuroscience, Physiology & Pharmacology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| |
Collapse
|
10
|
Mikati MA, Grintsevich EE, Reisler E. Drebrin-induced stabilization of actin filaments. J Biol Chem 2013; 288:19926-38. [PMID: 23696644 DOI: 10.1074/jbc.m113.472647] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Drebrin is a mammalian neuronal protein that binds to and organizes filamentous actin (F-actin) in dendritic spines, the receptive regions of most excitatory synapses that play a crucial role in higher brain functions. Here, the structural effects of drebrin on F-actin were examined in solution. Depolymerization and differential scanning calorimetry assays show that F-actin is stabilized by the binding of drebrin. Drebrin inhibits depolymerization mainly at the barbed end of F-actin. Full-length drebrin and its C-terminal truncated constructs were used to clarify the domain requirements for these effects. The actin binding domain of drebrin decreases the intrastrand disulfide cross-linking of Cys-41 (in the DNase I binding loop) to Cys-374 (C-terminal) but increases the interstrand disulfide cross-linking of Cys-265 (hydrophobic loop) to Cys-374 in the yeast mutants Q41C and S265C, respectively. We also demonstrate, using solution biochemistry methods and EM, the rescue of filament formation by drebrin in different cases of longitudinal interprotomer contact perturbation: the T203C/C374S yeast actin mutant and grimelysin-cleaved skeletal actin (between Gly-42 and Val-43). Additionally, we show that drebrin rescues the polymerization of V266G/L267G, a hydrophobic loop yeast actin mutant with an impaired lateral interface formation between the two filament strands. Overall, our data suggest that drebrin stabilizes actin filaments through its effect on their interstrand and intrastrand contacts.
Collapse
Affiliation(s)
- Mouna A Mikati
- Department of Chemistry and Biochemistry, of California Los Angeles, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
11
|
Myelin Proteome Analysis: Methods and Implications for the Myelin Cytoskeleton. THE CYTOSKELETON 2013. [DOI: 10.1007/978-1-62703-266-7_15] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
12
|
Sargsyan SA, Blackburn DJ, Barber SC, Grosskreutz J, De Vos KJ, Monk PN, Shaw PJ. A comparison of in vitro properties of resting SOD1 transgenic microglia reveals evidence of reduced neuroprotective function. BMC Neurosci 2011; 12:91. [PMID: 21943126 PMCID: PMC3191510 DOI: 10.1186/1471-2202-12-91] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2011] [Accepted: 09/23/2011] [Indexed: 12/11/2022] Open
Abstract
Background Overexpression of mutant copper/zinc superoxide dismutase (SOD1) in rodents has provided useful models for studying the pathogenesis of amyotrophic lateral sclerosis (ALS). Microglia have been shown to contribute to ALS disease progression in these models, although the mechanism of this contribution remains to be elucidated. Here, we present the first evidence of the effects of overexpression of mutant (TG G93A) and wild type (TG WT) human SOD1 transgenes on a set of functional properties of microglia relevant to ALS progression, including expression of integrin β-1, spreading and migration, phagocytosis of apoptotic neuronal cell debris, and intracellular calcium changes in response to an inflammatory stimulus. Results TG SOD1 G93A but not TG SOD1 WT microglia had lower expression levels of the cell adhesion molecule subunit integrin β-1 than their NTG control cells [NTG (G93A) and NTG (WT), respectively, 92.8 ± 2.8% on TG G93A, 92.0 ± 6.6% on TG WT, 100.0 ± 1.6% on NTG (G93A), and 100.0 ± 2.7% on NTG (WT) cells], resulting in decreased spreading ability, with no effect on ability to migrate. Both TG G93A and TG WT microglia had reduced capacity to phagocytose apoptotic neuronal cell debris (13.0 ± 1.3% for TG G93A, 16.5 ± 1.9% for TG WT, 28.6 ± 1.8% for NTG (G93A), and 26.9 ± 2.8% for NTG (WT) cells). Extracellular stimulation of microglia with ATP resulted in smaller increase in intracellular free calcium in TG G93A and TG WT microglia relative to NTG controls (0.28 ± 0.02 μM for TG G93A, 0.24 ± 0.03 μM for TG WT, 0.39 ± 0.03 μM for NTG (G93A), and 0.37 ± 0.05 μM for NTG (WT) microglia). Conclusions These findings indicate that, under resting conditions, microglia from mutant SOD1 transgenic mice have a reduced capacity to elicit physiological responses following tissue disturbances and that higher levels of stimulatory signals, and/or prolonged stimulation may be necessary to initiate these responses. Overall, resting mutant SOD1-overexpressing microglia may have reduced capacity to function as sensors of disturbed tissue/cellular homeostasis in the CNS and thus have reduced neuroprotective function.
Collapse
Affiliation(s)
- Siranush A Sargsyan
- Department of Medicine, University of Colorado Denver School of Medicine, CO, USA.
| | | | | | | | | | | | | |
Collapse
|
13
|
Farris SM. Are mushroom bodies cerebellum-like structures? ARTHROPOD STRUCTURE & DEVELOPMENT 2011; 40:368-79. [PMID: 21371566 DOI: 10.1016/j.asd.2011.02.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Revised: 02/08/2011] [Accepted: 02/19/2011] [Indexed: 05/20/2023]
Abstract
The mushroom bodies are distinctive neuropils in the protocerebral brain segments of many protostomes. A defining feature of mushroom bodies is their intrinsic neurons, masses of cytoplasm-poor globuli cells that form a system of lobes with their densely-packed, parallel-projecting axon-like processes. In insects, the role of the mushroom bodies in olfactory processing and associative learning and memory has been studied in depth, but several lines of evidence suggest that the function of these higher brain centers cannot be restricted to these roles. The present account considers whether insight into an underlying function of mushroom bodies may be provided by cerebellum-like structures in vertebrates, which are similarly defined by the presence of masses of tiny granule cells that emit thin parallel fibers forming a dense molecular layer. In vertebrates, the shared neuroarchitecture of cerebellum-like structures has been suggested to underlie a common functional role as adaptive filters for the removal of predictable sensory elements, such as those arising from reafference, from the total sensory input. Cerebellum-like structures include the vertebrate cerebellum, the electrosensory lateral line lobe, dorsal and medial octavolateral nuclei of fish, and the dorsal cochlear nucleus of mammals. The many architectural and physiological features that the insect mushroom bodies share with cerebellum-like structures suggest that it might be fruitful to consider mushroom body function in light of a possible role as adaptive sensory filters. The present account thus presents a detailed comparison of the insect mushroom bodies with vertebrate cerebellum-like structures.
Collapse
Affiliation(s)
- Sarah M Farris
- Department of Biology, West Virginia University, 3139 Life Sciences Building, 53 Campus Drive, Morgantown, WV 26505, USA.
| |
Collapse
|
14
|
Li YC, Sun LK, Zhou L, Zhang HN. Clarification of the peripherally located F-actin network around the primary afferent neurons. Brain Res 2011; 1392:54-61. [PMID: 21459082 DOI: 10.1016/j.brainres.2011.03.063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Revised: 03/19/2011] [Accepted: 03/25/2011] [Indexed: 10/18/2022]
Abstract
The primary afferent neurons are enriched for F-actin in the peripheries, giving rise to ring-like appearances when stained with fluorescence probes for actin. A variety of data, mainly obtained from in vitro experiments, suggested that the satellite cell (SC) envelopes around sensory neurons may be a candidate for the F-actin positive structures. However, immunohistochemical studies on dorsal root ganglia (DRGs) suggested that F-actin was located exclusively within the cortical regions of the sensory neurons. Given this discrepancy, the present study has reexamined the distribution of F-actin in the DRGs at both the fluorescence light and electron microscopic levels. Double staining of F-actin with S-100 beta and neural cell adhesion molecule showed that the fluorescence signals for F-actin around the DRG neurons were restricted to the inner margins of the SC envelope. Subsequent ultrastructural observations, by use of a modified anti-FITC system, demonstrated that the F-actin-positive sites corresponded mostly to the adneuronal SC lamellae, while the cortical cytoskeletons in the neurons seemed to contribute little to the typical F-actin staining. The high concentration of F-actin in the SC envelope has not yet described before, and is thought to assist in understanding the functions of the SCs in the ganglia.
Collapse
Affiliation(s)
- Yan-Chao Li
- Department of Histology and Embryology, Norman Bethune College of Medicine, Jilin University, Changchun, Jilin Province 130021, China.
| | | | | | | |
Collapse
|
15
|
Köhler CN. Histochemical localization of caldesmon in the CNS and ganglia of the mouse. J Histochem Cytochem 2011; 59:504-17. [PMID: 21411712 DOI: 10.1369/0022155411400875] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The author has recently reported the distribution of the cytoskeleton-associated protein caldesmon in spleen and lymph nodes detected with different antibodies against caldesmon (J Histochem Cytochem 58:183-193, 2010). Here the author reports the distribution of caldesmon in the CNS and ganglia of the mouse using the same antibodies. Western blot analysis of mouse brain and spinal cord showed the preponderance of l-caldesmon and suggested at least two l-caldesmon isoforms in the brain. Immunostaining revealed the predominant reactivity of smooth muscle cells and cells resembling pericytes of many large and small blood vessels, ependymocytes, and secretory cells of the pineal gland and pituitary gland. Neuronal perikarya and neuropil in general displayed no or weak immunoreactivity, but there was stronger labeling of neuronal perikarya in dorsal root and trigeminal ganglia. In the brain, staining of the neuropil was stronger in the molecular layers of the dentate gyrus and cerebellum. Results show that caldesmon is expressed in many different cell types in the CNS and ganglia, consistent with the notion that l-caldesmon is ubiquitously expressed, but it appears most concentrated in smooth muscle cells, pericytes, epithelial cells, secretory cells, and neuronal perikarya in dorsal root and trigeminal ganglia.
Collapse
Affiliation(s)
- Christoph N Köhler
- Institute II of Anatomy, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
16
|
El Ghazi I, Sheng WS, Hu S, Reilly BG, Lokensgard JR, Rock RB, Peterson PK, Wilcox GL, Armitage IM. Changes in the NMR metabolic profile of human microglial cells exposed to lipopolysaccharide or morphine. J Neuroimmune Pharmacol 2010; 5:574-81. [PMID: 20333557 PMCID: PMC4115445 DOI: 10.1007/s11481-010-9197-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2009] [Accepted: 02/14/2010] [Indexed: 02/08/2023]
Abstract
Microglial cells play a major role in host defense of the central nervous system. Once activated, several functional properties are up-regulated including migration, phagocytosis, and secretion of inflammatory mediators such as cytokines and chemokines. Little, if anything, is known about the metabolic changes that occur during the activation process. High-resolution (1)H nuclear magnetic resonance spectra obtained from perchloric acid extracts of human microglial cell cultures exposed to lipopolysaccharide (LPS) or morphine were used to both identify and quantify the metabolites. We found that human microglia exposed to LPS had increased concentrations of glutamate and lactate, whereas the cells exposed to morphine had decreased concentrations in creatinine, taurine, and thymine. Glutamate and creatinine were the key metabolites differentiating between the two stimuli. These results are discussed in terms of activation and differences in the inflammatory response of human microglial cells to LPS and morphine.
Collapse
Affiliation(s)
- Issam El Ghazi
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| | - Wen S. Sheng
- University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Shuxian Hu
- University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Brian G. Reilly
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| | | | - R. Bryan Rock
- University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | - George L. Wilcox
- Departments of Neuroscience, Pharmacology and Dermatology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Ian M. Armitage
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, 6-155 Jackson Hall, 321 Church Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
17
|
Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. J Neurosci 2009; 29:13971-80. [PMID: 19890007 DOI: 10.1523/jneurosci.3799-09.2009] [Citation(s) in RCA: 247] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Leucine-rich repeat kinase 2 (LRRK2) functions as a putative protein kinase of ezrin, radixin, and moesin (ERM) family proteins. A Parkinson's disease-related G2019S substitution in the kinase domain of LRRK2 further enhances the phosphorylation of ERM proteins. The phosphorylated ERM (pERM) proteins are restricted to the filopodia of growing neurites in which they tether filamentous actin (F-actin) to the cytoplasmic membrane and regulate the dynamics of filopodia protrusion. Here, we show that, in cultured neurons derived from LRRK2 G2019S transgenic mice, the number of pERM-positive and F-actin-enriched filopodia was significantly increased, and this correlates with the retardation of neurite outgrowth. Conversely, deletion of LRRK2, which lowered the pERM and F-actin contents in filopodia, promoted neurite outgrowth. Furthermore, inhibition of ERM phosphorylation or actin polymerization rescued the G2019S-dependent neuronal growth defects. These data support a model in which the G2019S mutation of LRRK2 causes a gain-of-function effect that perturbs the homeostasis of pERM and F-actin in sprouting neurites critical for neuronal morphogenesis.
Collapse
|
18
|
Mironov AA, Beznoussenko GV. Correlative microscopy: a potent tool for the study of rare or unique cellular and tissue events. J Microsc 2009; 235:308-21. [PMID: 19754725 DOI: 10.1111/j.1365-2818.2009.03222.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Biological studies have relied on two complementary microscope technologies - light (fluorescence) microscopy and electron microscopy. Light microscopy is used to study phenomena at a global scale to look for unique or rare events, and it also provides an opportunity for live imaging, whereas the forte of electron microscopy is the high resolution. Traditionally light and electron microscopy observations are carried out in different populations of cells/tissues and a 'correlative' inference is drawn. The advent of true correlative light-electron microscopy has allowed high-resolution imaging by electron microscopy of the same structure observed by light microscopy, and in advanced cases by video microscopy. Thus a rare event captured by low-resolution imaging of a population or transient events captured by live imaging can now also be studied at high resolution by electron microscopy. Here, the potential and difficulties of this approach, along with the most impressive breakthroughs obtained by these methods, are discussed.
Collapse
Affiliation(s)
- A A Mironov
- Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, S. Maria Imbaro (Chieti), Italy.
| | | |
Collapse
|
19
|
Stillman AA, Krsnik Z, Sun J, Rasin MR, State MW, Sestan N, Louvi A. Developmentally regulated and evolutionarily conserved expression of SLITRK1 in brain circuits implicated in Tourette syndrome. J Comp Neurol 2009; 513:21-37. [PMID: 19105198 DOI: 10.1002/cne.21919] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tourette syndrome (TS) is an inherited developmental neuropsychiatric disorder characterized by vocal and motor tics. Multiple lines of neurophysiological evidence implicate dysfunction in the corticostriatal-thalamocortical circuits in the etiology of TS. We recently identified rare sequence variants in the Slit and Trk-like family member 1 (SLITRK1) gene associated with TS. SLITRK1, a single-pass transmembrane protein, displays similarities to the SLIT family of secreted ligands, which have roles in axonal repulsion and dendritic patterning, but its function and developmental expression remain largely unknown. Here we provide evidence that SLITRK1 has a developmentally regulated expression pattern in projection neurons of the corticostriatal-thalamocortical circuits. SLITRK1 is further enriched in the somatodendritic compartment and cytoplasmic vesicles of cortical pyramidal neurons in mouse, monkey, and human brain, observations suggestive of an evolutionarily conserved function in mammals. SLITRK1 is transiently expressed in the striosomal/patch compartment of the mammalian striatum and moreover is associated with the direct output pathway; adult striatal expression is confined to cholinergic interneurons. These analyses demonstrate that the expression of SLITRK1 is dynamic and specifically associated with the circuits most commonly implicated in TS and related disorders, suggesting that SLITRK1 contributes to the development of corticostriatal-thalamocortical circuits.
Collapse
Affiliation(s)
- Althea A Stillman
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Potential role of pyridoxal-5'-phosphate phosphatase/chronopin in epilepsy. Exp Neurol 2008; 211:128-40. [PMID: 18346735 DOI: 10.1016/j.expneurol.2008.01.029] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 01/18/2008] [Accepted: 01/18/2008] [Indexed: 11/20/2022]
Abstract
Changes in actin dynamics and pyridoxal-5'-phosphate (PLP) metabolisms are closely related to the pathophysiological profiles of the epileptic hippocampus. Recently, it has been reported that PLP phosphatase/chronophin (PLPP/CIN) directly dephosphorylates actin-depolymerizing factor (ADF)/cofilin as well as PLP. In the present study, therefore, we have investigated whether PLPP/CIN is linked to the dynamics of actin filament assembly and the excitability in the rat hippocampus. In control animals, pyridoxine chloride (PNP) treatment increased PLPP/CIN immunoreactivity only in astrocytes, which did not affect electrophysiological properties. Following status epilepticus, the PLPP/CIN protein level increased in granule cells and reactive astrocytes. These changes in PLPP/CIN protein level showed an inverse correlation with phospho-ADF (pADF)/cofilin levels and F-actin content. These changes were also accompanied by alterations in the excitability ratio and paired-pulse inhibition. Transduction of PLPP/CIN by Tat-PLPP/CIN showed similar effects on pADF/cofilin levels, F-actin content and excitability ratio in normal animals. These findings suggest that PLPP/CIN-mediated actin dynamics may play an important role in the changes of morphological properties and excitability of the epileptic hippocampus.
Collapse
|
21
|
Elkin BS, Azeloglu EU, Costa KD, Morrison B. Mechanical heterogeneity of the rat hippocampus measured by atomic force microscope indentation. J Neurotrauma 2007; 24:812-22. [PMID: 17518536 DOI: 10.1089/neu.2006.0169] [Citation(s) in RCA: 205] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Knowledge of brain tissue mechanical properties may be critical for formulating hypotheses about traumatic brain injury (TBI) mechanisms and for accurate TBI simulations. To determine the local mechanical properties of anatomical subregions within the rat hippocampus, the atomic force microscope (AFM) was adapted for use on living brain tissue. The AFM provided advantages over alternative methods for measuring local mechanical properties of brain because of its high spatial resolution, high sensitivity, and ability to measure live samples under physiologic conditions. From AFM indentations, a mean pointwise or depth-dependent apparent elastic modulus, E, was determined for the following hippocampal subregions: CA1 pyramidal cell layer (CA1P) and stratum radiatum (CA1SR), CA3 pyramidal cell layer (CA3P) and stratum radiatum (CA3SR), and the dentate gyrus (DG). For all regions, E was indentation-depth-dependent, reflecting the nonlinearity of brain tissue. At an indentation depth of 3microm, E was 234 +/- 152 Pa for CA3P, 308 +/- 184 Pa for CA3SR, 137 +/- 97 Pa for CA1P, 169 +/- 52 Pa for CA1SR, and 201 +/- 133 Pa for DG (mean +/- SD). Our results demonstrate for the first time that the hippocampus is mechanically heterogeneous. Based on our findings, we discuss hypotheses accounting for experimentally observed patterns of hippocampal cell death, which can be tested with biofidelic finite element models of TBI.
Collapse
Affiliation(s)
- Benjamin S Elkin
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, USA
| | | | | | | |
Collapse
|
22
|
Sekerková G, Diño MR, Ilijic E, Russo M, Zheng L, Bartles JR, Mugnaini E. Postsynaptic enrichment of Eps8 at dendritic shaft synapses of unipolar brush cells in rat cerebellum. Neuroscience 2007; 145:116-29. [PMID: 17223277 PMCID: PMC1892609 DOI: 10.1016/j.neuroscience.2006.11.061] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2006] [Revised: 11/17/2006] [Accepted: 11/20/2006] [Indexed: 01/07/2023]
Abstract
Epidermal growth factor receptor pathway substrate 8 (Eps8) is a widely expressed multidomain signaling protein that coordinates two disparate GTPase-dependent mechanisms: actin reorganization via Ras/Rac pathways and receptor trafficking via Rab5. Expression of Eps8, the gene encoding the founding member of the Eps8 family of proteins, was found in cerebellum by virtual Northern analysis and in situ hybridization. Because the cerebellum has a well-known cellular architecture and is a favored model to study synaptic plasticity and actin dynamics, we sought to analyze Eps8 localization in rat cerebellar neurons and synapses by light and electron microscopy. Specificity of Eps8-antibody was demonstrated by immunoblots and in brain sections. In cerebellum, unipolar brush cells (UBCs) were densely Eps8 immunopositive and granule cells were moderately immunostained. In both types of neuron immunoreaction product was localized to the somatodendritic and axonal compartments. Postsynaptic immunostained foci were demonstrated in the glomeruli in correspondence of the synapses formed by mossy fiber terminals with granule cell and UBC dendrites. These foci appeared especially evident in the UBC brush, which contains an extraordinary postsynaptic apparatus of actin microfilaments facing synaptic junctions of the long and segmented varieties. Eps8 immunoreactivity was conspicuously absent in Purkinje cells and their actin-rich dendritic spines, in all types of inhibitory interneurons of the cerebellum, cerebellar nuclei neurons, and astrocytes. In conclusion, Eps8 protein in cerebellum is expressed exclusively by excitatory cortical interneurons and is intracellularly compartmentalized in a cell-class specific manner. This is the first demonstration of the presence of a member of the Eps8 protein family in UBCs and its enrichment at postsynaptic sites.
Collapse
Affiliation(s)
- G Sekerková
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, 320 East Superior Street, Chicago, IL 60611, USA.
| | | | | | | | | | | | | |
Collapse
|
23
|
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: 155] [Impact Index Per Article: 9.1] [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
|
24
|
Sosinsky GE, Giepmans BNG, Deerinck TJ, Gaietta GM, Ellisman MH. Markers for correlated light and electron microscopy. Methods Cell Biol 2007; 79:575-91. [PMID: 17327175 DOI: 10.1016/s0091-679x(06)79023-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Gina E Sosinsky
- National Center for Microscopy and Imaging Research and Center for Research in Biological Systems, University of California, San Diego, La Jolla, California 92093, USA
| | | | | | | | | |
Collapse
|
25
|
Offenhäuser N, Castelletti D, Mapelli L, Soppo BE, Regondi MC, Rossi P, D'Angelo E, Frassoni C, Amadeo A, Tocchetti A, Pozzi B, Disanza A, Guarnieri D, Betsholtz C, Scita G, Heberlein U, Di Fiore PP. Increased ethanol resistance and consumption in Eps8 knockout mice correlates with altered actin dynamics. Cell 2006; 127:213-26. [PMID: 17018287 DOI: 10.1016/j.cell.2006.09.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Revised: 07/30/2006] [Accepted: 09/08/2006] [Indexed: 01/18/2023]
Abstract
Dynamic modulation of the actin cytoskeleton is critical for synaptic plasticity, abnormalities of which are thought to contribute to mental illness and addiction. Here we report that mice lacking Eps8, a regulator of actin dynamics, are resistant to some acute intoxicating effects of ethanol and show increased ethanol consumption. In the brain, the N-methyl-D-aspartate (NMDA) receptor is a major target of ethanol. We show that Eps8 is localized to postsynaptic structures and is part of the NMDA receptor complex. Moreover, in Eps8 null mice, NMDA receptor currents and their sensitivity to inhibition by ethanol are abnormal. In addition, Eps8 null neurons are resistant to the actin-remodeling activities of NMDA and ethanol. We propose that proper regulation of the actin cytoskeleton is a key determinant of cellular and behavioral responses to ethanol.
Collapse
Affiliation(s)
- Nina Offenhäuser
- Fondazione Istituto FIRC di Oncologia Molecolare, Via Adamello 16, 20139 Milan, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Nimmerjahn A, Kirchhoff F, Helmchen F. Resting Microglial Cells Are Highly Dynamic Surveillants of Brain Parenchyma in Vivo. Science 2005; 308:1314-8. [PMID: 15831717 DOI: 10.1126/science.1110647] [Citation(s) in RCA: 4065] [Impact Index Per Article: 213.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Microglial cells represent the immune system of the mammalian brain and therefore are critically involved in various injuries and diseases. Little is known about their role in the healthy brain and their immediate reaction to brain damage. By using in vivo two-photon imaging in neocortex, we found that microglial cells are highly active in their presumed resting state, continually surveying their microenvironment with extremely motile processes and protrusions. Furthermore, blood-brain barrier disruption provoked immediate and focal activation of microglia, switching their behavior from patroling to shielding of the injured site. Microglia thus are busy and vigilant housekeepers in the adult brain.
Collapse
Affiliation(s)
- Axel Nimmerjahn
- Abteilung Zellphysiologie, Max Planck Institut für Medizinische Forschung, Jahnstrasse 29, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
27
|
Bobik M, Ellisman MH, Rudy B, Martone ME. Potassium channel subunit Kv3.2 and the water channel aquaporin-4 are selectively localized to cerebellar pinceau. Brain Res 2005; 1026:168-78. [PMID: 15488478 DOI: 10.1016/j.brainres.2004.07.088] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2004] [Indexed: 11/30/2022]
Abstract
The pinceau is a cerebellar structure formed by descending GABA-ergic basket cell axonal terminals converging on the initial axonal segment of Purkinje cell. Although basket cells exert a powerful inhibitory influence on the output of the cerebellar cortex, the function and mode of action of the pinceau are not understood because the majority of basket cell axons fail to make identifiable synaptic contacts with the Purkinje cell axon. Several proteins were previously reported to cluster specifically in this area, including a number of voltage-activated potassium channel subunits. In this study, we used immunohistochemistry, electron microscopy, and electron tomography to examine the ultrastructural localization of a novel voltage-gated potassium channel subunit, Kv3.2, in the pinceau. We found strong, selective localization of Kv3.2 to basket cell axons. Additionally, because potassium buffering is often conducted through water channels, we studied the extent of a brain-specific water channel, aquaporin-4 (AQP4), using confocal and electron microscopy. As expected, we found AQP4 was heavily localized to astrocytic processes of the pinceau. The abundance of potassium channels and AQP4 in this area suggests rapid ionic dynamics in the pinceau, and the unusual, highly specialized morphology of this region implies that the structural features may combine with the molecular composition to regulate the microenvironment of the initial segment of the Purkinje cell axon.
Collapse
Affiliation(s)
- Marketta Bobik
- Department of Neurosciences, National Center for Microscopy and Imaging Research, University of California, San Diego, La Jolla, CA 92093-0608, USA
| | | | | | | |
Collapse
|
28
|
Nikonenko I, Boda B, Alberi S, Muller D. Application of photoconversion technique for correlated confocal and ultrastructural studies in organotypic slice cultures. Microsc Res Tech 2005; 68:90-6. [PMID: 16228980 DOI: 10.1002/jemt.20239] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Photoconversion of fluorescent staining into stable diaminobenzidine (DAB) precipitate is widely used for neuroanatomical and developmental studies. An important advantage of the approach is to make correlations between light and electron microscopy analyses possible, the DAB reaction product formed during photoconversion being electron dense. By combining a photoconversion approach with biolistic transfection of neurons in organotypic hippocampal slice cultures, we describe here a methodology that allowed us to study at the electron microscopy level the fine details of cells expressing specific genes of interest. The same approach has also been used to analyze the ultrastructural characteristics of specific cells such as neurons recorded with patch clamp techniques. This approach revealed particularly useful for studies of dendritic arborisation, dendritic spines, and axon varicosities of identified cells, as precise morphometric parameters of these structures can only be obtained by electron microscopy. The techniques used for fluorescent staining and photoconversion of these different cell structures and the results obtained by electron microscopic analyses are described.
Collapse
Affiliation(s)
- Irina Nikonenko
- Département des Neurosciences Fondamentales, Centre Médical Universitaire, Geneva, Switzerland.
| | | | | | | |
Collapse
|