101
|
Miwa JM, Lester HA, Walz A. Optimizing cholinergic tone through lynx modulators of nicotinic receptors: implications for plasticity and nicotine addiction. Physiology (Bethesda) 2012; 27:187-99. [PMID: 22875450 DOI: 10.1152/physiol.00002.2012] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The cholinergic system underlies both adaptive (learning and memory) and nonadaptive (addiction and dependency) behavioral changes through its ability to shape and regulate plasticity. Protein modulators such as lynx family members can fine tune the activity of the cholinergic system and contribute to the graded response of the cholinergic system, stabilizing neural circuitry through direct interaction with nicotinic receptors. Release of this molecular brake can unmask cholinergic-dependent mechanisms in the brain. Lynx proteins have the potential to provide top-down control over plasticity mechanisms, including addictive propensity. If this is indeed the case, then, what regulates the regulator? Transcriptional changes of lynx genes in response to pharmacological, physiological, and pathological alterations are explored in this review.
Collapse
Affiliation(s)
- Julie M Miwa
- California Institute of Technology, Pasadena, California, USA.
| | | | | |
Collapse
|
102
|
Herrera-Rincon C, Torets C, Sanchez-Jimenez A, Avendaño C, Panetsos F. Chronic electrical stimulation of transected peripheral nerves preserves anatomy and function in the primary somatosensory cortex. Eur J Neurosci 2012; 36:3679-90. [DOI: 10.1111/ejn.12000] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Revised: 08/01/2012] [Accepted: 08/13/2012] [Indexed: 01/18/2023]
Affiliation(s)
- Celia Herrera-Rincon
- Neurocomputing and Neurorobotics Research Group; Universidad Complutense de Madrid; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Madrid; Spain
| | - Carlos Torets
- Neurocomputing and Neurorobotics Research Group; Universidad Complutense de Madrid; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Madrid; Spain
| | | | - Carlos Avendaño
- Department of Anatomy, Histology and Neuroscience; Universidad Autonoma de Madrid; Madrid; Spain
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group; Universidad Complutense de Madrid; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC); Madrid; Spain
| |
Collapse
|
103
|
IGF1 as a Potential Treatment for Rett Syndrome: Safety Assessment in Six Rett Patients. AUTISM RESEARCH AND TREATMENT 2012; 2012:679801. [PMID: 22934177 PMCID: PMC3420537 DOI: 10.1155/2012/679801] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 04/03/2012] [Indexed: 11/18/2022]
Abstract
Rett syndrome (RTT) is a devastating neurodevelopmental disorder that affects one in ten thousand girls and has no cure. The majority of RTT patients display mutations in the gene that codes for the methyl-CpG-binding protein 2 (MeCP2). Clinical observations and neurobiological analysis of mouse models suggest that defects in the expression of MeCP2 protein compromise the development of the central nervous system, especially synaptic and circuit maturation. Thus, agents that promote brain development and synaptic function, such as insulin-like growth factor 1 (IGF1), are good candidates for ameliorating the symptoms of RTT. IGF1 and its active peptide, (1–3) IGF1, cross the blood brain barrier, and (1–3) IGF1 ameliorates the symptoms of RTT in a mouse model of the disease; therefore they are ideal treatments for neurodevelopmental disorders, including RTT. We performed a pilot study to establish whether there are major risks associated with IGF1 administration in RTT patients. Six young girls with classic RTT received IGF1 subcutaneous injections twice a day for six months, and they were regularly monitored by their primary care physicians and by the unit for RTT in Versilia Hospital (Italy). This study shows that there are no risks associated with IGF1 administration.
Collapse
|
104
|
Abstract
There are numerous examples of enduring effects of early experience on gene transcription and neural function. We review the emerging evidence for epigenetics as a candidate mechanism for such effects. There is now evidence that intracellular signals activated by environmental events can directly modify the epigenetic state of the genome, including CpG methylation, histone modifications and microRNAs. We suggest that this process reflects an activity-dependent epigenetic plasticity at the level of the genome, comparable with that observed at the synapse. This epigenetic plasticity mediates neuronal differentiation and phenotypic plasticity, including that associated with learning and memory. Altered epigenetic states are also associated with the risk for and expression of mental disorders. In a broader context, these studies define a biological basis for the interplay between environmental signals and the genome in the regulation of individual differences in behavior, cognition and physiology, as well as the risk for psychopathology.
Collapse
Affiliation(s)
- Judy Sng
- Integrative Neuroscience Program, Singapore Institute for Clinical Sciences, 30 Medial Drive, Singapore.
| | | |
Collapse
|
105
|
Cortical GABAergic interneurons in cross-modal plasticity following early blindness. Neural Plast 2012; 2012:590725. [PMID: 22720175 PMCID: PMC3377178 DOI: 10.1155/2012/590725] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 04/04/2012] [Indexed: 11/30/2022] Open
Abstract
Early loss of a given sensory input in mammals causes anatomical and functional modifications in the brain via a process called cross-modal plasticity. In the past four decades, several animal models have illuminated our understanding of the biological substrates involved in cross-modal plasticity. Progressively, studies are now starting to emphasise on cell-specific mechanisms that may be responsible for this intermodal sensory plasticity. Inhibitory interneurons expressing γ-aminobutyric acid (GABA) play an important role in maintaining the appropriate dynamic range of cortical excitation, in critical periods of developmental plasticity, in receptive field refinement, and in treatment of sensory information reaching the cerebral cortex. The diverse interneuron population is very sensitive to sensory experience during development. GABAergic neurons are therefore well suited to act as a gate for mediating cross-modal plasticity. This paper attempts to highlight the links between early sensory deprivation, cortical GABAergic interneuron alterations, and cross-modal plasticity, discuss its implications, and further provide insights for future research in the field.
Collapse
|
106
|
IGF-1 restores visual cortex plasticity in adult life by reducing local GABA levels. Neural Plast 2012; 2012:250421. [PMID: 22720172 PMCID: PMC3375096 DOI: 10.1155/2012/250421] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Accepted: 04/01/2012] [Indexed: 01/10/2023] Open
Abstract
The central nervous system architecture is markedly modified by sensory experience during early life, but a decline of plasticity occurs with age. Recent studies have challenged this dogma providing evidence that both pharmacological treatments and paradigms based on the manipulation of environmental stimulation levels can be successfully employed as strategies for enhancing plasticity in the adult nervous system. Insulin-like growth factor 1 (IGF-1) is a peptide implicated in prenatal and postnatal phases of brain development such as neurogenesis, neuronal differentiation, synaptogenesis, and experience-dependent plasticity. Here, using the visual system as a paradigmatic model, we report that IGF-1 reactivates neural plasticity in the adult brain. Exogenous administration of IGF-1 in the adult visual cortex, indeed, restores the susceptibility of cortical neurons to monocular deprivation and promotes the recovery of normal visual functions in adult amblyopic animals. These effects were accompanied by a marked reduction of intracortical GABA levels. Moreover, we show that a transitory increase of IGF-1 expression is associated to the plasticity reinstatement induced by environmental enrichment (EE) and that blocking IGF-1 action by means of the IGF-1 receptor antagonist JB1 prevents EE effects on plasticity processes.
Collapse
|
107
|
Corvin AP, Molinos I, Little G, Donohoe G, Gill M, Morris DW, Tropea D. Insulin-like growth factor 1 (IGF1) and its active peptide (1–3)IGF1 enhance the expression of synaptic markers in neuronal circuits through different cellular mechanisms. Neurosci Lett 2012; 520:51-6. [DOI: 10.1016/j.neulet.2012.05.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 05/02/2012] [Accepted: 05/08/2012] [Indexed: 10/28/2022]
|
108
|
Sommeijer JP, Levelt CN. Synaptotagmin-2 is a reliable marker for parvalbumin positive inhibitory boutons in the mouse visual cortex. PLoS One 2012; 7:e35323. [PMID: 22539967 PMCID: PMC3335159 DOI: 10.1371/journal.pone.0035323] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Accepted: 03/15/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Inhibitory innervation by parvalbumin (PV) expressing interneurons has been implicated in the onset of the sensitive period of visual plasticity. Immunohistochemical analysis of the development and plasticity of these inhibitory inputs is difficult because PV expression is low in young animals and strongly influenced by neuronal activity. Moreover, the synaptic boutons that PV neurons form onto each other cannot be distinguished from the innervated cell bodies by immunostaining for this protein because it is present throughout the cells. These problems call for the availability of a synaptic, activity-independent marker for PV+ inhibitory boutons that is expressed before sensitive period onset. We investigated whether synaptotagmin-2 (Syt2) fulfills these properties in the visual cortex. Syt2 is a synaptic vesicle protein involved in fast Ca(2+) dependent neurotransmitter release. Its mRNA expression follows a pattern similar to that of PV throughout the brain and is present in 30-40% of hippocampal PV expressing basket cells. Up to now, no quantitative analyses of Syt2 expression in the visual cortex have been carried out. METHODOLOGY/PRINCIPAL FINDINGS We used immunohistochemistry to analyze colocalization of Syt2 with multiple interneuron markers including vesicular GABA transporter VGAT, calbindin, calretinin, somatostatin and PV in the primary visual cortex of mice during development and after dark-rearing. CONCLUSIONS/SIGNIFICANCE We show that in the adult visual cortex Syt2 is only found in inhibitory, VGAT positive boutons. Practically all Syt2 positive boutons also contain PV and vice versa. During development, Syt2 expression can be detected in synaptic boutons prior to PV and in contrast to PV expression, Syt2 is not down-regulated by dark-rearing. These properties of Syt2 make it an excellent marker for analyzing the development and plasticity of perisomatic inhibitory innervations onto both excitatory and inhibitory neurons in the visual cortex.
Collapse
Affiliation(s)
- Jean-Pierre Sommeijer
- Department of Molecular Visual Neuroscience, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - Christiaan N. Levelt
- Department of Molecular Visual Neuroscience, Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
109
|
Kind PC, Sengpiel F, Beaver CJ, Crocker-Buque A, Kelly GM, Matthews RT, Mitchell DE. The development and activity-dependent expression of aggrecan in the cat visual cortex. ACTA ACUST UNITED AC 2012; 23:349-60. [PMID: 22368089 DOI: 10.1093/cercor/bhs015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The Cat-301 monoclonal antibody identifies aggrecan, a chondroitin sulfate proteoglycan in the cat visual cortex and dorsal lateral geniculate nucleus (dLGN). During development, aggrecan expression increases in the dLGN with a time course that matches the decline in plasticity. Moreover, examination of tissue from selectively visually deprived cats shows that expression is activity dependent, suggesting a role for aggrecan in the termination of the sensitive period. Here, we demonstrate for the first time that the onset of aggrecan expression in area 17 also correlates with the decline in experience-dependent plasticity in visual cortex and that this expression is experience dependent. Dark rearing until 15 weeks of age dramatically reduced the density of aggrecan-positive neurons in the extragranular layers, but not in layer IV. This effect was reversible as dark-reared animals that were subsequently exposed to light showed normal numbers of Cat-301-positive cells. The reduction in aggrecan following certain early deprivation regimens is the first biochemical correlate of the functional changes to the γ-aminobutyric acidergic system that have been reported following early deprivation in cats.
Collapse
Affiliation(s)
- P C Kind
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh EH8 9XD, UK
| | | | | | | | | | | | | |
Collapse
|
110
|
Rietman ML, Sommeijer JP, Levelt CN, Heimel JA. Candidate genes in ocular dominance plasticity. Front Neurosci 2012; 6:11. [PMID: 22347157 PMCID: PMC3269753 DOI: 10.3389/fnins.2012.00011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 01/16/2012] [Indexed: 11/16/2022] Open
Abstract
Many studies have been devoted to the identification of genes involved in experience-dependent plasticity in the visual cortex. To discover new candidate genes, we have reexamined data from one such study on ocular dominance (OD) plasticity in recombinant inbred BXD mouse strains. We have correlated the level of plasticity with the gene expression data in the neocortex that have become available for these same strains. We propose that genes with a high correlation are likely to play a role in OD plasticity. We have tested this hypothesis for genes whose inactivation is known to affect OD plasticity. The expression levels of these genes indeed correlated with OD plasticity if their levels showed strong differences between the BXD strains. To narrow down our candidate list of correlated genes, we have selected only those genes that were previously found to be regulated by visual experience and associated with pathways implicated in OD plasticity. This resulted in a list of 32 candidate genes. The list contained unproven, but not unexpected candidates such as the genes for IGF-1, NCAM1, NOGO-A, the gamma2 subunit of the GABA(A) receptor, acetylcholine esterase, and the catalytic subunit of cAMP-dependent protein kinase A. This demonstrates the viability of our approach. More interestingly, the following novel candidate genes were identified: Akap7, Akt1, Camk2d, Cckbr, Cd44, Crim1, Ctdsp2, Dnajc5, Gnai1, Itpka, Mapk8, Nbea, Nfatc3, Nlk, Npy5r, Phf21a, Phip, Ppm1l, Ppp1r1b, Rbbp4, Slc1a3, Slit2, Socs2, Spock3, St8sia1, Zfp207. Whether all these novel candidates indeed function in OD plasticity remains to be established, but possible roles of some of them are discussed in the article.
Collapse
Affiliation(s)
- M Liset Rietman
- Department of Molecular Visual Plasticity, Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Arts and Sciences Amsterdam, Netherlands
| | | | | | | | | |
Collapse
|
111
|
Miyata S, Komatsu Y, Yoshimura Y, Taya C, Kitagawa H. Persistent cortical plasticity by upregulation of chondroitin 6-sulfation. Nat Neurosci 2012; 15:414-22, S1-2. [PMID: 22246436 DOI: 10.1038/nn.3023] [Citation(s) in RCA: 255] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 12/06/2011] [Indexed: 11/09/2022]
Abstract
Cortical plasticity is most evident during a critical period in early life, but the mechanisms that restrict plasticity after the critical period are poorly understood. We found that a developmental increase in the 4-sulfation/6-sulfation (4S/6S) ratio of chondroitin sulfate proteoglycans (CSPGs), which are components of the brain extracellular matrix, leads to the termination of the critical period for ocular dominance plasticity in the mouse visual cortex. Condensation of CSPGs into perineuronal nets that enwrapped synaptic contacts on parvalbumin-expressing interneurons was prevented by cell-autonomous overexpression of chondroitin 6-sulfation, which maintains a low 4S/6S ratio. Furthermore, the increase in the 4S/6S ratio was required for the accumulation of Otx2, a homeoprotein that activates the development of parvalbumin-expressing interneurons, and for functional maturation of the electrophysiological properties of these cells. Our results indicate that the critical period for cortical plasticity is regulated by the 4S/6S ratio of CSPGs, which determines the maturation of parvalbumin-expressing interneurons.
Collapse
Affiliation(s)
- Shinji Miyata
- Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe, Japan
| | | | | | | | | |
Collapse
|
112
|
Modulation of CREB in the dorsal lateral geniculate nucleus of dark-reared mice. Neural Plast 2012; 2012:426437. [PMID: 22292123 PMCID: PMC3265102 DOI: 10.1155/2012/426437] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 10/04/2011] [Indexed: 11/18/2022] Open
Abstract
The cAMP-response element-binding protein (CREB) plays an important role in visual cortical plasticity that follows the disruption of sensory activity, as induced by dark rearing (DR). Recent findings indicate that the dorsal lateral geniculate nucleus (dLGN) of thalamus is also sensitive to altered sensory activity. DR disrupts retinogeniculate synaptic strength and pruning in mice, but only when DR starts one week after eye opening (delayed DR, DDR) and not after chronic DR (CDR) from birth. While DR upregulates CREB in visual cortex, whether it also modulates this pathway in dLGN remains unknown. Here we investigate the role of CREB in the dLGN of mice that were CDR or DDR using western blot and immunofluorescence. Similar to findings in visual cortex, CREB is upregulated in dLGN after CDR and DDR. These findings are consistent with the proposal that DR up-regulates the CREB pathway in response to decreased visual drive.
Collapse
|
113
|
Mower AF, Kwok S, Yu H, Majewska AK, Okamoto KI, Hayashi Y, Sur M. Experience-dependent regulation of CaMKII activity within single visual cortex synapses in vivo. Proc Natl Acad Sci U S A 2011; 108:21241-6. [PMID: 22160721 PMCID: PMC3248554 DOI: 10.1073/pnas.1108261109] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Unbalanced visual input during development induces persistent alterations in the function and structure of visual cortical neurons. The molecular mechanisms that drive activity-dependent changes await direct visualization of underlying signals at individual synapses in vivo. By using a genetically engineered Förster resonance energy transfer (FRET) probe for the detection of CaMKII activity, and two-photon imaging of single synapses within identified functional domains, we have revealed unexpected and differential mechanisms in specific subsets of synapses in vivo. Brief monocular deprivation leads to activation of CaMKII in most synapses of layer 2/3 pyramidal cells within deprived eye domains, despite reduced visual drive, but not in nondeprived eye domains. Synapses that are eliminated in deprived eye domains have low basal CaMKII activity, implying a protective role for activated CaMKII against synapse elimination.
Collapse
Affiliation(s)
- Amanda F. Mower
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Showming Kwok
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
- RIKEN–MIT Neuroscience Research Center, Massachusetts Institute of Technology, Cambridge, MA 02139; and
| | - Hongbo Yu
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ania K. Majewska
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ken-Ichi Okamoto
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
- RIKEN–MIT Neuroscience Research Center, Massachusetts Institute of Technology, Cambridge, MA 02139; and
| | - Yasunori Hayashi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
- RIKEN–MIT Neuroscience Research Center, Massachusetts Institute of Technology, Cambridge, MA 02139; and
- Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | - Mriganka Sur
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
114
|
Lee H, Sawatari A. Medium spiny neurons of the neostriatal matrix exhibit specific, stereotyped changes in dendritic arborization during a critical developmental period in mice. Eur J Neurosci 2011; 34:1345-54. [PMID: 21995728 DOI: 10.1111/j.1460-9568.2011.07852.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In mice, the matrix compartment of the striatum (caudate/putamen) undergoes major developmental changes during the second postnatal week, including the establishment of corticostriatal and nigrostriatal afferents, the maturation of parvalbumin-positive interneurons and the appearance of perineuronal nets. It is not known if any of these events influence the dendritic structure of medium spiny neurons, the principal output cells of the striatum. To determine whether any measurable changes in the dendrites of matrix medium spiny neurons occur during this important developmental period, we labeled individual cells at different time points flanking the second postnatal week. These cells exhibit distinct dendritic morphologies from the earliest postnatal time points examined. Furthermore, our data show that the dendritic arbors of these neurons change in length, branch points, diameter and tortuosity, regardless of morphological type. The increase in dendritic length is accompanied by a decrease in the number of branch points that occur in different, but consistent, parts of the dendritic arbor. All of these changes are most pronounced during the second postnatal week, coinciding with a number of developmental events considered important for consolidating circuitry within the striatal matrix. Our results further support the critical importance of this early postnatal period in striatal development.
Collapse
Affiliation(s)
- Hyunchul Lee
- Discipline of Physiology, School of Medical Sciences and the Bosch Institute, University of Sydney, Sydney, NSW, Australia
| | | |
Collapse
|
115
|
Landi S, Putignano E, Boggio EM, Giustetto M, Pizzorusso T, Ratto GM. The short-time structural plasticity of dendritic spines is altered in a model of Rett syndrome. Sci Rep 2011; 1:45. [PMID: 22355564 PMCID: PMC3216532 DOI: 10.1038/srep00045] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 07/04/2011] [Indexed: 11/09/2022] Open
Abstract
The maturation of excitatory transmission comes about through a developmental period in which dendritic spines are highly motile and their number, form and size are rapidly changing. Surprisingly, although these processes are crucial for the formation of cortical circuitry, little is known about possible alterations of these processes in brain disease. By means of acute in vivo 2-photon imaging we show that the dynamic properties of dendritic spines of layer V cortical neurons are deeply affected in a mouse model of Rett syndrome (RTT) at a time around P25 when the neuronal phenotype of the disease is still mild. Then, we show that 24h after a subcutaneous injection of IGF-1 spine dynamics is restored. Our study demonstrates that spine dynamics in RTT mice is severely impaired early during development and suggest that treatments for RTT should be started very early in order to reestablish a normal period of spine plasticity.
Collapse
Affiliation(s)
- Silvia Landi
- NEST, Scuola Normale Superiore, Pisa, Italy; NEST Institute Nanoscience CNR, Pisa, Italy
| | | | | | | | | | | |
Collapse
|
116
|
Funke K, Benali A. Modulation of cortical inhibition by rTMS - findings obtained from animal models. J Physiol 2011; 589:4423-35. [PMID: 21768267 DOI: 10.1113/jphysiol.2011.206573] [Citation(s) in RCA: 125] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Transcranial magnetic stimulation (TMS) has become a popular method to non-invasively stimulate the human brain. The opportunity to modify cortical excitability with repetitive stimulation (rTMS) has especially gained interest for its therapeutic potential. However, details of the cellular mechanisms of the effects of rTMS are scarce. Currently favoured are long-term changes in the efficiency of excitatory synaptic transmission, with low-frequency rTMS depressing it, but high-frequency rTMS augmenting. Only recently has modulation of cortical inhibition been considered as an alternative way to explain lasting changes in cortical excitability induced by rTMS. Adequate animal models help to highlight stimulation-induced changes in cellular processes which are not assessable in human rTMS studies. In this review article, we summarize findings obtained with our rat models which indicate that distinct inhibitory cell classes, like the fast-spiking cells characterized by parvalbumin expression, are most sensitive to certain stimulation protocols, e.g. intermittent theta burst stimulation. We discuss how our findings can support the recently suggested models of gating and homeostatic plasticity as possible mechanisms of rTMS-induced changes in cortical excitability.
Collapse
Affiliation(s)
- Klaus Funke
- Institute of Physiology, Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, 44780 Bochum, Germany.
| | | |
Collapse
|
117
|
West AE, Greenberg ME. Neuronal activity-regulated gene transcription in synapse development and cognitive function. Cold Spring Harb Perspect Biol 2011; 3:cshperspect.a005744. [PMID: 21555405 DOI: 10.1101/cshperspect.a005744] [Citation(s) in RCA: 390] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Activity-dependent plasticity of vertebrate neurons allows the brain to respond to its environment. During brain development, both spontaneous and sensory-driven neural activity are essential for instructively guiding the process of synapse development. These effects of neuronal activity are transduced in part through the concerted regulation of a set of activity-dependent transcription factors that coordinate a program of gene expression required for the formation and maturation of synapses. Here we review the cellular signaling networks that regulate the activity of transcription factors during brain development and discuss the functional roles of specific activity-regulated transcription factors in specific stages of synapse formation, refinement, and maturation. Interestingly, a number of neurodevelopmental disorders have been linked to abnormalities in activity-regulated transcriptional pathways, indicating that these signaling networks are critical for cognitive development and function.
Collapse
Affiliation(s)
- Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
118
|
Jimenez S, Torres M, Vizuete M, Sanchez-Varo R, Sanchez-Mejias E, Trujillo-Estrada L, Carmona-Cuenca I, Caballero C, Ruano D, Gutierrez A, Vitorica J. Age-dependent accumulation of soluble amyloid beta (Abeta) oligomers reverses the neuroprotective effect of soluble amyloid precursor protein-alpha (sAPP(alpha)) by modulating phosphatidylinositol 3-kinase (PI3K)/Akt-GSK-3beta pathway in Alzheimer mouse model. J Biol Chem 2011; 286:18414-25. [PMID: 21460223 PMCID: PMC3099658 DOI: 10.1074/jbc.m110.209718] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2010] [Revised: 03/04/2011] [Indexed: 01/04/2023] Open
Abstract
Neurotrophins, activating the PI3K/Akt signaling pathway, control neuronal survival and plasticity. Alterations in NGF, BDNF, IGF-1, or insulin signaling are implicated in the pathogenesis of Alzheimer disease. We have previously characterized a bigenic PS1×APP transgenic mouse displaying early hippocampal Aβ deposition (3 to 4 months) but late (17 to 18 months) neurodegeneration of pyramidal cells, paralleled to the accumulation of soluble Aβ oligomers. We hypothesized that PI3K/Akt/GSK-3β signaling pathway could be involved in this apparent age-dependent neuroprotective/neurodegenerative status. In fact, our data demonstrated that, as compared with age-matched nontransgenic controls, the Ser-9 phosphorylation of GSK-3β was increased in the 6-month PS1×APP hippocampus, whereas in aged PS1×APP animals (18 months), GSK-3β phosphorylation levels displayed a marked decrease. Using N2a and primary neuronal cell cultures, we demonstrated that soluble amyloid precursor protein-α (sAPPα), the predominant APP-derived fragment in young PS1×APP mice, acting through IGF-1 and/or insulin receptors, activated the PI3K/Akt pathway, phosphorylated the GSK-3β activity, and in consequence, exerted a neuroprotective action. On the contrary, several oligomeric Aβ forms, present in the soluble fractions of aged PS1×APP mice, inhibited the induced phosphorylation of Akt/GSK-3β and decreased the neuronal survival. Furthermore, synthetic Aβ oligomers blocked the effect mediated by different neurotrophins (NGF, BDNF, insulin, and IGF-1) and sAPPα, displaying high selectivity for NGF. In conclusion, the age-dependent appearance of APP-derived soluble factors modulated the PI3K/Akt/GSK-3β signaling pathway through the major neurotrophin receptors. sAPPα stimulated and Aβ oligomers blocked the prosurvival signaling. Our data might provide insights into the selective vulnerability of specific neuronal groups in Alzheimer disease.
Collapse
Affiliation(s)
- Sebastian Jimenez
- From the Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
- the Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Manuel Torres
- From the Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
- the Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Marisa Vizuete
- From the Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
- the Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Raquel Sanchez-Varo
- the Departamento Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
| | - Elisabeth Sanchez-Mejias
- the Departamento Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
| | - Laura Trujillo-Estrada
- the Departamento Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
| | - Irene Carmona-Cuenca
- From the Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
- the Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Cristina Caballero
- From the Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
- the Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Diego Ruano
- From the Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
- the Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| | - Antonia Gutierrez
- the Departamento Biología Celular, Genética y Fisiología, Facultad de Ciencias, Universidad de Málaga, 29071 Málaga
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
| | - Javier Vitorica
- From the Departamento Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla
- the Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas, 41013 Sevilla, and
- the Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, 41013 Sevilla, Spain
| |
Collapse
|
119
|
Vallès A, Boender AJ, Gijsbers S, Haast RAM, Martens GJM, de Weerd P. Genomewide analysis of rat barrel cortex reveals time- and layer-specific mRNA expression changes related to experience-dependent plasticity. J Neurosci 2011; 31:6140-58. [PMID: 21508239 PMCID: PMC6632955 DOI: 10.1523/jneurosci.6514-10.2011] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 01/26/2011] [Accepted: 02/26/2011] [Indexed: 12/12/2022] Open
Abstract
Because of its anatomical organization, the rodent whisker-to-barrel system is an ideal model to study experience-dependent plasticity. Manipulation of sensory input causes changes in the properties of the barrels at the physiological, structural, and functional levels. However, much less is known about the molecular events underlying these changes. To explore such molecular events, we have used a genomewide approach to identify key genes and molecular pathways involved in experience-induced plasticity in the barrel cortex of adult rats. Given the natural tendency of rats to explore novel objects, exposure to an enriched environment (EE) was used to stimulate the activity of the whisker-to-barrel cortex in vivo. Microarray analysis at two different time points after EE revealed differential expression of genes encoding transcription factors, including nuclear receptors, as well as of genes involved in the regulation of synaptic plasticity, cell differentiation, metabolism, and, surprisingly, blood vessel morphogenesis. These expression differences reflect changes in somatosensory information processing because unilateral whisker clipping showed EE-induced differential expression patterns in the spared versus deprived barrel cortex. Finally, in situ hybridization revealed cortical layer patterns specific for each selected gene. Together, the present study offers the first genomewide exploration of the key genes regulated by somatosensory stimulation in the barrel cortex and thus provides a solid experimental framework for future in-depth analysis of the mechanisms underlying experience-dependent plasticity.
Collapse
Affiliation(s)
- Astrid Vallès
- Department of Neurocognition, Faculty of Psychology and Neurosciences, Maastricht University, 6200 MD Maastricht, The Netherlands, and
- Department of Molecular Animal Physiology, Radboud University Nijmegen, Donders Institute for Brain, Cognition, and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Arjen J. Boender
- Department of Molecular Animal Physiology, Radboud University Nijmegen, Donders Institute for Brain, Cognition, and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Steef Gijsbers
- Department of Molecular Animal Physiology, Radboud University Nijmegen, Donders Institute for Brain, Cognition, and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Roy A. M. Haast
- Department of Molecular Animal Physiology, Radboud University Nijmegen, Donders Institute for Brain, Cognition, and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Gerard J. M. Martens
- Department of Molecular Animal Physiology, Radboud University Nijmegen, Donders Institute for Brain, Cognition, and Behaviour (Centre for Neuroscience), Nijmegen Centre for Molecular Life Sciences, 6525 GA Nijmegen, The Netherlands
| | - Peter de Weerd
- Department of Neurocognition, Faculty of Psychology and Neurosciences, Maastricht University, 6200 MD Maastricht, The Netherlands, and
| |
Collapse
|
120
|
Dahlhaus M, Li KW, van der Schors RC, Saiepour MH, van Nierop P, Heimel JA, Hermans JM, Loos M, Smit AB, Levelt CN. The synaptic proteome during development and plasticity of the mouse visual cortex. Mol Cell Proteomics 2011; 10:M110.005413. [PMID: 21398567 PMCID: PMC3098591 DOI: 10.1074/mcp.m110.005413] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
During brain development, the neocortex shows periods of enhanced plasticity, which enables the acquisition of knowledge and skills that we use and build on in adult life. Key to persistent modifications of neuronal connectivity and plasticity of the neocortex are molecular changes occurring at the synapse. Here we used isobaric tag for relative and absolute quantification to measure levels of 467 synaptic proteins in a well-established model of plasticity in the mouse visual cortex and the regulation of its critical period. We found that inducing visual cortex plasticity by monocular deprivation during the critical period increased levels of kinases and proteins regulating the actin-cytoskeleton and endocytosis. Upon closure of the critical period with age, proteins associated with transmitter vesicle release and the tubulin- and septin-cytoskeletons increased, whereas actin-regulators decreased in line with augmented synapse stability and efficacy. Maintaining the visual cortex in a plastic state by dark rearing mice into adulthood only partially prevented these changes and increased levels of G-proteins and protein kinase A subunits. This suggests that in contrast to the general belief, dark rearing does not simply delay cortical development but may activate signaling pathways that specifically maintain or increase the plasticity potential of the visual cortex. Altogether, this study identified many novel candidate plasticity proteins and signaling pathways that mediate synaptic plasticity during critical developmental periods or restrict it in adulthood.
Collapse
Affiliation(s)
- Martijn Dahlhaus
- Netherlands Institute for Neuroscience, an institute of the Royal Netherlands Academy of Arts and Sciences, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Upregulation of insulin-like growth factor binding protein 3 in astrocytes of transgenic mice that express Borna disease virus phosphoprotein. J Virol 2011; 85:4567-71. [PMID: 21325425 DOI: 10.1128/jvi.01817-10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
In a previous study, we demonstrated that transgenic mice that express Borna disease virus (BDV) phosphoprotein (P) in astrocytes show striking neurobehavioral abnormalities resembling those in BDV-infected animals. To understand the molecular disturbances induced by the expression of P in astrocytes, we performed microarray analysis with cultured astroglial cells transiently expressing P. We showed that expression of insulin-like growth factor binding protein 3 mRNA increases not only in P-expressing cultured cells but also in astrocytes from the cerebella of P transgenic mice (P-Tg). Furthermore, we demonstrated that insulin-like growth factor signaling is disturbed in the P-Tg cerebellum, a factor that might be involved in the increased vulnerability of Purkinje cell neurons in the brain.
Collapse
|
122
|
Benali A, Trippe J, Weiler E, Mix A, Petrasch-Parwez E, Girzalsky W, Eysel UT, Erdmann R, Funke K. Theta-burst transcranial magnetic stimulation alters cortical inhibition. J Neurosci 2011; 31:1193-203. [PMID: 21273404 PMCID: PMC6623597 DOI: 10.1523/jneurosci.1379-10.2011] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 09/24/2010] [Accepted: 11/16/2010] [Indexed: 11/21/2022] Open
Abstract
Human cortical excitability can be modified by repetitive transcranial magnetic stimulation (rTMS), but the cellular mechanisms are largely unknown. Here, we show that the pattern of delivery of theta-burst stimulation (TBS) (continuous versus intermittent) differently modifies electric activity and protein expression in the rat neocortex. Intermittent TBS (iTBS), but not continuous TBS (cTBS), enhanced spontaneous neuronal firing and EEG gamma band power. Sensory evoked cortical inhibition increased only after iTBS, although both TBS protocols increased the first sensory response arising from the resting cortical state. Changes in the cortical expression of the calcium-binding proteins parvalbumin (PV) and calbindin D-28k (CB) indicate that changes in spontaneous and evoked cortical activity following rTMS are in part related to altered activity of inhibitory systems. By reducing PV expression in the fast-spiking interneurons, iTBS primarily affected the inhibitory control of pyramidal cell output activity, while cTBS, by reducing CB expression, more likely affected the dendritic integration of synaptic inputs controlled by other classes of inhibitory interneurons. Calretinin, the third major calcium-binding protein expressed by another class of interneurons was not affected at all. We conclude that different patterns of TBS modulate the activity of inhibitory cell classes differently, probably depending on the synaptic connectivity and the preferred discharge pattern of these inhibitory neurons.
Collapse
Affiliation(s)
- Alia Benali
- Institute of Physiology, Department of Neurophysiology
- Department of Cognitive Neurology, Hertie-Institute for Clinical Brain Research and Center for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany
| | - Jörn Trippe
- Institute of Physiology, Department of Neurophysiology
| | - Elke Weiler
- Institute of Physiology, Department of Neurophysiology
| | - Annika Mix
- Institute of Physiology, Department of Neurophysiology
| | | | - Wolfgang Girzalsky
- Institute of Physiological Chemistry, Department of Systems Biochemistry, Ruhr-University Bochum, 44780 Bochum, Germany, and
| | - Ulf T. Eysel
- Institute of Physiology, Department of Neurophysiology
| | - Ralf Erdmann
- Institute of Physiological Chemistry, Department of Systems Biochemistry, Ruhr-University Bochum, 44780 Bochum, Germany, and
| | - Klaus Funke
- Institute of Physiology, Department of Neurophysiology
| |
Collapse
|
123
|
Abstract
The work of recent decades has shown that the nervous system changes continually throughout life. Activity-dependent central nervous system (CNS) plasticity has many different mechanisms and involves essentially every region, from the cortex to the spinal cord. This new knowledge radically changes the challenge of explaining learning and memory and greatly increases the relevance of the spinal cord. The challenge is now to explain how continual and ubiquitous plasticity accounts for the initial acquisition and subsequent stability of many different learned behaviors. The spinal cord has a key role because it is the final common pathway for all behavior and is a site of substantial plasticity. Furthermore, because it is simple, accessible, distant from the rest of the CNS, and directly connected to behavior, the spinal cord is uniquely suited for identifying sites and mechanisms of plasticity and for determining how they account for behavioral change. Experimental models based on spinal cord reflexes facilitate study of the gradual plasticity that makes possible most rapid learning phenomena. These models reveal principles and generate concepts that are likely to apply to learning and memory throughout the CNS. In addition, they offer new approaches to guiding activity-dependent plasticity so as to restore functions lost to injury or disease.
Collapse
Affiliation(s)
- Jonathan R Wolpaw
- Laboratory of Neural Injury and Repair, Wadsworth Center, New York State Department of Health, Albany, NY 12201-0509, USA.
| |
Collapse
|
124
|
Ribic A, Flügge G, Schlumbohm C, Mätz-Rensing K, Walter L, Fuchs E. Activity-dependent regulation of MHC class I expression in the developing primary visual cortex of the common marmoset monkey. Behav Brain Funct 2011; 7:1. [PMID: 21205317 PMCID: PMC3023691 DOI: 10.1186/1744-9081-7-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Accepted: 01/04/2011] [Indexed: 01/31/2023] Open
Abstract
Background Several recent studies have highlighted the important role of immunity-related molecules in synaptic plasticity processes in the developing and adult mammalian brains. It has been suggested that neuronal MHCI (major histocompatibility complex class I) genes play a role in the refinement and pruning of synapses in the developing visual system. As a fast evolutionary rate may generate distinct properties of molecules in different mammalian species, we studied the expression of MHCI molecules in a nonhuman primate, the common marmoset monkey (Callithrix jacchus). Methods and results Analysis of expression levels of MHCI molecules in the developing visual cortex of the common marmoset monkeys revealed a distinct spatio-temporal pattern. High levels of expression were detected very early in postnatal development, at a stage when synaptogenesis takes place and ocular dominance columns are formed. To determine whether the expression of MHCI molecules is regulated by retinal activity, animals were subjected to monocular enucleation. Levels of MHCI heavy chain subunit transcripts in the visual cortex were found to be elevated in response to monocular enucleation. Furthermore, MHCI heavy chain immunoreactivity revealed a banded pattern in layer IV of the visual cortex in enucleated animals, which was not observed in control animals. This pattern of immunoreactivity indicated that higher expression levels were associated with retinal activity coming from the intact eye. Conclusions These data demonstrate that, in the nonhuman primate brain, expression of MHCI molecules is regulated by neuronal activity. Moreover, this study extends previous findings by suggesting a role for neuronal MHCI molecules during synaptogenesis in the visual cortex.
Collapse
Affiliation(s)
- Adema Ribic
- German Primate Center/Leibniz Institute for Primate Research, Göttingen, Germany.
| | | | | | | | | | | |
Collapse
|
125
|
|
126
|
Desgent S, Boire D, Ptito M. Altered expression of parvalbumin and calbindin in interneurons within the primary visual cortex of neonatal enucleated hamsters. Neuroscience 2010; 171:1326-40. [PMID: 20937364 DOI: 10.1016/j.neuroscience.2010.10.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 09/29/2010] [Accepted: 10/04/2010] [Indexed: 01/08/2023]
Abstract
In the present study, we tested the hypothesis that the expression of calcium binding proteins (CaBPs), parvalbumin (PV), calretinin (CR) and calbindin (CB), is dependent upon sensory experience as emphasized in visual deprivation and deafferentation studies. The expression of CaBPs was studied in interneurons within the primary and extrastriate visual cortices (V1, V2M, V2L) and auditory cortex (AC) of adult hamsters enucleated at birth. The effects of enucleation were mainly confined to area V1 where there was a significant volume reduction (26%) and changes in the laminar distribution of PV and CB immunoreactive (IR) cells. The density of PV-IR cell bodies was significantly increased in layer IV and reduced in layer V. Moreover, the density of CB-IR neurons was inferior in layer V of V1 in enucleated hamsters (EH) compared to controls. These results suggest that some features of the laminar distribution of specific CaBPs, in primary sensory cortices, are dependent upon or modulated by sensory input.
Collapse
Affiliation(s)
- S Desgent
- École d'Optométrie, Université de Montréal, Québec, Canada, H3C 3J7
| | | | | |
Collapse
|
127
|
Mix A, Benali A, Eysel UT, Funke K. Continuous and intermittent transcranial magnetic theta burst stimulation modify tactile learning performance and cortical protein expression in the rat differently. Eur J Neurosci 2010; 32:1575-86. [PMID: 20950358 DOI: 10.1111/j.1460-9568.2010.07425.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Repetitive transcranial magnetic stimulation (rTMS) can modulate cortical excitability in a stimulus-frequency-dependent manner. Two kinds of theta burst stimulation (TBS) [intermittent TBS (iTBS) and continuous TBS (cTBS)] modulate human cortical excitability differently, with iTBS increasing it and cTBS decreasing it. In rats, we recently showed that this is accompanied by changes in the cortical expression of proteins related to the activity of inhibitory neurons. Expression levels of the calcium-binding protein parvalbumin (PV) and of the 67-kDa isoform of glutamic acid decarboxylase (GAD67) were strongly reduced following iTBS, but not cTBS, whereas both increased expression of the 65-kDa isoform of glutamic acid decarboxylase. In the present study, to investigate possible functional consequences, we applied iTBS and cTBS to rats learning a tactile discrimination task. Conscious rats received either verum or sham rTMS prior to the task. Finally, to investigate how rTMS and learning effects interact, protein expression was determined for cortical areas directly involved in the task and for those either not, or indirectly, involved. We found that iTBS, but not cTBS, improved learning and strongly reduced cortical PV and GAD67 expression. However, the combination of learning and iTBS prevented this effect in those cortical areas involved in the task, but not in unrelated areas. We conclude that the improved learning found following iTBS is a result of the interaction of two effects, possibly in a homeostatic manner: a general weakening of inhibition mediated by the fast-spiking interneurons, and re-established activity in those neurons specifically involved in the learning task, leading to enhanced contrast between learning-induced and background activity.
Collapse
Affiliation(s)
- Annika Mix
- Department of Neurophysiology, Medical Faculty, Ruhr-University Bochum, Universitaetsstrasse 150, 44780 Bochum, Germany
| | | | | | | |
Collapse
|
128
|
Sugo N, Oshiro H, Takemura M, Kobayashi T, Kohno Y, Uesaka N, Song WJ, Yamamoto N. Nucleocytoplasmic translocation of HDAC9 regulates gene expression and dendritic growth in developing cortical neurons. Eur J Neurosci 2010; 31:1521-32. [PMID: 20525066 DOI: 10.1111/j.1460-9568.2010.07218.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Transcriptional regulation of gene expression is thought to play a pivotal role in activity-dependent neuronal differentiation and circuit formation. Here, we investigated the role of histone deacetylase 9 (HDAC9), which regulates transcription by histone modification, in the development of neocortical neurons. The translocation of HDAC9 from nucleus to cytoplasm was induced by an increase of spontaneous firing activity in cultured mouse cortical neurons. This nucleocytoplasmic translocation was also observed in postnatal development in vivo. The translocation-induced gene expression and cellular morphology was further examined by introducing an HDAC9 mutant that disrupts the nucleocytoplasmic translocation. Expression of c-fos, an immediately-early gene, was suppressed in the mutant-transfected cells regardless of neural activity. Moreover, the introduction of the mutant decreased the total length of dendritic branches, whereas knockdown of HDAC9 promoted dendritic growth. These findings indicate that chromatin remodeling with nucleocytoplasmic translocation of HDAC9 regulates activity-dependent gene expression and dendritic growth in developing cortical neurons.
Collapse
Affiliation(s)
- Noriyuki Sugo
- Neuroscience Laboratories, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
129
|
Beston BR, Jones DG, Murphy KM. Experience-dependent changes in excitatory and inhibitory receptor subunit expression in visual cortex. Front Synaptic Neurosci 2010; 2:138. [PMID: 21423524 PMCID: PMC3059668 DOI: 10.3389/fnsyn.2010.00138] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 08/06/2010] [Indexed: 02/01/2023] Open
Abstract
Experience-dependent development of visual cortex depends on the balance between excitatory and inhibitory activity. This activity is regulated by key excitatory (NMDA, AMPA) and inhibitory (GABAA) receptors. The composition of these receptors changes developmentally, affecting the excitatory–inhibitory (E/I) balance and synaptic plasticity. Until now, it has been unclear how abnormal visual experience affects this balance. To examine this question, we measured developmental changes in excitatory and inhibitory receptor subunits in visual cortex following normal visual experience and monocular deprivation. We used Western blot analysis to quantify expression of excitatory (NR1, NR2A, NR2B, GluR2) and inhibitory (GABAAα1, GABAAα3) receptor subunits. Monocular deprivation promoted a complex pattern of changes in receptor subunit expression that varied with age and was most severe in the region of visual cortex representing the central visual field. To characterize the multidimensional pattern of experience-dependent change in these synaptic mechanisms, we applied a neuroinformatics approach using principal component analysis. We found that monocular deprivation (i) causes a large portion of the normal developmental trajectory to be bypassed, (ii) shifts the E/I balance in favor of more inhibition, and (iii) accelerates the maturation of receptor subunits. Taken together, these results show that monocularly deprived animals have an abnormal balance of the synaptic machinery needed for functional maturation of cortical circuits and for developmental plasticity. This raises the possibility that interventions intended to treat amblyopia may need to address multiple synaptic mechanisms to produce optimal recovery.
Collapse
Affiliation(s)
- Brett R Beston
- McMaster Integrative Neuroscience Discovery and Study Program, McMaster University Hamilton, ON, Canada
| | | | | |
Collapse
|
130
|
Structural dynamics of synapses in vivo correlate with functional changes during experience-dependent plasticity in visual cortex. J Neurosci 2010; 30:11086-95. [PMID: 20720116 DOI: 10.1523/jneurosci.1661-10.2010] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The impact of activity on neuronal circuitry is complex, involving both functional and structural changes whose interaction is largely unknown. We have used optical imaging of mouse visual cortex responses and two-photon imaging of superficial layer spines on layer 5 neurons to monitor network function and synaptic structural dynamics in the mouse visual cortex in vivo. Total lack of vision due to dark-rearing from birth dampens visual responses and shifts spine dynamics and morphologies toward an immature state. The effects of vision after dark rearing are strongly dependent on the timing of exposure: over a period of days, functional and structural changes are temporally related such that light stabilizes spines while increasing visually driven activity. The effects of long-term light exposure can be partially mimicked by experimentally enhancing inhibitory signaling in the darkness. Brief light exposure, however, results in a rapid, transient, NMDA-dependent increase of cortical responses, accompanied by increased dynamics of dendritic spines. These findings indicate that visual experience induces rapid reorganization of cortical circuitry followed by a period of stabilization, and demonstrate a close relationship between dynamic changes at single synapses and cortical network function.
Collapse
|
131
|
Cerri C, Restani L, Caleo M. Callosal contribution to ocular dominance in rat primary visual cortex. Eur J Neurosci 2010; 32:1163-9. [PMID: 20726891 DOI: 10.1111/j.1460-9568.2010.07363.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Ocular dominance (OD) plasticity triggered by monocular eyelid suture is a classic paradigm for studying experience-dependent changes in neural connectivity. Recently, rodents have become the most popular model for studies of OD plasticity. It is therefore important to determine how OD is determined in the rodent primary visual cortex. In particular, cortical cells receive considerable inputs from the contralateral hemisphere via callosal axons, but the role of these connections in controlling eye preference remains controversial. Here we have examined the role of callosal connections in binocularity of the visual cortex in naïve young rats. We recorded cortical responses evoked by stimulation of each eye before and after acute silencing, via stereotaxic tetrodotoxin (TTX) injection, of the lateral geniculate nucleus ipsilateral to the recording site. This protocol allowed us to isolate visual responses transmitted via the corpus callosum. Cortical binocularity was assessed by visual evoked potential (VEP) and single-unit recordings. We found that acute silencing of afferent geniculocortical input produced a very significant reduction in the contralateral-to-ipsilateral (C/I) VEP ratio, and a marked shift towards the ipsilateral eye in the OD distribution of cortical cells. Analysis of absolute strength of each eye indicated a dramatic decrease in contralateral eye responses following TTX, while those of the ipsilateral eye were reduced but maintained a more evident input. We conclude that callosal connections contribute to normal OD mainly by carrying visual input from the ipsilateral eye. These data have important implications for the interpretation of OD plasticity following alterations of visual experience.
Collapse
|
132
|
Silingardi D, Scali M, Belluomini G, Pizzorusso T. Epigenetic treatments of adult rats promote recovery from visual acuity deficits induced by long-term monocular deprivation. Eur J Neurosci 2010; 31:2185-92. [DOI: 10.1111/j.1460-9568.2010.07261.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
133
|
Talaei SA, Sheibani V, Salami M. Light deprivation improves melatonin related suppression of hippocampal plasticity. Hippocampus 2010; 20:447-55. [PMID: 19475653 DOI: 10.1002/hipo.20650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In early postnatal life, sensory inputs deeply influence development as well as function of the brain. Plasticity of synaptic transmission including its experimentally induced form, long-term potentiation (LTP), is affected by sensory deprivation in neocortex. This study is devoted to assess if dark rearing and a dark phase synthesized hormone melatonin influence LTP in the hippocampus, an area of brain involved in learning and memory. In vivo experiments were carried out on two groups of 45-days-old male Wistar rats kept in standard 12-h light/dark condition [light reared (LR) tested during the light phase] or in complete darkness [dark reared (DR)] since birth to testing. Each group, in turn, was divided to two, vehicle- and melatonin-treated, groups. Stimulating the Schaffer collaterals of CA3 area of hippocampus extracellular postsynaptic potentials (EPSPs) were recorded in the CA1 area. Having the stable baseline responses to the test pulses, the hippocampus was perfused by either vehicle or 2 microg melatonin and EPSPs were recorded for 30 min. Then, for induction of LTP, the tetanus was applied to the Schaffer collaterals and the field potentials were pooled for 120-min post-tetanus. The light deprivation resulted in a significant augmentation in the amplitude of baseline responses. Also, we observed a melatonin-induced increase in amplitude of the baseline recordings in either LR or DR animals. Tetanic stimulation elicited LTP of EPSPs in both LR and DR groups, robustly in the former where it lasted for about 90 min. Generally, melatonin inhibited the production of LTP in the two groups especially in the LR animals leading to a noticeable depression. We concluded that higher level of neuronal activity in the DR rats gives rise to a lower level of LTP. Weaker effect of melatonin on blocking the potentiation of post-tetanus EPSPs in the DR rats may be the result of a desensitization of melatonin receptors due to chronically increased levels of this hormone in the visually deprived rats.
Collapse
Affiliation(s)
- Sayyed Alireza Talaei
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, I. R. Iran
| | | | | |
Collapse
|
134
|
Woo TUW, Spencer K, McCarley RM. Gamma oscillation deficits and the onset and early progression of schizophrenia. Harv Rev Psychiatry 2010; 18:173-89. [PMID: 20415633 PMCID: PMC2860612 DOI: 10.3109/10673221003747609] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
A fascinating convergence of evidence in recent years has implicated the disturbances of neural synchrony in the gamma frequency band (30-100 Hz) as a major pathophysiologic feature of schizophrenia. Evidence suggests that reduced glutamatergic neurotransmission via the N-methyl-D-aspartate (NMDA) receptors that are localized to inhibitory interneurons, perhaps especially the fast-spiking cells that contain the calcium-binding protein parvalbumin (PV), may contribute to gamma band synchrony deficits. These deficits may underlie the brain's failure to integrate information and hence the manifestations of many symptoms and deficits of schizophrenia. Furthermore, because gamma oscillations are thought to provide the temporal structure that is necessary for synaptic plasticity, gamma oscillation deficits may disturb the developmental synaptic reorganization process that is occurring during the period of late adolescence and early adulthood. This disturbance may contribute to the onset of schizophrenia and the functional deterioration that is characteristic of the early stage of the illness. Finally, reduced NMDA neurotransmission on inhibitory interneurons, including the PV-containing cells, may inflict excitotoxic or oxidative injury to downstream pyramidal neurons, leading to further loss of synapses and dendritic branchings. Hence, a key element in the conceptualization of rational early-intervention and prevention strategies for schizophrenia may involve correcting the abnormal NMDA neurotransmission on inhibitory interneurons-possibly that on the PV-containing neurons, in particular-thereby normalizing gamma oscillation deficits and attenuating downstream neuronal pathology.
Collapse
Affiliation(s)
- Tsung-Ung W. Woo
- Laboratory of Translational Psychiatry, Mailman Research Center McLean Hospital Belmont, MA 02478,Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA 02215,Department of Psychiatry, Harvard Medical School, Boston, MA 02115
| | - Kevin Spencer
- Department of Psychiatry, VA Boston Healthcare System, Brockton, MA 02301,Department of Psychiatry, Harvard Medical School, Boston, MA 02115
| | - Robert M. McCarley
- Laboratory of Translational Psychiatry, Mailman Research Center McLean Hospital Belmont, MA 02478,Department of Psychiatry, VA Boston Healthcare System, Brockton, MA 02301,Department of Psychiatry, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
135
|
Zhou M, Lei Z, Li H, Yi W, Zhang Z, Guo A. NMDA receptors-dependent plasticity in the phototaxis preference behavior induced by visual deprivation in young and adult flies. GENES BRAIN AND BEHAVIOR 2010; 9:325-34. [DOI: 10.1111/j.1601-183x.2009.00562.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
136
|
Loss of Arc renders the visual cortex impervious to the effects of sensory experience or deprivation. Nat Neurosci 2010; 13:450-7. [PMID: 20228806 PMCID: PMC2864583 DOI: 10.1038/nn.2508] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 01/27/2010] [Indexed: 12/01/2022]
Abstract
A myriad of mechanisms are suggested to account for the full richness of visual cortical plasticity. We report that visual cortex lacking Arc is impervious to the effects of deprivation or experience. Using intrinsic signal imaging and chronic visually evoked potential recordings, we find that Arc−/− mice do not exhibit depression of deprived eye responses or a shift in ocular dominance after brief monocular deprivation. Extended deprivation also fails to elicit a shift in ocular dominance or open eye potentiation. Moreover, Arc−/− mice lack stimulus–selective response potentiation. Although Arc−/− mice exhibit normal visual acuity, baseline ocular dominance is abnormal and resembles that observed after dark–rearing. These data suggest that Arc is required for the experience–dependent processes that normally establish and modify synaptic connections in visual cortex.
Collapse
|
137
|
Dnmt1 and Dnmt3a maintain DNA methylation and regulate synaptic function in adult forebrain neurons. Nat Neurosci 2010; 13:423-30. [PMID: 20228804 DOI: 10.1038/nn.2514] [Citation(s) in RCA: 756] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Accepted: 02/15/2010] [Indexed: 12/11/2022]
Abstract
Dnmt1 and Dnmt3a are important DNA methyltransferases that are expressed in postmitotic neurons, but their function in the CNS is unclear. We generated conditional mutant mice that lack Dnmt1, Dnmt3a or both exclusively in forebrain excitatory neurons and found that only double knockout (DKO) mice showed abnormal long-term plasticity in the hippocampal CA1 region together with deficits in learning and memory. Although we found no neuronal loss, hippocampal neurons in DKO mice were smaller than in the wild type; furthermore, DKO neurons showed deregulated expression of genes, including the class I MHC genes and Stat1, that are known to contribute to synaptic plasticity. In addition, we observed a significant decrease in DNA methylation in DKO neurons. We conclude that Dnmt1 and Dnmt3a are required for synaptic plasticity, learning and memory through their overlapping roles in maintaining DNA methylation and modulating neuronal gene expression in adult CNS neurons.
Collapse
|
138
|
Schaevitz LR, Moriuchi JM, Nag N, Mellot TJ, Berger-Sweeney J. Cognitive and social functions and growth factors in a mouse model of Rett syndrome. Physiol Behav 2010; 100:255-63. [PMID: 20045424 DOI: 10.1016/j.physbeh.2009.12.025] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Revised: 12/18/2009] [Accepted: 12/22/2009] [Indexed: 11/18/2022]
Abstract
Rett syndrome (RTT) is an autism-spectrum disorder caused by mutations in the X-linked gene encoding methyl-CpG-binding protein 2 (MeCP2). Abnormalities in social behavior, stereotyped movements, and restricted interests are common features in both RTT and classic autism. While mouse models of both RTT and autism exist, social behaviors have not been explored extensively in mouse models of RTT. Here, we report cognitive and social abnormalities in Mecp2(1lox) null mice, an animal model of RTT. The null mice show severe deficits in short- and long-term object recognition memories, reminiscent of the severe cognitive deficits seen in RTT girls. Social behavior, however, is abnormal in that the null mice spend more time in contact with stranger mice than do wildtype controls. These findings are consistent with reports of increased reciprocal social interaction in RTT girls relative to classic autism. We also report here that the levels of the neurotrophins brain-derived neurotrophic factor (BDNF), insulin-like growth factor-1 (IGF-1), and nerve growth factor (NGF) are decreased in the hippocampus of the null mice, and discuss how this may provide an underlying mechanism for both the cognitive deficits and the increased motivation for social contact observed in the Mecp2(1lox) null mice. These studies support a differential etiology between RTT and autism, particularly with respect to sociability deficits.
Collapse
Affiliation(s)
- Laura R Schaevitz
- Department of Biological Sciences, Wellesley College, Wellesley MA 02481, United States
| | | | | | | | | |
Collapse
|
139
|
Loebrich S, Nedivi E. The function of activity-regulated genes in the nervous system. Physiol Rev 2009; 89:1079-103. [PMID: 19789377 DOI: 10.1152/physrev.00013.2009] [Citation(s) in RCA: 165] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The mammalian brain is plastic in the sense that it shows a remarkable capacity for change throughout life. The contribution of neuronal activity to brain plasticity was first recognized in relation to critical periods of development, when manipulating the sensory environment was found to profoundly affect neuronal morphology and receptive field properties. Since then, a growing body of evidence has established that brain plasticity extends beyond development and is an inherent feature of adult brain function, spanning multiple domains, from learning and memory to adaptability of primary sensory maps. Here we discuss evolution of the current view that plasticity of the adult brain derives from dynamic tuning of transcriptional control mechanisms at the neuronal level, in response to external and internal stimuli. We then review the identification of "plasticity genes" regulated by changes in the levels of electrical activity, and how elucidating their cellular functions has revealed the intimate role transcriptional regulation plays in fundamental aspects of synaptic transmission and circuit plasticity that occur in the brain on an every day basis.
Collapse
Affiliation(s)
- Sven Loebrich
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
140
|
Laprairie JL, Murphy AZ. Neonatal injury alters adult pain sensitivity by increasing opioid tone in the periaqueductal gray. Front Behav Neurosci 2009; 3:31. [PMID: 19862348 PMCID: PMC2766783 DOI: 10.3389/neuro.08.031.2009] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Accepted: 08/27/2009] [Indexed: 11/13/2022] Open
Abstract
Studies in both rodents and humans have shown that acute inflammatory pain experienced during the perinatal period produces long-term decreases in pain sensitivity (hypoalgesia) (Grunau et al., 1994a, 2001; Ren et al., 2004; LaPrairie and Murphy, 2007). To date, the mechanisms underlying these long-term adaptations, however, have yet to be elucidated. The present studies tested the hypothesis that neonatal inflammatory pain induces an upregulation in endogenous opioid tone that is maintained into adulthood, and that this increase in opioid tone provides the underlying mechanism for the observed hypoalgesia. On the day of birth (P0), inflammatory pain was induced in male and female Sprague-Dawley rats by intraplantar administration of carrageenan (CGN; 1%). In adulthood (P60), these animals displayed significantly increased paw withdrawal latencies in response to a noxious thermal stimulus in comparison to controls. Systemic administration of the brain-penetrant opioid receptor antagonist naloxone HCl, but not the peripherally restricted naloxone methiodide, significantly attenuated the injury-induced hypoalgesia. Direct administration of naloxone HCl or antagonists directed at the mu or delta opioid receptors into the midbrain periaqueductal gray (PAG) also significantly reversed the injury-induced hypoalgesia in adult rats. Parallel anatomical studies revealed that inflammatory pain experienced on the day of birth significantly increased beta-endorphin and met/leu-enkephalin protein levels and decreased opioid receptor expression in the PAG of the adult rat. Thus, early noxious insult produces long-lasting alterations in endogenous opioid tone, thereby profoundly impacting nociceptive responsiveness in adulthood.
Collapse
|
141
|
Regionalized loss of parvalbumin interneurons in the cerebral cortex of mice with deficits in GFRalpha1 signaling. J Neurosci 2009; 29:10695-705. [PMID: 19710321 DOI: 10.1523/jneurosci.2658-09.2009] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Inhibitory interneurons are crucially important for cerebral cortex function and behavior. The mechanisms controlling inhibitory interneuron diversification and allocation to distinct cortical areas remain poorly understood. GDNF (glial cell line-derived neurotrophic factor) and its receptor GFRalpha1 have been implicated in the development of GABAergic precursors but, because of the early lethality of null mutants, their roles in postnatal maturation and function of cortical interneurons are unknown. "cis-only" mutant mice lack GFRalpha1 only in cells that do not express the RET signaling receptor subunit and survive to adulthood. At birth, both null mutants and cis-only mice showed a specific loss of GABAergic interneurons in rostro- and caudolateral cortical regions but not in more medial areas. Unexpectedly, the adult cortex of cis-only mice displayed a complete loss of parvalbumin (PV)-expressing GABAergic interneurons in discrete regions (PV holes) interspersed among areas of normal PV cell density. PV holes predominantly occurred in the visual and frontal cortices, and their size could be affected by neuronal activity. Consistent with deficits in cortical inhibitory activity, these mice showed enhanced cortical excitability, increased sensitivity to epileptic seizure, and increased social behavior. We propose that GFRalpha1 signaling guides the development of a subset of PV-expressing GABAergic interneurons populating discrete regions of the cerebral cortex and may thus contribute to the diversification and allocation of specific cortical interneuron subtypes.
Collapse
|
142
|
Yang L, Pan Z, Zhou L, Lin S, Wu K. Continuously changed genes during postnatal periods in rat visual cortex. Neurosci Lett 2009; 462:162-5. [DOI: 10.1016/j.neulet.2009.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 06/10/2009] [Accepted: 07/02/2009] [Indexed: 10/20/2022]
|
143
|
Theta burst and conventional low-frequency rTMS differentially affect GABAergic neurotransmission in the rat cortex. Exp Brain Res 2009; 199:411-21. [DOI: 10.1007/s00221-009-1961-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2008] [Accepted: 07/22/2009] [Indexed: 01/16/2023]
|
144
|
Rittenhouse CD, Majewska AK. Synaptic Mechanisms of Activity-Dependent Remodeling in Visual Cortex during Monocular Deprivation. J Exp Neurosci 2009. [DOI: 10.4137/jen.s2559] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
It has long been appreciated that in the visual cortex, particularly within a postnatal critical period for experience-dependent plasticity, the closure of one eye results in a shift in the responsiveness of cortical cells toward the experienced eye. While the functional aspects of this ocular dominance shift have been studied for many decades, their cortical substrates and synaptic mechanisms remain elusive. Nonetheless, it is becoming increasingly clear that ocular dominance plasticity is a complex phenomenon that appears to have an early and a late component. Early during monocular deprivation, deprived eye cortical synapses depress, while later during the deprivation open eye synapses potentiate. Here we review current literature on the cortical mechanisms of activity-dependent plasticity in the visual system during the critical period. These studies shed light on the role of activity in shaping neuronal structure and function in general and can lead to insights regarding how learning is acquired and maintained at the neuronal level during normal and pathological brain development.
Collapse
Affiliation(s)
| | - Ania K Majewska
- Department of Neurobiology and Anatomy, University of Rochester, Rochester, NY
| |
Collapse
|
145
|
McCoy PA, Huang HS, Philpot BD. Advances in understanding visual cortex plasticity. Curr Opin Neurobiol 2009; 19:298-304. [PMID: 19540104 DOI: 10.1016/j.conb.2009.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 05/21/2009] [Accepted: 05/25/2009] [Indexed: 01/21/2023]
Abstract
Visual cortical plasticity can be either rapid, occurring in response to abrupt changes in neural activity, or slow, occurring over days as a homeostatic process for adapting neuronal responsiveness. Recent advances have shown that the magnitude and polarity of rapid synaptic modifications are regulated by neuromodulators, while homeostatic modifications can occur through regulation of cytokine actions or N-methyl-d-aspartate (NMDA) receptor subunit composition. Synaptic and homeostatic plasticity together produce the normal physiological response to monocular impairments. In vivo studies have now overturned the dogma that robust plasticity is limited to an early critical period. Indeed, rapid physiological plasticity in the adult can be enabled by prior, experience-driven anatomical rearrangements or through pharmacological manipulations of the epigenome.
Collapse
Affiliation(s)
- Portia A McCoy
- Department of Cell and Molecular Physiology, Neuroscience Center, Curriculum in Neurobiology, and Neurodevelopmental Disorders Research Center, University of North Carolina, Chapel Hill, NC 27599-7545, USA.
| | | | | |
Collapse
|
146
|
Leamey CA, Van Wart A, Sur M. Intrinsic patterning and experience-dependent mechanisms that generate eye-specific projections and binocular circuits in the visual pathway. Curr Opin Neurobiol 2009; 19:181-7. [PMID: 19502049 DOI: 10.1016/j.conb.2009.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2009] [Revised: 05/14/2009] [Accepted: 05/15/2009] [Indexed: 01/10/2023]
Abstract
A defining feature of the mammalian nervous system is its complex yet precise circuitry. The mechanisms which underlie the generation of neural connectivity are the topic of intense study in developmental neuroscience. The mammalian visual pathway demonstrates precise retinotopic organization in subcortical and cortical pathways, together with the alignment and matching of eye-specific projections, and sophisticated cortical circuitry that enables the extraction of features underlying vision. New approaches employing molecular-genetic analyses, transgenic mice, novel recombinant probes, and high-resolution imaging are contributing to rapid progress and a new synthesis in the field. These approaches are revealing the ways in which intrinsic patterning mechanisms act in concert with experience-dependent mechanisms to shape visual projections and circuits.
Collapse
Affiliation(s)
- Catherine A Leamey
- Discipline of Physiology, School of Medical Sciences and Bosch Institute, University of Sydney, NSW 2006, Australia.
| | | | | |
Collapse
|
147
|
Yashiro K, Riday TT, Condon KH, Roberts AC, Bernardo DR, Prakash R, Weinberg RJ, Ehlers MD, Philpot BD. Ube3a is required for experience-dependent maturation of the neocortex. Nat Neurosci 2009; 12:777-83. [PMID: 19430469 PMCID: PMC2741303 DOI: 10.1038/nn.2327] [Citation(s) in RCA: 254] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2008] [Accepted: 03/24/2009] [Indexed: 02/06/2023]
Abstract
Experience-dependent maturation of neocortical circuits is required for normal sensory and cognitive abilities, which are distorted in neurodevelopmental disorders. We tested whether experience-dependent neocortical modifications require Ube3a, an E3 ubiquitin ligase whose dysregulation has been implicated in autism and Angelman syndrome. Using visual cortex as a model, we found that experience-dependent maturation of excitatory cortical circuits was severely impaired in Angelman syndrome model mice deficient in Ube3a. This developmental defect was associated with profound impairments in neocortical plasticity. Normal plasticity was preserved under conditions of sensory deprivation, but was rapidly lost by sensory experiences. The loss of neocortical plasticity is reversible, as late-onset visual deprivation restored normal synaptic plasticity. Furthermore, Ube3a-deficient mice lacked ocular dominance plasticity in vivo when challenged with monocular deprivation. We conclude that Ube3a is necessary for maintaining plasticity during experience-dependent neocortical development and suggest that the loss of neocortical plasticity contributes to deficits associated with Angelman syndrome.
Collapse
Affiliation(s)
- Koji Yashiro
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
- K.Y. present address: Urogenix Inc, Durham NC 27713, USA
| | - Thorfinn T. Riday
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Kathryn H. Condon
- Department of Neurobiology Duke University Medical Center, Durham, NC 27710, USA
| | - Adam C. Roberts
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Neurodevelopmental Disorders Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Danilo R. Bernardo
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rohit Prakash
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Richard J. Weinberg
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Cell and Developmental Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Michael D. Ehlers
- Department of Neurobiology Duke University Medical Center, Durham, NC 27710, USA
- Howard Hughes Medical Institute, Duke University Medical Center, Durham, NC 27710, USA
| | - Benjamin D. Philpot
- Department of Cell and Molecular Physiology, University of North Carolina, Chapel Hill, NC 27599, USA
- Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Neurodevelopmental Disorders Research Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
148
|
Abstract
Environmental enrichment (EE) was shown recently to accelerate brain development in rodents. Increased levels of maternal care, and particularly tactile stimulation through licking and grooming, may represent a key component in the early phases of EE. We hypothesized that enriching the environment in terms of body massage may thus accelerate brain development in infants. We explored the effects of body massage in preterm infants and found that massage accelerates the maturation of electroencephalographic activity and of visual function, in particular visual acuity. In massaged infants, we found higher levels of blood IGF-1. Massage accelerated the maturation of visual function also in rat pups and increased the level of IGF-1 in the cortex. Antagonizing IGF-1 action by means of systemic injections of the IGF-1 antagonist JB1 blocked the effects of massage in rat pups. These results demonstrate that massage has an influence on brain development and in particular on visual development and suggest that its effects are mediated by specific endogenous factors such as IGF-1.
Collapse
|
149
|
Enrich the environment to empower the brain. Trends Neurosci 2009; 32:233-9. [DOI: 10.1016/j.tins.2008.12.004] [Citation(s) in RCA: 242] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2008] [Revised: 11/15/2008] [Accepted: 12/10/2008] [Indexed: 11/23/2022]
|
150
|
Partial reversal of Rett Syndrome-like symptoms in MeCP2 mutant mice. Proc Natl Acad Sci U S A 2009; 106:2029-34. [PMID: 19208815 DOI: 10.1073/pnas.0812394106] [Citation(s) in RCA: 429] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Rett Syndrome (RTT) is a severe form of X-linked mental retardation caused by mutations in the gene coding for methyl CpG-binding protein 2 (MECP2). Mice deficient in MeCP2 have a range of physiological and neurological abnormalities that mimic the human syndrome. Here we show that systemic treatment of MeCP2 mutant mice with an active peptide fragment of Insulin-like Growth Factor 1 (IGF-1) extends the life span of the mice, improves locomotor function, ameliorates breathing patterns, and reduces irregularity in heart rate. In addition, treatment with IGF-1 peptide increases brain weight of the mutant mice. Multiple measurements support the hypothesis that RTT results from a deficit in synaptic maturation in the brain: MeCP2 mutant mice have sparse dendritic spines and reduced PSD-95 in motor cortex pyramidal neurons, reduced synaptic amplitude in the same neurons, and protracted cortical plasticity in vivo. Treatment with IGF-1 peptide partially restores spine density and synaptic amplitude, increases PSD-95, and stabilizes cortical plasticity to wild-type levels. Our results thus strongly suggest IGF-1 as a candidate for pharmacological treatment of RTT and potentially of other CNS disorders caused by delayed synapse maturation.
Collapse
|