Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Critical period plasticity in local cortical circuits

Key Points

  • Critical periods of brain development have been identified across sensory systems, but are best characterized in the visual cortex where the inputs from the two eyes first converge and compete for space, both functionally and structurally. In children born with opacity or deviation of the eyes, the consequent loss of cortical spiking response to the deprived eye (ocular dominance plasticity) leads to lifelong amblyopia (although the retina may remain healthy). Critical periods for other receptive field properties (for example, orientation bias or visual motion) might vary in their developmental timing.

  • Although in vitro models of excitatory homosynaptic plasticity have been doubly dissociated from ocular dominance changes in vivo, genetic or pharmacological manipulation of the development of GABA (γ-aminobutyric acid)-mediated transmission yields consistent and direct control over critical period timing and its eventual anatomical outcomes (such as spine pruning and column spacing).

  • Several lines of evidence implicate one particular GABA-containing cell type — the parvalbumin-positive large basket cell — as the critical period trigger. (1) They develop with a late postnatal time course in tight correlation with plasticity onset; (2) brain-derived neurotrophic factor (BDNF) accelerates the onset of both development and plasticity; (3) dark-rearing delays the onset of both; (4) they make somatic synapses containing GABAA (GABA type A) receptor α1-subunits, which mediate critical period acceleration by benzodiazepines; (5) their ability to fire at high rates determines the rate of plasticity; (6) their long, horizontal axons span and can influence ocular dominance column size; and (7) with age they are preferentially enwrapped by perineuronal nets, the removal of which in adulthood (by chondroitinases) reactivates plasticity.

  • After induction by GABA cell development, critical period expression follows a molecular cascade leading toward structural refinement through the actions of proteases and neurotrophins. Proteolytic (tissue-type plasminogen activator, tPA) action increases on sensory deprivation, leading, during the critical period only, to dendritic spine motility, pruning, and axonal retraction followed by growth.

  • How might GABA circuits trigger plasticity in vivo? Two heuristic models are proposed. First, 'instructive' editing of back-propagating action potentials at the soma to control synaptic plasticity in the dendrites. Second, 'permissive' synchrony detection as a network of parvalbumin-positive cells coupled by gap junctions and mutual inhibitory connections to regulate the release and uptake of proteases and growth factors. In this case, competition occurs extracellularly.

  • The article also discusses a general role for local circuit inhibition in the experience-dependent development of other brain systems.

Abstract

Neuronal circuits in the brain are shaped by experience during 'critical periods' in early postnatal life. In the primary visual cortex, this activity-dependent development is triggered by the functional maturation of local inhibitory connections and driven by a specific, late-developing subset of interneurons. Ultimately, the structural consolidation of competing sensory inputs is mediated by a proteolytic reorganization of the extracellular matrix that occurs only during the critical period. The reactivation of this process, and subsequent recovery of function in conditions such as amblyopia, can now be studied with realistic circuit models that might generalize across systems.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Local circuit control of developing columnar architecture in the neocortex.
Figure 2: GABA-mediated control of the critical period.
Figure 3: Heterogeneity of local GABA circuits in the neocortex.
Figure 4: Specific GABAA circuits for visual cortical plasticity.
Figure 5: Structural consolidation during the critical period.
Figure 6: Molecular mechanisms of visual cortical plasticity.
Figure 7: Two models for inhibitory control of sensory plasticity.

Similar content being viewed by others

References

  1. Kim, K. H., Relkin, N. R., Lee, K. M. & Hirsch, J. Distinct cortical areas associated with native and second languages. Nature 388, 171–174 (1997).

    Article  CAS  PubMed  Google Scholar 

  2. Elbert, T., Pantev, C., Wienbruch, C., Rockstroh, B. & Taub, E. Increased cortical representation of the fingers of the left hand in string players. Science 270, 305–307 (1995).

    Article  CAS  PubMed  Google Scholar 

  3. Pantev, C. et al. Increased auditory cortical representation in musicians. Nature 392, 811–814 (1998).

    Article  CAS  PubMed  Google Scholar 

  4. Hensch, T. K. Critical period regulation. Annu. Rev. Neurosci. 27, 549–579 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Wiesel, T. N. & Hubel, D. H. Single-cell responses in striate cortex of kittens deprived of vision in one eye. J. Neurophysiol. 26, 1003–1017 (1963). A classic paper for anyone interested in the fields of development, plasticity and vision.

    Article  CAS  PubMed  Google Scholar 

  6. Berardi, N., Pizzorusso, T. & Maffei, L. Critical periods during sensory development. Curr. Opin. Neurobiol. 10, 138–145 (2000).

    Article  CAS  PubMed  Google Scholar 

  7. Daw, N. Visual Development (Plenum, New York, 1995).

    Book  Google Scholar 

  8. Hubel, D. H., Wiesel, T. N. & LeVay, S. Functional architecture of area 17 in normal and monocularly deprived macaque monkeys. Cold Spring Harb. Symp. Quant. Biol. 40, 581–589 (1976).

    Article  CAS  PubMed  Google Scholar 

  9. Shatz, C. J. & Stryker, M. P. Ocular dominance in layer IV of the cat's visual cortex and the effects of monocular deprivation. J. Physiol. (Lond.) 281, 267–283 (1978).

    Article  CAS  Google Scholar 

  10. Antonini, A. & Stryker, M. P. Plasticity of geniculocortical afferents following brief or prolonged monocular occlusion in the cat. J. Comp. Neurol. 369, 64–82 (1996).

    Article  CAS  PubMed  Google Scholar 

  11. Antonini, A., Fagiolini, M. & Stryker, M. P. Anatomical correlates of functional plasticity in mouse visual cortex. J. Neurosci. 19, 4388–4406 (1999). Details the well known anatomical consequences of monocular deprivation in terms of thalamic input to the neocortex. These structural events are far slower than intracortical changes (discussed in references 12, 13, 85 and 86).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Trachtenberg, J. T., Trepel, C. & Stryker, M. P. Rapid extragranular plasticity in the absence of thalamocortical plasticity in the developing primary visual cortex. Science 287, 2029–2032 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Trachtenberg, J. T. & Stryker, M. P. Rapid anatomical plasticity of horizontal connections in the developing visual cortex. J. Neurosci. 21, 3476–3482 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Katz, L. C. & Crowley, J. C. Development of cortical circuits: lessons from ocular dominance columns. Nature Rev. Neurosci. 3, 34–42 (2002).

    Article  CAS  Google Scholar 

  15. Kaschube, M., Wolf, F., Geisel, T. & Lowel, S. Genetic influence on quantitative features of neocortical architecture. J. Neurosci. 22, 7206–7217 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Adams, D. L. & Horton, J. C. Shadows cast by retinal blood vessels mapped in primary visual cortex. Science 298, 572–576 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Willshaw, D. J. & von der Malsburg, C. How patterned neural connections can be set up by self-organization. Proc. R. Soc. Lond. B 194, 431–445 (1976).

    Article  CAS  PubMed  Google Scholar 

  18. Miller, K. D., Keller, J. B. & Stryker, M. P. Ocular dominance column development: analysis and simulation. Science 245, 605–615 (1989).

    Article  CAS  PubMed  Google Scholar 

  19. Hensch, T. K. & Stryker, M. P. Columnar architecture sculpted by GABA circuits in developing cat visual cortex. Science 303, 1678–1681 (2004). First bidirectional shaping of cortical column size by the direct manipulation of lateral inhibition, as predicted by earlier theoretical models (described in references 17 and18).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sieghart, W. Structure and pharmacology of γ-aminobutyric acidA receptor subtypes. Pharmacol. Rev. 47, 181–234 (1995).

    CAS  PubMed  Google Scholar 

  21. Lowel, S. Ocular dominance column development: strabismus changes the spacing of adjacent columns in cat visual cortex. J. Neurosci. 14, 7451–7468 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Horton, J. C. & Hocking, D. R. Intrinsic variability of ocular dominance column periodicity in normal macaque monkeys. J. Neurosci. 16, 7228–7239 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kasthuri, N. & Lichtman, J. W. The role of neuronal identity in synaptic competition. Nature 424, 426–430 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Buffelli, M. et al. Genetic evidence that relative synaptic efficacy biases the outcome of synaptic competition. Nature 424, 430–434 (2003). An elegant genetic dissection of competition at a visible peripheral synapse.

    Article  CAS  PubMed  Google Scholar 

  25. Somogyi, P., Tamas, G., Lujan, R. & Buhl, E. H. Salient features of synaptic organisation in the cerebral cortex. Brain Res. Rev. 26, 113–135 (1998).

    Article  CAS  PubMed  Google Scholar 

  26. Hensch, T. K. & Fagiolini, M. (eds) Excitatory–Inhibitory Balance: Synapses, Circuits, Systems (Kluwer/Plenum, New York, 2004).

    Book  Google Scholar 

  27. Liu, G. Local structural balance and functional interaction of excitatory and inhibitory synapses in hippocampal dendrites. Nature Neurosci. 7, 373–379 (2004).

    Article  CAS  PubMed  Google Scholar 

  28. Turrigiano, G. G. & Nelson, S. B. Homeostatic plasticity in the developing nervous system. Nature Rev. Neurosci. 5, 97–107 (2004).

    Article  CAS  Google Scholar 

  29. Desai, N. S., Cudmore, R. H., Nelson, S. B. & Turrigiano, G. G. Critical periods for experience-dependent synaptic scaling in visual cortex. Nature Neurosci. 5, 783–789 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Maffei, A., Nelson, S. B. & Turrigiano, G. G. Selective reconfiguration of layer 4 visual cortical circuitry by visual deprivation. Nature Neurosci. 7, 1353–1359 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Long, M. A., Cruikshank, S. J., Jutras, M. J. & Connors, B. W. Abrupt maturation of a spike-synchronizing mechanism in neocortex. J. Neurosci. 25, 7309–7316 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Shaw, C. & Cynader, M. Disruption of cortical activity prevents ocular dominance changes in monocularly deprived kittens. Nature 308, 731–734 (1984).

    Article  CAS  PubMed  Google Scholar 

  33. Ramoa, A. S., Paradiso, M. A. & Freeman, R. D. Blockade of intracortical inhibition in kitten striate cortex: effects on receptive field properties and associated loss of ocular dominance plasticity. Exp. Brain Res. 73, 285–296 (1988).

    Article  CAS  PubMed  Google Scholar 

  34. Reiter, H. O., Waitzman, D. M. & Stryker, M. P. Cortical activity blockade prevents ocular dominance plasticity in the kitten visual cortex. Exp. Brain Res. 65, 182–188 (1986).

    Article  CAS  PubMed  Google Scholar 

  35. Bear, M. F., Kleinschmidt, A., Gu, Q. A. & Singer, W. Disruption of experience-dependent synaptic modifications in striate cortex by infusion of an NMDA receptor antagonist. J. Neurosci. 10, 909–925 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Reiter, H. O. & Stryker, M. P. Neural plasticity without postsynaptic action potentials: less-active inputs become dominant when kitten visual cortical cells are pharmacologically inhibited. Proc. Natl Acad. Sci. USA 85, 3623–3627 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Hata, Y. & Stryker, M. P. Control of thalamocortical afferent rearrangement by postsynaptic activity in developing visual cortex. Science 265, 1732–1735 (1994).

    Article  CAS  PubMed  Google Scholar 

  38. Gordon, J. A. & Stryker, M. P. Experience-dependent plasticity of binocular responses in the primary visual cortex of the mouse. J. Neurosci. 16, 3274–3286 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Prusky, G. T. & Douglas, R. M. Developmental plasticity of mouse visual acuity. Eur. J. Neurosci. 17, 167–173 (2003). A behavioural study revealing a clear critical period for amblyopia that matches the shift of single-unit responses in the mouse visual cortex (detailed in reference 38), which indicates that there are no lasting consequence of sub-threshold changes reported with evoked potentials or immediate early gene expression (discussed further in references 172–174).

    Article  PubMed  Google Scholar 

  40. Soghomonian, J. J. & Martin, D. L. Two isoforms of glutamate decarboxylase: why? Trends Pharmacol. 19, 500–505 (1998).

    Article  CAS  Google Scholar 

  41. Asada, H. et al. Cleft palate and decreased brain γ-aminobutyric acid in mice lacking the 67-kDa isoform of glutamic acid decarboxylase. Proc. Natl Acad. Sci. USA 94, 6496–6499 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Tian, N. et al. The role of the synthetic enzyme GAD65 in the control of neuronal γ-aminobutyric acid release. Proc. Natl Acad. Sci. USA 96, 12911–12916 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hensch, T. K. et al. Local GABA circuit control of experience-dependent plasticity in developing visual cortex. Science 282, 1504–1508 (1998). The first evidence that GABA-mediated transmission is required for plasticity in vivo . Lays the foundation for a series of studies confirming that critical period timing can be controlled through inhibitory interneurons (discussed further in references 44–48), quite unlike LTP models in vitro that are routinely blocked by inhibition (see references 105, 121).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Fagiolini, M. & Hensch, T. K. Inhibitory threshold for critical-period activation in primary visual cortex. Nature 404, 183–186 (2000).

    Article  CAS  PubMed  Google Scholar 

  45. Iwai, Y., Fagiolini, M., Obata, K. & Hensch, T. K. Rapid critical period induction by tonic inhibition in visual cortex. J. Neurosci. 23, 6695–6702 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Fagiolini, M. et al. Specific GABAA circuits for visual cortical plasticity. Science 303, 1681–1683 (2004). Provides striking evidence that not all GABA-mediated connections are involved in critical period induction, which has strong implications for computational models and the design of specific benzodiazepines for use in human infants.

    Article  CAS  PubMed  Google Scholar 

  47. Huang, Z. J. et al. BDNF regulates the maturation of inhibition and the critical period of plasticity in mouse visual cortex. Cell 98, 739–755 (1999).

    Article  CAS  PubMed  Google Scholar 

  48. Hanover, J. L., Huang, Z. J., Tonegawa, S. & Stryker, M. P. Brain-derived neurotrophic factor overexpression induces precocious critical period in mouse visual cortex. J. Neurosci. 19, RC40 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Castren, E., Zafra, F., Thoenen, H. & Lindholm, D. Light regulates expression of brain-derived neurotrophic factor mRNA in rat visual cortex. Proc. Natl Acad. Sci. USA 89, 9444–9448 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Morales, B., Choi, S. Y. & Kirkwood, A. Dark rearing alters the development of GABAergic transmission in visual cortex. J. Neurosci. 22, 8084–8090 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, L., Yang, C. & Mower, G. D. Developmental changes in the expression of GABAA receptor subunits (α1, α2, α3) in the cat visual cortex and the effects of dark rearing. Mol. Brain Res. 88, 135–143 (2001).

    Article  CAS  PubMed  Google Scholar 

  52. Mower, G. D. The effect of dark rearing on the time course of the critical period in cat visual cortex. Dev. Brain Res. 58, 151–158 (1991).

    Article  CAS  Google Scholar 

  53. Fagiolini, M. et al. Separable features of visual cortical plasticity revealed by N-methyl-D-aspartate receptor 2A signaling. Proc. Natl Acad. Sci. USA 100, 2854–2859 (2003). Debunks the 'NR2A subunit switch' hypothesis for critical period closure (see also reference 101). Also reveals distinct molecular pathways for individual receptive field properties.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Gianfranceschi, L. et al. Visual cortex is rescued from the effects of dark rearing by overexpression of BDNF. Proc. Natl Acad. Sci. USA 100, 12486–12491 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Bartoletti, A., Medini, P., Berardi, N. & Maffei, L. Environmental enrichment prevents effects of dark-rearing in the rat visual cortex. Nature Neurosci. 7, 215–216 (2004). References 54 and 55, along with reference 45, strikingly demonstrate that direct modulation of tonic GABA-mediated function in the cortex is sufficient to trigger the critical period, even in the absence of visual input.

    Article  CAS  PubMed  Google Scholar 

  56. Kawaguchi, Y. & Kubota, Y. GABAergic cell subtypes and their synaptic connections in rat frontal cortex. Cereb. Cortex 7, 476–486 (1997).

    Article  CAS  PubMed  Google Scholar 

  57. Markram, H. et al. Interneurons of the neocortical inhibitory system. Nature Rev. Neurosci. 5, 793–807 (2004).

    Article  CAS  Google Scholar 

  58. DeFelipe, J. Types of neurons, synaptic connections and chemical characteristics of cells immunoreactive for calbindin-D28K, parvalbumin and calretinin in the neocortex. J. Chem. Neuroanat. 14, 1–19 (1997).

    Article  CAS  PubMed  Google Scholar 

  59. Del Rio, J. A., De Lecea, L., Ferrer, I. & Soriano, E. The development of parvalbumin-immunoreactivity in the neocortex of the mouse. Dev. Brain Res. 81, 247–259 (1994).

    Article  CAS  Google Scholar 

  60. Rudy, B. & McBain, C. J. Kv3 channels: voltage-gated K+ channels designed for high-frequency repetitive firing. Trends Neurosci. 24, 517–526 (2001).

    Article  CAS  PubMed  Google Scholar 

  61. Erisir, A., Lau, D., Rudy, B. & Leonard, C. S. Function of specific K+ channels in sustained high-frequency firing of fast-spiking neocortical interneurons. J. Neurophysiol. 82, 2476–2489 (1999).

    Article  CAS  PubMed  Google Scholar 

  62. Lien, C. C. & Jonas, P. Kv3 potassium conductance is necessary and kinetically optimized for high-frequency action potential generation in hippocampal interneurons. J. Neurosci. 23, 2058–2068 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Cherubini, E. & Conti, F. Generating diversity at GABAergic synapses. Trends Neurosci. 24, 155–162 (2001).

    Article  CAS  PubMed  Google Scholar 

  64. Rudolph, U., Crestani, F. & Möhler, H. GABAA receptor subtypes: dissecting their pharmacological functions. Trends Pharmacol. Sci. 22, 188–194 (2001).

    Article  CAS  PubMed  Google Scholar 

  65. Di Cristo, G. et al. Subcellular domain-restricted GABAergic innervation in primary visual cortex in the absence of sensory and thalamic inputs. Nature Neurosci. 7, 1184–1186 (2004).

    Article  CAS  PubMed  Google Scholar 

  66. Chattopadhyaya, B. et al. Experience and activity-dependent maturation of perisomatic GABAergic innervation in primary visual cortex during a postnatal critical period. J. Neurosci. 24, 9598–9611 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Buzas, P., Eysel, U. T., Adorjan, P. & Kisvarday, Z. F. Axonal topography of cortical basket cells in relation to orientation, direction, and ocular dominance maps. J. Comp. Neurol. 437, 259–285 (2001).

    Article  CAS  PubMed  Google Scholar 

  68. Klausberger, T., Roberts, J. D. & Somogyi, P. Cell type- and input-specific differences in the number and subtypes of synaptic GABAA receptors in the hippocampus. J. Neurosci. 22, 2513–2521 (2002). Demonstrates remarkable subcellular sorting of individual GABA A receptor subtypes to receive distinct inhibitory inputs based on α subunit composition.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Nusser, Z., Sieghart, W., Benke, D., Fritschy, J. M. & Somogyi, P. Differential synaptic localization of two major γ-aminobutyric acid type A receptor a subunits on hippocampal pyramidal cells. Proc. Natl Acad. Sci. USA 93, 11939–11944 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Härtig, W. et al. Cortical neurons immunoreactive for the potassium channel Kv3.1b subunit are predominantly surrounded by perineuronal nets presumed as a buffering system for cations. Brain Res. 842, 15–29 (1999).

    Article  PubMed  Google Scholar 

  71. Saghatelyan, A. K. et al. Reduced perisomatic inhibition, increased excitatory transmission, and impaired long-term potentiation in mice deficient for the extra-cellular matrix glycoprotein tenascin-R. Mol. Cell. Neurosci. 17, 226–240 (2001).

    Article  CAS  PubMed  Google Scholar 

  72. Pizzorusso, T. et al. Reactivation of ocular dominance plasticity in the adult visual cortex. Science 298, 1248–1251 (2002). The holy grail of critical period studies that showed it is possible to reactivate plasticity in adulthood, achieved by disrupting ECM components of perineuronal nets with chondroitinases. These structures preferentially surround and influence the function of large parvalbumin-positive basket cells (see also references 70 and 71).

    Article  CAS  PubMed  Google Scholar 

  73. Berardi, N., Pizzorusso, T. & Maffei, L. Extracellular matrix and visual cortical plasticity; freeing the synapse. Neuron 44, 905–908 (2004).

    CAS  PubMed  Google Scholar 

  74. Liu, Y., Fields, R. D., Fitzgerald, S., Festoff, B. W. & Nelson, P. G. Proteolytic activity, synapse elimination, and the Hebb synapse. J. Neurobiol. 25, 325–335 (1994).

    Article  CAS  PubMed  Google Scholar 

  75. Shiosaka, S. & Yoshida, S. Synaptic microenvironments — structural plasticity, adhesion molecules, proteases and their inhibitors. Neurosci. Res. 37, 85–89 (2000).

    Article  CAS  PubMed  Google Scholar 

  76. Mataga, N. & Hensch, T. K. in Proteases in Biology and Disease Vol. 3 (eds Lendeckel, U. & Hooper, N.) Chapter 11 (Kluwer/Plenum, New York, in the press).

  77. Qian, Z., Gilbert, M. E., Colicos, M. A., Kandel, E. R. & Kuhl, D. Tissue-plasminogen activator is induced as an immediate-early gene during seizure, kindling and long-term potentiation. Nature 361, 453–457 (1993).

    Article  CAS  PubMed  Google Scholar 

  78. Mataga, N., Nagai, N. & Hensch, T. K. Permissive proteolytic activity for visual cortical plasticity. Proc. Natl Acad. Sci. USA 99, 7717–7721 (2002). Definitive evidence that proteases are required for ocular dominance plasticity through the use of gene-targeted animals, supporting earlier pharmacological studies in cats (detailed in references 79 and 80).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Mataga, N. et al. Enhancement of mRNA expression of tissue-type plasminogen activator by L-threo-3,4-dihydrophenylserine in association with ocular dominance plasticity. Neurosci. Lett. 218, 149–152 (1996).

    Article  CAS  PubMed  Google Scholar 

  80. Müller, C. M. & Griesinger, C. B. Tissue plasminogen activator mediates reverse occlusion plasticity in visual cortex. Nature Neurosci. 1, 47–53 (1998).

    Article  PubMed  Google Scholar 

  81. Berardi, N., Pizzorusso, T., Ratto, G. M. & Maffei, L. Molecular basis of plasticity in the visual cortex. Trends Neurosci. 26, 369–378 (2003).

    Article  CAS  PubMed  Google Scholar 

  82. Silver, M. A. & Stryker, M. P. Synaptic density in geniculocortical afferents remains constant after monocular deprivation in the cat. J. Neurosci. 19, 10829–10842 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Grutzendler, J., Kasthuri, N. & Gan, W. B. Long-term dendritic spine stability in the adult cortex. Nature 420, 812–816 (2002).

    Article  CAS  PubMed  Google Scholar 

  84. Majewska, A. & Sur, M. Motility of dendritic spines in visual cortex in vivo: changes during the critical period and effects of visual deprivation. Proc. Natl Acad. Sci. USA 100, 16024–16029 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Oray, S., Majewska, A. & Sur, M. Dendritic spine dynamics are regulated by monocular deprivation and extracellular matrix degradation. Neuron 44, 1021–1030 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Mataga, N., Mizuguchi, Y. & Hensch, T. K. Experience-dependent pruning of dendritic spines in visual cortex by tissue plasminogen activator. Neuron 44, 1031–1041 (2004). References 85 and 86 reveal the first anatomical events that occur on monocular deprivation. Spine motility followed by transient pruning faithfully reflects competition during the critical period and is mediated by tPA–plasmin in vivo.

    Article  CAS  PubMed  Google Scholar 

  87. Taha, S. & Stryker, M. P. Rapid ocular dominance plasticity requires cortical but not geniculate protein synthesis. Neuron 34, 425–436 (2002).

    Article  CAS  PubMed  Google Scholar 

  88. Pang, P. et al. Cleavage of ProBDNF by tPA/plasmin is essential for long-term hippocampal plasticity. Science 306, 487–491 (2004).

    Article  CAS  PubMed  Google Scholar 

  89. Tyler, W. J. & Pozzo-Miller, L. Miniature synaptic transmission and BDNF modulates dendritic spine growth and form in rat CA1 neurons. J. Physiol. (Lond.) 553, 497–509 (2004).

    Article  CAS  Google Scholar 

  90. Miller, K. D. Synaptic economics: competition and cooperation in synaptic plasticity. Neuron 17, 371–374 (1996). Careful consideration of the theory and cellular events behind the competitive nature of critical period plasticity in the visual cortex.

    Article  CAS  PubMed  Google Scholar 

  91. Barry, M. F. & Ziff, E. B. Receptor trafficking and the plasticity of excitatory synapses. Curr. Opin. Neurobiol. 12, 279–286 (2002).

    Article  CAS  PubMed  Google Scholar 

  92. Takahashi, T., Svoboda, K. & Malinow, R. Experience strengthening transmission by driving AMPA receptors into synapses. Science 299, 1585–1588 (2003).

    Article  CAS  PubMed  Google Scholar 

  93. Allen, C. B., Celikel, T. & Feldman, D. E. Long-term depression induced by sensory deprivation during cortical map plasticity in vivo. Nature Neurosci. 6, 291–299 (2003).

    Article  CAS  PubMed  Google Scholar 

  94. Heynen, A. J. et al. Molecular mechanism for loss of visual cortical responsiveness following brief monocular deprivation. Nature Neurosci. 6, 854–862 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Flint, A. C., Maisch, U. S., Weishaupt, J. H., Kriegstein, A. R. & Monyer, H. NR2A subunit expression shortens NMDA receptor synaptic currents in developing neocortex. J. Neurosci. 17, 2469–2476 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Nase, G., Weishaupt, J., Stern, P., Singer, W. & Monyer, H. Genetic and epigenetic regulation of NMDA receptor expression in the rat visual cortex. Eur. J. Neurosci. 11, 4320–4326 (1999).

    Article  CAS  PubMed  Google Scholar 

  97. Quinlan, E. M., Philpot, B. D., Huganir, R. L. & Bear, M. F. Rapid, experience-dependent expression of synaptic NMDA receptors in visual cortex in vivo. Nature Neurosci. 2, 352–357 (1999).

    Article  CAS  PubMed  Google Scholar 

  98. Philpot, B. D., Sekhar, A. K., Shouval, H. Z. & Bear, M. F. Visual experience and deprivation bidirectionally modify the composition and function of NMDA receptors in visual cortex. Neuron 29, 157–169 (2001).

    Article  CAS  PubMed  Google Scholar 

  99. Tang, Y. P. et al. Genetic enhancement of learning and memory in mice. Nature 401, 63–69 (1999).

    Article  CAS  PubMed  Google Scholar 

  100. Liu, L. et al. Role of NMDA receptor subtypes in governing the direction of hippocampal synaptic plasticity. Science 304, 1021–1024 (2004).

    Article  CAS  PubMed  Google Scholar 

  101. Lu, H. C., Gonzalez, E. & Crair, M. C. Barrel cortex critical period plasticity is independent of changes in NMDA receptor subunit composition. Neuron 32, 619–634 (2001).

    Article  PubMed  Google Scholar 

  102. Barth, A. L. & Malenka, R. C. NMDAR EPSC kinetics do not regulate the critical period for LTP at thalamocortical synapses. Nature Neurosci. 4, 235–236 (2001).

    Article  CAS  PubMed  Google Scholar 

  103. Datwani, A., Iwasato, T., Itohara, S. & Erzurumlu, R. S. Lesion-induced thalamocortical axonal plasticity in the S1 cortex is independent of NMDA receptor function in excitatory cortical neurons. J. Neurosci. 22, 9171–9175 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Rebsam, A., Seif, I. & Gaspar, P. Dissociating barrel development and plasticity in the mouse somatosensory cortex. J. Neurosci. 25, 706–710 (2005). Clever use of the monoamine oxidise A-knockout mouse, whose barrel formation can be rescued by lowering serotonin levels with parachlorophenylalanine (PCPA) at ages beyond the critical period for barrel plasticity. Closure of the whisker cautery effect on the barrelfield is independent and probably not determined at the cortical level.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Feldman, D. E. Inhibition and plasticity. Nature Neurosci. 3, 303–304 (2000).

    Article  CAS  PubMed  Google Scholar 

  106. Dudai, Y. Molecular bases of long-term memories: a question of persistence. Curr. Opin. Neurobiol. 12, 211–216 (2002).

    Article  CAS  PubMed  Google Scholar 

  107. O'Connor, D. H., Wittenberg, G. M. & Wang, S. S. -H. Graded bidirectional synaptic plasticity is composed of switch-like unitary events. Proc. Natl Acad. Sci. USA 102, 9679–9684 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Sajikumar, S., Navakkode, S. & Frey, J. U. Protein synthesis-dependent long-term functional plasticity: methods and techniques. Curr. Opin. Neurobiol. (in the press).

  109. Jones, M. W. et al. A requirement for the immediate early gene Zif268 in the expression of late LTP and long-term memories. Nature Neurosci. 4, 289–296 (2001).

    Article  CAS  PubMed  Google Scholar 

  110. Mataga, N., Fujishima, S., Condie, B. G. & Hensch, T. K. Experience-dependent plasticity of mouse visual cortex in the absence of the neuronal activity-dependent marker egr1/zif268. J. Neurosci. 21, 9724–9732 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Baranes, D. et al. Tissue plasminogen activator contributes to the late phase of LTP and to synaptic growth in the hippocampal mossy fiber pathway. Neuron 21, 813–825 (1998).

    Article  CAS  PubMed  Google Scholar 

  112. Neuhoff, H., Roeper, J. & Schweizer, M. Activity-dependent formation of perforated synapses in cultured hippocampal neurons. Eur. J. Neurosci. 11, 4241–4250 (1999).

    Article  CAS  PubMed  Google Scholar 

  113. Daw, N. in The Visual Neurosciences Vol. 1 (eds Chalupa, L. & Werner, J. S.) 126–145 (MIT Press, Cambridge, Massachusetts, USA, 2004).

    Google Scholar 

  114. Hensch, T. K. Controlling the critical period. Neurosci. Res. 47, 17–22 (2003).

    Article  PubMed  Google Scholar 

  115. Renger, J. J. et al. Experience-dependent plasticity without long-term depression by type 2 metabotropic glutamate receptors in developing visual cortex. Proc. Natl Acad. Sci. USA 99, 1041–1046 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Bartoletti, A. et al. Heterozygous knock-out mice for brain-derived neurotrophic factor show a pathway-specific impairment of long-term potentiation but normal critical period for monocular deprivation. J. Neurosci. 22, 10072–10077 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Daw, N., Rao, Y., Wang, X. F., Fischer, Q. & Yang, Y. LTP and LTD vary with layer in rodent visual cortex. Vision Res. 44, 3377–3380 (2004).

    Article  PubMed  Google Scholar 

  118. Zhou, Q., Homma, K. J. & Poo, M. M. Shrinkage of dendritic spines associated with long-term depression of hippocampal synapses. Neuron 44, 749–757 (2004). Demonstrates that spine shape changes can occur on a fast time scale, but independent of mechanisms that underlie the LTD of synaptic transmission.

    Article  CAS  PubMed  Google Scholar 

  119. Hayashi, Y. & Majewska, A. K. Dendritic spine geometry: functional implication and regulation. Neuron 46, 529–532 (2005).

    Article  CAS  PubMed  Google Scholar 

  120. Yang, Y. et al. Reversible blockade of experience-dependent plasticity by calcineurin in mouse visual cortex. Nature Neurosci. 8, 791–796 (2005).

    Article  CAS  PubMed  Google Scholar 

  121. Wan, H. et al. Benzodiazepine impairment of perirhinal cortical plasticity and recognition memory. Eur. J. Neurosci. 20, 2214–2224 (2004).

    Article  CAS  PubMed  Google Scholar 

  122. Jiang, B., Akaneya, Y., Hata, Y. & Tsumoto, T. Long-term depression is not induced by low-frequency stimulation in rat visual cortex in vivo: a possible preventing role of endogenous brain-derived neurotrophic factor. J. Neurosci. 23, 3761–3770 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Bi, G. & Poo, M. Synaptic modification by correlated activity: Hebb's postulate revisited. Annu. Rev. Neurosci. 24, 139–166 (2001).

    Article  CAS  PubMed  Google Scholar 

  124. Song, S., Miller, K. D. & Abbott, L. F. Competitive Hebbian learning through spike-timing-dependent synaptic plasticity. Nature Neurosci. 3, 919–926 (2000).

    Article  CAS  PubMed  Google Scholar 

  125. Pouille, F. & Scanziani, M. Enforcement of temporal fidelity in pyramidal cells by somatic feed-forward inhibition. Science 293, 1159–1163 (2001).

    Article  CAS  PubMed  Google Scholar 

  126. Sanes, J. R. & Lichtman, J. W. Can molecules explain long-term potentiation? Nature Neurosci. 2, 597–604 (1999).

    Article  CAS  PubMed  Google Scholar 

  127. Zhu, Y., Stornetta, R. L. & Zhu, J. J. Chandelier cells control excessive cortical excitation: characteristics of whisker-evoked synaptic responses of layer 2/3 nonpyramidal and pyramidal neurons. J. Neurosci. 24, 5101–5108 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Galarreta, M. & Hestrin, S. Electrical synapses between GABA-releasing interneurons. Nature Rev. Neurosci. 2, 425–433 (2001).

    Article  CAS  Google Scholar 

  129. Meyer, A. H., Katona, I., Blatow, M., Rozov, A. & Monyer, H. In vivo labeling of parvalbumin-positive interneurons and analysis of electrical coupling in identified neurons. J. Neurosci. 22, 7055–7064 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Galarreta, M. & Hestrin, S. Spike transmission and synchrony detection in networks of GABAergic interneurons. Science 292, 2295–2299 (2001). Paired recordings from electrically-coupled, parvalbumin-positive cells reveals a novel property of such networks, namely synchrony detection, which could operate on a columnar scale.

    Article  CAS  PubMed  Google Scholar 

  131. Gao, B. & Fritschy, J. M. Selective allocation of GABAA receptors containing the α 1 subunit to neurochemically distinct subpopulations of rat hippocampal interneurons. Eur. J. Neurosci. 6, 837–853 (1994).

    Article  CAS  PubMed  Google Scholar 

  132. Deans, M. R., Gibson, J. R., Sellitto, C., Connors, B. W. & Paul, D. L. Synchronous activity of inhibitory networks in neocortex requires electrical synapses containing connexin36. Neuron 31, 477–485 (2001).

    Article  CAS  PubMed  Google Scholar 

  133. Hormuzdi, S. G. et al. Impaired electrical signaling disrupts gamma frequency oscillations in connexin 36-deficient mice. Neuron 31, 487–495 (2001).

    Article  CAS  PubMed  Google Scholar 

  134. Guldenagel, M. et al. Expression patterns of connexin genes in mouse retina. J. Comp. Neurol. 425, 193–201 (2000).

    Article  CAS  PubMed  Google Scholar 

  135. Dityatev, A. & Schachner, M. Extracellular matrix molecules and synaptic plasticity. Nature Rev. Neurosci. 4, 456–468 (2003).

    Article  CAS  Google Scholar 

  136. Lochner, J. E. et al. Real-time imaging of the axonal transport of granules containing a tissue plasminogen activator/green fluorescent protein hybrid. Mol. Biol. Cell 9, 2463–2476 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Gualandris, A., Jones, T. E., Strickland, S. & Tsirka, S. E. Membrane depolarization induces calcium-dependent secretion of tissue plasminogen activator. J. Neurosci. 16, 2220–2225 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Parmer, R. J. et al. Tissue plasminogen activator (tPA) is targeted to the regulated secretory pathway. J. Biol. Chem. 272, 1976–1982 (1997).

    Article  CAS  PubMed  Google Scholar 

  139. Collin, T. et al. Developmental changes in parvalbumin regulate presynaptic Ca2+ signaling. J. Neurosci. 25, 96–107 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Murase, S., Mosser, E. & Schuman, E. M. Depolarization drives β-catenin into neuronal spines promoting changes in synaptic structure and function. Neuron 35, 91–105 (2002).

    Article  CAS  PubMed  Google Scholar 

  141. Tanaka, H. et al. Molecular modification of N-cadherin in response to synaptic activity. Neuron 25, 93–107 (2000). Strongly suggests that synaptic adhesion is locally controlled and dynamically modulated by neuronal activity through the acquisition of protease resistance by dimerization of N-cadherin on depolarization.

    Article  CAS  PubMed  Google Scholar 

  142. Wannier-Morino, P., Rager, G., Sonderegger, P. & Grabs, D. Expression of neuroserpin in the visual cortex of the mouse during the developmental critical period. Eur. J. Neurosci. 17, 1853–1860 (2003).

    Article  PubMed  Google Scholar 

  143. Sakaguchi, H. Sex differences in the developmental changes of GABAergic neurons in zebra finch song control nuclei. Exp. Brain Res. 108, 62–68 (1996). Intriguing evidence that the motor phase of the zebra finch critical period for song acquisition reflects maturation of GABA-containing cells in male nucleus RA.

    Article  CAS  PubMed  Google Scholar 

  144. Freund, T. F., Katona, I. & Piomelli, D. Role of endogenous cannabinoids in synaptic signaling. Physiol. Rev. 83, 1017–1066 (2003).

    Article  CAS  PubMed  Google Scholar 

  145. Soderstrom, K. & Johnson, F. Cannabinoid exposure alters learning of zebra finch vocal patterns. Brain Res. 142, 215–217 (2003).

    Article  CAS  Google Scholar 

  146. Knudsen, E. I., Zheng, W. & DeBello, W. M. Traces of learning in the auditory localization pathway. Proc. Natl Acad. Sci. USA 97, 11815–11820 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  147. Knudsen, E. I. Capacity for plasticity in the adult owl auditory system expanded by juvenile experience. Science 279, 1531–1533 (1998). Demonstrates the power of the critical period in laying down multiple neural representations of early experienced environments that can be reactivated when re-encountered later in life. Active suppression by newly formed GABA circuits prevents confusion among multiple maps (see reference 148).

    Article  CAS  PubMed  Google Scholar 

  148. Zheng, W. & Knudsen, E. I. Functional selection of adaptive auditory space map by GABAA-mediated inhibition. Science 284, 962–965 (1999).

    Article  CAS  PubMed  Google Scholar 

  149. Linkenhoker, B. A. & Knudsen, E. I. Incremental training increases the plasticity of the auditory space map in adult barn owls. Nature 419, 293–296 (2002). Suggests a new strategy for adult plasticity (without drugs) based on incremental training to cumulatively overcome the anatomical constraints established during the critical period.

    Article  CAS  PubMed  Google Scholar 

  150. Linkenhoker, B. A., von der Ohe, C. G. & Knudsen, E. I. Anatomical traces of juvenile learning in the auditory system of adult barn owls. Nature Neurosci. 8, 93–98 (2005).

    Article  CAS  PubMed  Google Scholar 

  151. Fuchs, J. L. & Salazar, E. Effects of whisker trimming on GABAA receptor binding in the barrel cortex of developing and adult rats. J. Comp. Neurol. 395, 209–216 (1998).

    Article  CAS  PubMed  Google Scholar 

  152. Micheva, K. D. & Beaulieu, C. An anatomical substrate for experience-dependent plasticity of the rat barrel field cortex. Proc. Natl Acad. Sci. USA 92, 11834–11838 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Knott, G. W., Quairiaux, C., Genoud, C. & Welker, E. Formation of dendritic spines with GABAergic synapses induced by whisker stimulation in adult mice. Neuron 34, 265–273 (2002). Demonstrates plasticity of GABA-mediated connections in the adult barrel cortex.

    Article  CAS  PubMed  Google Scholar 

  154. Diamond, M. E., Armstrong-James, M. & Ebner, F. F. Experience-dependent plasticity in adult rat barrel cortex. Proc. Natl Acad. Sci. USA 90, 2082–2086 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Gheusi, G. et al. Importance of newly generated neurons in the adult olfactory bulb for odor discrimination. Proc. Natl Acad. Sci. USA 97, 1823–1828 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Cecchi, G. A., Petreanu, L. T., Alvarez-Buylla, A. & Magnasco, M. O. Unsupervised learning and adaptation in a model of adult neurogenesis. J. Comput. Neurosci. 11, 175–182 (2001).

    Article  CAS  PubMed  Google Scholar 

  157. Yokoi, M., Mori, K. & Nakanishi, S. Refinement of odor molecule tuning by dendro-dendritic synaptic inhibition in the olfactory bulb. Proc. Natl Acad. Sci. USA 92, 3371–3375 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Lagier, S., Carleton, A. & Lledo, P. M. Interplay between local GABA-mediated interneurons and relay neurons generates gamma oscillations in the rat olfactory bulb. J. Neurosci. 24, 4382–4392 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Murphy, K. M., Beston, B. R., Boley, P. M. & Jones, D. G. Development of human visual cortex: a balance between excitatory and inhibitory plasticity mechanisms. Dev. Psychobiol. 46, 209–221 (2005). Crucial evidence from human autopsy samples showing slow maturation of GAD65 and GABA A receptor α1-subunits in the visual cortex that better matches the prolonged critical period for binocular vision in this species (see references 6 and 7) than does NMDA-receptor subunit switching (for further information, see references 43, 46 and 53).

    Article  CAS  PubMed  Google Scholar 

  160. De Negri, M., Baglietto, M. G. & Biancheri, R. Electrical status epilepticus in childhood: treatment with short cycles of high dosage benzodiazepine. Brain Dev. 15, 311–312 (1993).

    Article  CAS  PubMed  Google Scholar 

  161. Rubenstein, J. L. & Merzenich, M. M. Model of autism: increased ratio of excitation/inhibition in key neural systems. Genes Brain Behav. 2, 255–267 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  162. Möhler, H., Fritschy, J. M., Crestani, F., Hensch, T. & Rudolph, U. Specific GABAA circuits in brain development and therapy. Biochem. Pharmacol. 68, 1685–1690 (2004).

    Article  PubMed  CAS  Google Scholar 

  163. Arckens, L. et al. Cooperative changes in GABA, glutamate and activity levels: the missing link in cortical plasticity. Eur. J. Neurosci. 12, 4222–4232 (2000).

    Article  CAS  PubMed  Google Scholar 

  164. Lodder, J., Luijckx, G., van Raak, L. & Kessels, F. Diazepam treatment to increase the cerebral GABAergic activity in acute stroke: a feasibility study in 104 patients. Cerebrovasc. Dis. 10, 437–440 (2000).

    Article  CAS  PubMed  Google Scholar 

  165. Dani, V. S. et al. Reduced cortical activity due to a shift in the balance between excitation and inhibition in a mouse model of Rett syndrome. Proc. Natl Acad. Sci. USA 102, 12560–12565 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Kalanithi, P. S. A. et al. Altered parvalbumin-positive neuron distribution in basal ganglia of individuals with Tourette syndrome. Proc. Natl Acad. Sci. USA 102, 13307–13312 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Lewis, D. A., Hashimoto, T. & Volk, D. W. Cortical inhibitory neurons and schizophrenia. Nature Rev. Neurosci. 6, 312–324 (2005).

    Article  CAS  Google Scholar 

  168. Paulsen, O. & Moser, E. I. A model of hippocampal memory encoding and retrieval: GABAergic control of synaptic plasticity. Trends Neurosci. 21, 273–378 (1998).

    Article  CAS  PubMed  Google Scholar 

  169. Miyamoto, H., Katagiri, H. & Hensch, T. Experience-dependent slow-wave sleep development. Nature Neurosci. 6, 553–554 (2003).

    Article  CAS  PubMed  Google Scholar 

  170. Prasad, S. S. et al. Gene expression patterns during enhanced periods of visual cortex plasticity. Neuroscience 111, 35–45 (2002).

    Article  CAS  PubMed  Google Scholar 

  171. Ossipow, V., Pellissier, F., Schaad, O. & Ballivet, M. Gene expression analysis of the critical period in the visual cortex. Mol. Cell. Neurosci. 27, 70–83 (2004).

    Article  CAS  PubMed  Google Scholar 

  172. Sawtell, N. B. et al. NMDA receptor-dependent ocular dominance plasticity in adult visual cortex. Neuron 38, 977–985 (2003). Erratum Neuron 39, 727 (2003).

    Article  CAS  PubMed  Google Scholar 

  173. Pham, T. A. et al. A semi-persistent adult ocular dominance plasticity in visual cortex is stabilized by activated CREB. Learn. Mem. 11, 738–747 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  174. Tagawa, Y., Kanold, P. O., Majdan, M. & Shatz, C. J. Multiple periods of functional ocular dominance plasticity in mouse visual cortex. Nature Neurosci. 8, 380–388 (2005).

    Article  CAS  PubMed  Google Scholar 

  175. Fagiolini, M., Pizzorusso, T., Berardi, N., Domenici, L. & Maffei, L. Functional postnatal development of the rat primary visual cortex and the role of visual experience: dark rearing and monocular deprivation. Vision Res. 34, 709–720 (1994).

    Article  CAS  PubMed  Google Scholar 

  176. Wang, X., Merzenich, M. M., Sameshima, K. & Jenkins, W. M. Remodelling of hand representation in adult cortex determined by timing of tactile stimulation. Nature 378, 71–75 (1995).

    Article  CAS  PubMed  Google Scholar 

  177. Kilgard, M. P. & Merzenich, M. M. Cortical map reorganization enabled by nucleus basalis activity. Science 279, 1714–1718 (1998).

    Article  CAS  PubMed  Google Scholar 

  178. Bao, S., Chan, V. T. & Merzenich, M. M. Cortical remodelling induced by activity of ventral tegmental dopamine neurons. Nature 412, 79–83 (2001). Two studies that demonstrate the potential for neuromodulatory systems (attention) to restore near critical period levels of plasticity to the adult brain.

    Article  CAS  PubMed  Google Scholar 

  179. Miyamoto, H. & Hensch, T. K. Reciprocal interaction of sleep and synaptic plasticity. Mol. Interv. 3, 404–417 (2003).

    Article  PubMed  Google Scholar 

  180. Hinkle, D. J. & Macdonald, R. L. β subunit phosphorylation selectively increases fast desensitization and prolongs deactivation of α1β1γ2L and α1β3γ2L GABAA receptor currents. J. Neurosci. 23, 11698–11710 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Fischer, Q. S. et al. Requirement for the RIIβ isoform of PKA, but not calcium-stimulated adenylyl cyclase, in visual cortical plasticity. J. Neurosci. 24, 9049–9058 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. McGee, A. et al. Experience-driven plasticity of visual cortex limited by myelin and Nogo receptor. Science 309, 2222–2226 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Mechelli, A. et al. Neurolinguistics: structural plasticity in the bilingual brain. Nature 431, 757 (2004).

    Article  CAS  PubMed  Google Scholar 

  184. Bengtsson, S. L. et al. Extensive piano practicing has regionally specific effects on white matter development. Nature Neurosci. 8, 1148–1150 (2005).

    Article  CAS  PubMed  Google Scholar 

  185. Fiumelli, H., Jabaudon, D., Magistretti, P. J. & Martin, J. -L. BDNF stimulates expression, activity and release of tissue-type plasminogen activator in mouse cortical neurons. Eur. J. Neurosci. 11, 1639–1646 (1999).

    Article  CAS  PubMed  Google Scholar 

  186. Lewis, T. L. & Maurer, D. Multiple sensitive periods in human visual development: evidence from visually deprived children. Dev. Psychobiol. 46, 163–183 (2005).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

I thank M. Fagiolini for comments, N. Mataga for immunostaining in Figure 5b and all members of the lab for Neuronal Circuit Development (RIKEN BSI) for their dedication to understanding critical period mechanisms.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Related links

Related links

DATABASES

Entrez Gene

BDNF

Gad65

Gad67

Zif268

OMIM

Rett syndrome

Tourette's syndrome

FURTHER INFORMATION

RIKEN Brain Science Institute

Glossary

CRITICAL PERIOD

A strict time window during which experience provides information that is essential for normal development and permanently alters performance.

SENSITIVE PERIOD

A limited time during development, during which the effect of experience on brain function is particularly strong.

AMBLYOPIA

Poor vision through an eye that is otherwise physically healthy due to little or no transmission of the visual image to the brain through circuits that are hard-wired during a developmental critical period. It affects 2–5% of the population.

OCULAR DOMINANCE

Relative anatomical or physiological strength of connections from either eye to individual cells in the primary visual cortex.

THALAMOCORTICAL AFFERENTS

Axons from the thalamus (for example, the dLGN) that relay sensory input from the periphery (for example, the retina) to layer 4 of the neocortex.

BENZODIAZEPINES

Modulate chloride flux through GABAA receptors that contain the γ2 and any combination of α1, α2, α3 or α5 subunits. Benzodiazepine agonists enhance and inverse agonists decrease GABA efficacy.

STRABISMUS

Deviation of the two eyes due to a weakening of extraocular musculature that results in either an inward (esotropic) or outward (exotropic) rotation of one orbit and consequent amblyopia.

TETRODOTOXIN

(TTX). A voltage-dependent sodium channel blocker that can be used to silence all neural activity except spontaneous neurotransmitter release.

MONOCULAR DEPRIVATION

(MD). Imbalanced visual input due to the occlusion of one eye by patching, eyelid suture or intraocular TTX injection.

PARVALBUMIN

One of three calcium-binding proteins, which, together with calretinin and calbindin, are expressed in most GABA-mediated neurons in the neocortex in largely non-overlapping groups.

FAST-SPIKING

Ability of parvalbumin-positive cells to fire non-adapting action potentials at rates of up to several hundred Hertz, due, in part, to unique potassium conductances (Kv3 class).

CHANDELIER CELL

Stereotypical GABA cell of the cerebral cortex that ensheathes the axon initial segment of up to 200 pyramidal cells with 'cartridge' synapses to directly control action potential generation.

LARGE BASKET CELL

Class of GABA cell with long, horizontally-extending axon that makes potent inhibitory contacts on the soma and proximal parts of the dendrites of target pyramidal cells.

CHOLECYSTOKININ

(CCK). A neuropeptide that is found in non-fast-spiking large basket cells that contact the soma of target pyramidal cells at synapses enriched in GABAA receptor α2-subunits.

PERINEURONAL NET

A conglomeration of chondroitin sulphate proteoglycans, extracellular matrix and cell-adhesion molecules that condenses around particular large-basket cells with age.

TISSUE-TYPE PLASMINOGEN ACTIVATOR

(tPA). The major serine protease in the brain, well known as an anti-clotting agent that works by cleaving fibrin in the blood.

IMMEDIATE EARLY GENES

Transcription factors that are induced within minutes of intense neuronal activity. Examples include zif268 and c-fos.

PLASMIN

The active form of plasminogen, which is the primary target of tPA action, and itself a protease that is known to cleave extracellular matrix molecules (for example, laminin and phosphacan).

LONG-TERM DEPRESSION

(LTD). A persistent reduction of synaptic transmission in response to weak, poorly-correlated input.

LONG-TERM POTENTIATION

(LTP). A persistent strengthening of synaptic transmission in response to strong, correlated input.

SPIKE-TIMING DEPENDENT PLASTICITY

Describes physiological windows for LTP or LTD of synaptic transmission based on the arrival time of incoming action potentials with respect to back-propagated spikes in the target dendrite.

PROTEIN KINASE A

(PKA). Phosphorylates multiple targets (including AMPA and GABAA receptors) when cyclic AMP binds its regulatory subunits to release the catalytic domains.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hensch, T. Critical period plasticity in local cortical circuits. Nat Rev Neurosci 6, 877–888 (2005). https://doi.org/10.1038/nrn1787

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn1787

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing