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:

Integrins in synapse regulation

Key Points

  • Integrins are major cell surface receptors for the extracellular matrix (ECM). Intracellularly, integrins link to the actin cytoskeleton and signalling systems and thus function as mechanochemical signal transducers between the extracellular environment and the cell.

  • Integrins are heterodimers of α- and β-subunits, and many integrin subtypes are expressed in the brain. During development, integrins have multifaceted roles in differentiation and maintenance of neural stem cells, axon outgrowth and dendrite arborization, in concert with signalling involving the ECM, growth factor receptors and adhesion proteins.

  • Synaptic integrins detect dynamic changes in the extracellular synaptic milieu and coordinate synapse structure and function. Integrins regulate synapse formation and maturation, in concert with glial signals, regulate postsynaptic strength by controlling neurotransmitter receptor dynamics and alter dendritic spine shape by triggering actin remodelling.

  • Different integrin subtypes participate in distinct forms of synaptic plasticity. Presumably, the differences reflect the differences in their biophysical properties, molecular interactors and signalling systems to which they are linked under specific cellular contexts.

  • The differential expression pattern of the integrin subtypes may also provide the basis for the subtle differences in behavioural phenotypes that are associated with mice deficient in particular subtypes of integrin. In the hippocampus, β1-containing integrins are required for working memory, whereas β3-containing integrins seem to be involved in emotional behaviours.

  • Much remains to be clarified about the basic properties of synaptic integrins, including the detailed cellular and subcellular expression of each of the subtypes in different brain regions and in circuits underlying specific behaviours, the native extracellular ligands of synaptic integrins and their dynamic regulation modulated by ECM remodelling, and the interactions of synaptic integrins with other binding partners within and outside synapses.

Abstract

Integrins are a large family of extracellular matrix (ECM) receptors. In the developing and adult brain, many integrins are present at high levels at synapses. The tetrapartite structure of synapses — which comprises presynaptic and postsynaptic neurons, the ECM and glial processes — places synaptic integrins in an excellent position to sense dynamic changes in the synaptic environment and use this information to coordinate further changes in synapse structure and function that will shape neural circuit properties. Recent developments in our understanding of the cellular and physiological roles of integrins, which range from control of neural process outgrowth and synapse formation to regulation of synaptic plasticity and memory, enable us to attempt a synthesis of synaptic integrin function.

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

Access options

Buy this article

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

Figure 1: Integrin functions are associated with every component of the tetrapartite synapse.
Figure 2: Integrin activation and signalling.
Figure 3: AMPA receptor regulation by β3-containing integrins.

Similar content being viewed by others

References

  1. Hynes, R. O. Integrins: bidirectional, allosteric signaling machines. Cell 110, 673–687 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Luo, B. H., Carman, C. V. & Springer, T. A. Structural basis of integrin regulation and signaling. Annu. Rev. Immunol. 25, 619–647 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Kim, C., Ye, F. & Ginsberg, M. H. Regulation of integrin activation. Annu. Rev. Cell Dev. Biol. 27, 321–345 (2011).

    CAS  PubMed  Google Scholar 

  4. Wehrle-Haller, B. Assembly and disassembly of cell matrix adhesions. Curr. Opin. Cell Biol. 24, 569–581 (2012).

    CAS  PubMed  Google Scholar 

  5. Bouvard, D., Pouwels, J., De Franceschi, N. & Ivaska, J. Integrin inactivators: balancing cellular functions in vitro and in vivo. Nat. Rev. Mol. Cell Biol. 14, 430–442 (2013).

    PubMed  Google Scholar 

  6. Dityatev, A., Schachner, M. & Sonderegger, P. The dual role of the extracellular matrix in synaptic plasticity and homeostasis. Nat. Rev. Neurosci. 11, 735–746 (2010).

    CAS  PubMed  Google Scholar 

  7. Huang, Z. et al. Distinct roles of the β1-class integrins at the developing and the mature hippocampal excitatory synapse. J. Neurosci. 26, 11208–11219 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Cingolani, L. A. et al. Activity-dependent regulation of synaptic AMPA receptor composition and abundance by β3 integrins. Neuron 58, 749–762 (2008). This study provides the first experimental evidence that β3-containing integrins control synaptic AMPAR number and composition, and mediate homeostatic synaptic scaling in a way that is similar to that demonstrated for glial TNF.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Bahr, B. A. et al. Arg-Gly-Asp-Ser-selective adhesion and the stabilization of long-term potentiation: pharmacological studies and the characterization of a candidate matrix receptor. J. Neurosci. 17, 1320–1329 (1997).

    CAS  PubMed  Google Scholar 

  10. Chun, D., Gall, C. M., Bi, X. & Lynch, G. Evidence that integrins contribute to multiple stages in the consolidation of long term potentiation in rat hippocampus. Neuroscience 105, 815–829 (2001).

    CAS  PubMed  Google Scholar 

  11. Stäubli, U., Chun, D. & Lynch, G. Time-dependent reversal of long-term potentiation by an integrin antagonist. J. Neurosci. 18, 3460–3469 (1998).

    PubMed  Google Scholar 

  12. Stäubli, U., Vanderklish, P. & Lynch, G. An inhibitor of integrin receptors blocks long-term potentiation. Behav. Neural Biol. 53, 1–5 (1990).

    PubMed  Google Scholar 

  13. Xiao, P., Bahr, B. A., Stäubli, U., Vanderklish, P. W. & Lynch, G. Evidence that matrix recognition contributes to stabilization but not induction of LTP. Neuroreport 2, 461–464 (1991).

    CAS  PubMed  Google Scholar 

  14. Kramar, E. A., Bernard, J. A., Gall, C. M. & Lynch, G. α3 integrin receptors contribute to the consolidation of long-term potentiation. Neuroscience 110, 29–39 (2002).

    CAS  PubMed  Google Scholar 

  15. Kramar, E. A., Lin, B., Rex, C. S., Gall, C. M. & Lynch, G. Integrin-driven actin polymerization consolidates long-term potentiation. Proc. Natl Acad. Sci. USA 103, 5579–5584 (2006). This study provides direct evidence that activity-dependent actin polymerization in dendritic spines is involved in LTP consolidation and requires activation of β1-containing integrins.

    CAS  PubMed  Google Scholar 

  16. Chan, C. S. et al. β1-integrins are required for hippocampal AMPA receptor-dependent synaptic transmission, synaptic plasticity, and working memory. J. Neurosci. 26, 223–232 (2006). Using mice in which β1-containing integrin was knocked out specifically in excitatory neurons of the postnatal forebrain, this study provides the initial evidence that β1-containing integrin, through the modulation of LTP, might have a role in a specific set of hippocampal-mediated behaviours, such as working memory.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Chan, C. S. et al. α3-integrins are required for hippocampal long-term potentiation and working memory. Learn. Mem. 14, 606–615 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Chan, C. S. et al. α8-integrins are required for hippocampal long-term potentiation but not for hippocampal-dependent learning. Genes Brain Behav. 9, 402–410 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. McGeachie, A. B. et al. β3 integrin is dispensable for conditioned fear and hebbian forms of plasticity in the hippocampus. Eur. J. Neurosci. 36, 2461–2469 (2012). This study suggests that β1- and β3-containing integrins might have distinctive roles in synaptic plasticity and hippocampal-mediated behaviours.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Bernard-Trifilo, J. A. et al. Integrin signaling cascades are operational in adult hippocampal synapses and modulate NMDA receptor physiology. J. Neurochem. 93, 834–849 (2005).

    CAS  PubMed  Google Scholar 

  21. McGeachie, A. B., Cingolani, L. A. & Goda, Y. A stabilising influence: integrins in regulation of synaptic plasticity. Neurosci. Res. 70, 24–29 (2011).

    PubMed  PubMed Central  Google Scholar 

  22. Kendall, T., Mukai, L., Jannuzi, A. L. & Bunch, T. A. Identification of integrin β subunit mutations that alter affinity for extracellular matrix ligand. J. Biol. Chem. 286, 30981–30993 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Pinkstaff, J. K., Detterich, J., Lynch, G. & Gall, C. Integrin subunit gene expression is regionally differentiated in adult brain. J. Neurosci. 19, 1541–1556 (1999).

    CAS  PubMed  Google Scholar 

  24. Webb, D. J., Zhang, H., Majumdar, D. & Horwitz, A. F. α5 integrin signaling regulates the formation of spines and synapses in hippocampal neurons. J. Biol. Chem. 282, 6929–6935 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Chan, C. S., Weeber, E. J., Kurup, S., Sweatt, J. D. & Davis, R. L. Integrin requirement for hippocampal synaptic plasticity and spatial memory. J. Neurosci. 23, 7107–7116 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Einheber, S., Schnapp, L. M., Salzer, J. L., Cappiello, Z. B. & Milner, T. A. Regional and ultrastructural distribution of the α8 integrin subunit in developing and adult rat brain suggests a role in synaptic function. J. Comp. Neurol. 370, 105–134 (1996).

    CAS  PubMed  Google Scholar 

  27. Chavis, P. & Westbrook, G. Integrins mediate functional pre- and postsynaptic maturation at a hippocampal synapse. Nature 411, 317–321 (2001).

    CAS  PubMed  Google Scholar 

  28. Shi, Y. & Ethell, I. M. Integrins control dendritic spine plasticity in hippocampal neurons through NMDA receptor and Ca2+/calmodulin-dependent protein kinase II-mediated actin reorganization. J. Neurosci. 26, 1813–1822 (2006).

    CAS  PubMed  Google Scholar 

  29. Wang, D. & Fawcett, J. The perineuronal net and the control of CNS plasticity. Cell Tissue Res. 349, 147–160 (2012).

    PubMed  Google Scholar 

  30. Takesian, A. E. & Hensch, T. K. Balancing plasticity/stability across brain development. Prog. Brain Res. 207, 3–34 (2013).

    PubMed  Google Scholar 

  31. Gundelfinger, E. D., Frischknecht, R., Choquet, D. & Heine, M. Converting juvenile into adult plasticity: a role for the brain's extracellular matrix. Eur. J. Neurosci. 31, 2156–2165 (2010).

    PubMed  Google Scholar 

  32. García-Alvarez, B. et al. Structural determinants of integrin recognition by talin. Mol. Cell 11, 49–58 (2003).

    PubMed  Google Scholar 

  33. Simpson, M. A. et al. Direct interactions with the integrin β1 cytoplasmic tail activate the Abl2/Arg kinase. J. Biol. Chem. 290, 8360–8372 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Warren, M. S. et al. Integrin β1 signals through Arg to regulate postnatal dendritic arborization, synapse density, and behavior. J. Neurosci. 32, 2824–2834 (2012). This paper provides the first evidence that a novel β1-containing integrin–ARG–p190GAP pathway regulating the structure and function of dendrites and synapses in the maturing hippocampus might underlie the higher vulnerability of adolescents to cocaine exposure.

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Pozo, K. et al. β3 integrin interacts directly with GluA2 AMPA receptor subunit and regulates AMPA receptor expression in hippocampal neurons. Proc. Natl Acad. Sci. USA 109, 1323–1328 (2012). This study provides the first evidence that a direct interaction between GluA2-containing AMPARs and β3-containing integrins play a part in regulating basal excitatory synaptic strength.

    CAS  PubMed  Google Scholar 

  36. Henley, J. M., Barker, E. A. & Glebov, O. O. Routes, destinations and delays: recent advances in AMPA receptor trafficking. Trends Neurosci. 34, 258–268 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Anggono, V. & Huganir, R. L. Regulation of AMPA receptor trafficking and synaptic plasticity. Curr. Opin. Neurobiol. 22, 461–469 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Chater, T. E. & Goda, Y. The role of AMPA receptors in postsynaptic mechanisms of synaptic plasticity. Front. Cell Neurosci. 8, 401 (2014).

    PubMed  PubMed Central  Google Scholar 

  39. Campos, L. S. β1 integrins and neural stem cells: making sense of the extracellular environment. Bioessays 27, 698–707 (2005).

    CAS  PubMed  Google Scholar 

  40. Yamada, K. M. & Even-Ram, S. Integrin regulation of growth factor receptors. Nat. Cell Biol. 4, E75–E76 (2002).

    CAS  PubMed  Google Scholar 

  41. Comoglio, P. M., Boccaccio, C. & Trusolino, L. Interactions between growth factor receptors and adhesion molecules: breaking the rules. Curr. Opin. Cell Biol. 15, 565–571 (2003).

    CAS  PubMed  Google Scholar 

  42. Chattopadhyay, N., Wang, Z., Ashman, L. K., Brady-Kalnay, S. M. & Kreidberg, J. A. α3β1 integrin–CD151, a component of the cadherin–catenin complex, regulates PTPμ expression and cell–cell adhesion. J. Cell Biol. 163, 1351–1362 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Förster, E. et al. Reelin, Disabled 1, and β1 integrins are required for the formation of the radial glial scaffold in the hippocampus. Proc. Natl Acad. Sci. USA 99, 13178–13183 (2002).

    PubMed  Google Scholar 

  44. Marsden, M. & DeSimone, D. W. Integrin–ECM interactions regulate cadherin-dependent cell adhesion and are required for convergent extension in Xenopus. Curr. Biol. 13, 1182–1191 (2003).

    CAS  PubMed  Google Scholar 

  45. Thelen, K. et al. The neural cell adhesion molecule L1 potentiates integrin-dependent cell migration to extracellular matrix proteins. J. Neurosci. 22, 4918–4931 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Carlstrom, L. P., Hines, J. H., Henle, S. J. & Henley, J. R. Bidirectional remodeling of β1-integrin adhesions during chemotropic regulation of nerve growth. BMC Biol. 9, 82 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Pasterkamp, R. J., Peschon, J. J., Spriggs, M. K. & Kolodkin, A. L. Semaphorin 7A promotes axon outgrowth through integrins and MAPKs. Nature 424, 398–405 (2003).

    PubMed  Google Scholar 

  48. Anton, E. S., Kreidberg, J. A. & Rakic, P. Distinct functions of α3 and αv integrin receptors in neuronal migration and laminar organization of the cerebral cortex. Neuron 22, 277–289 (1999).

    CAS  PubMed  Google Scholar 

  49. Myers, J. P., Santiago-Medina, M. & Gomez, T. M. Regulation of axonal outgrowth and pathfinding by integrin–ECM interactions. Dev. Neurobiol. 71, 901–923 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Hama, H., Hara, C., Yamaguchi, K. & Miyawaki, A. PKC signaling mediates global enhancement of excitatory synaptogenesis in neurons triggered by local contact with astrocytes. Neuron 41, 405–415 (2004).

    CAS  PubMed  Google Scholar 

  51. Xiao, X., Levy, A. D., Rosenberg, B. J., Higley, M. J. & Koleske, T. Disruption of coordinated presynaptic and postsynaptic maturation underlies the defects in hippocampal synapse stability and plasticity in Abl2/Arg-deficient mice. J. Neurosci. 36, 6778–6791 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Sfakianos, M. K. et al. Inhibition of Rho via Arg and p190RhoGAP in the postnatal mouse hippocampus regulates dendritic spine maturation, synapse and dendrite stability, and behavior. J. Neurosci. 27, 10982–10992 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Kerrisk, M. E., Greer, C. A. & Koleske, A. J. Integrin α3 is required for late postnatal stability of dendrite arbors, dendritic spines and synapses, and mouse behavior. J. Neurosci. 33, 6742–6752 (2013). This study demonstrates that α3-containing integrins and their downstream intracellular ARG–p190GAP–RHOA pathway developmentally regulate dendrite arbor stability, synapse density and ultrastructure in hippocampal neurons, similarly to the functions previously demonstrated for β1-containing integrins, and that α3-containing integrin signalling might have a key role in the cellular mechanism underlying novel-object recognition.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Lin, G. L. et al. Activation of β1 but not β3 integrin increases cell traction forces. FEBS Lett. 587, 763–769 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Charrier, C. et al. A crosstalk between β1 and β3 integrins controls glycine receptor and gephyrin trafficking at synapses. Nat. Neurosci. 13, 1388–1395 (2010). This study demonstrates that β1- and β3-containing integrins (and their corresponding ECM ligands) have surprisingly opposing roles in the scaling of basal inhibitory synaptic strength in cultured spinal cord neurons. These effects are mediated by modulating the dwell time of glycine receptors and their scaffolding protein gephyrin at synapses.

    CAS  PubMed  Google Scholar 

  56. Lin, Y. C., Yeckel, M. F. & Koleske, A. J. Abl2/Arg controls dendritic spine and dendrite arbor stability via distinct cytoskeletal control pathways. J. Neurosci. 33, 1846–1857 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Mortillo, S. et al. Compensatory redistribution of neuroligins and N-cadherin following deletion of synaptic β1-integrin. J. Comp. Neurol. 520, 2041–2052 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Michael, K. E., Dumbauld, D. W., Burns, K. L., Hanks, S. K. & García, A. J. Focal adhesion kinase modulates cell adhesion strengthening via integrin activation. Mol. Biol. Cell 20, 2508–2519 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Calderwood, D. A. & Ginsberg, M. H. Talin forges the links between integrins and actin. Nat. Cell Biol. 5, 694–697 (2003).

    CAS  PubMed  Google Scholar 

  60. Galbraith, C. G., Yamada, K. M. & Galbraith, J. A. Polymerizing actin fibers position integrins primed to probe for adhesion sites. Science 315, 992–995 (2007).

    CAS  PubMed  Google Scholar 

  61. Serrels, B. et al. Focal adhesion kinase controls actin assembly via a FERM-mediated interaction with the Arp2/3 complex. Nat. Cell Biol. 9, 1046–1056 (2007).

    CAS  PubMed  Google Scholar 

  62. Pirone, D. M. et al. An inhibitory role for FAK in regulating proliferation: a link between limited adhesion and RhoA–ROCK signaling. J. Cell Biol. 174, 277–288 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Cingolani, L. A. & Goda, Y. Differential involvement of β3 integrin in pre- and postsynaptic forms of adaptation to chronic activity deprivation. Neuron Glia Biol. 4, 179–187 (2008).

    PubMed  Google Scholar 

  64. Stellwagen, D. & Malenka, R. C. Synaptic scaling mediated by glial TNF-α. Nature 440, 1054–1059 (2006).

    CAS  PubMed  Google Scholar 

  65. Chen, B. S. & Roche, K. W. Regulation of NMDA receptors by phosphorylation. Neuropharmacology 53, 362–368 (2007).

    PubMed  PubMed Central  Google Scholar 

  66. Huntley, G. W. Synaptic circuit remodelling by matrix metalloproteinases in health and disease. Nat. Rev. Neurosci. 13, 743–757 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Shinoe, T. & Goda, Y. Tuning synapses by proteolytic remodeling of the adhesive surface. Curr. Opin. Neurobiol. 35, 148–155 (2015).

    CAS  PubMed  Google Scholar 

  68. Gawlak, M. et al. High resolution in situ zymography reveals matrix metalloproteinase activity at glutamatergic synapses. Neuroscience 158, 167–176 (2009).

    CAS  PubMed  Google Scholar 

  69. Michaluk, P. et al. Matrix metalloproteinase-9 controls NMDA receptor surface diffusion through integrin β1 signaling. J. Neurosci. 29, 6007–6012 (2009). Using single quantum dot tracking, this study provides direct evidence that MMP9 enzymatic activity increases NMDAR surface trafficking via β1-containing integrins.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Groc, L. et al. NMDA receptor surface trafficking and synaptic subunit composition are developmentally regulated by the extracellular matrix protein reelin. J. Neurosci. 27, 10165–10175 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Fatemi, S. H. Reelin mutations in mouse and man: from reeler mouse to schizophrenia, mood disorders, autism and lissencephaly. Mol. Psychiatry 6, 129–133 (2001).

    CAS  PubMed  Google Scholar 

  72. Hoe, H. S. et al. Interaction of reelin with amyloid precursor protein promotes neurite outgrowth. J. Neurosci. 29, 7459–7473 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Dong, E. et al. Reelin and glutamic acid decarboxylase67 promoter remodeling in an epigenetic methionine-induced mouse model of schizophrenia. Proc. Natl Acad. Sci. USA 102, 12578–12583 (2005).

    CAS  PubMed  Google Scholar 

  74. Samsom, J. N. & Wong, A. H. Schizophrenia and depression co-morbidity: what we have learned from animal models. Front. Psychiatry 6, 13 (2015).

    PubMed  PubMed Central  Google Scholar 

  75. Mota, S. I. et al. Impaired Src signaling and post-synaptic actin polymerization in Alzheimer's disease mice hippocampus — linking NMDA receptors and the reelin pathway. Exp. Neurol. 261, 698–709 (2014).

    CAS  PubMed  Google Scholar 

  76. Pujadas, L. et al. Reelin delays amyloid-β fibril formation and rescues cognitive deficits in a model of Alzheimer's disease. Nat. Commun. 5, 3443 (2014).

    PubMed  Google Scholar 

  77. Michaluk, P. et al. β-dystroglycan as a target for MMP-9, in response to enhanced neuronal activity. J. Biol. Chem. 282, 16036–16041 (2007).

    CAS  PubMed  Google Scholar 

  78. Wiera, G., Wozniak, G., Bajor, M., Kaczmarek, L. & Mozrzymas, J. W. Maintenance of long-term potentiation in hippocampal mossy fiber–CA3 pathway requires fine-tuned MMP-9 proteolytic activity. Hippocampus 23, 529–543 (2013).

    CAS  PubMed  Google Scholar 

  79. Nagy, V. et al. Matrix metalloproteinase-9 is required for hippocampal late-phase long-term potentiation and memory. J. Neurosci. 26, 1923–1934 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Wang, X. B. et al. Extracellular proteolysis by matrix metalloproteinase-9 drives dendritic spine enlargement and long-term potentiation coordinately. Proc. Natl Acad. Sci. USA 105, 19520–19525 (2008). This study provides experimental evidence that the MMP9-mediated mechanism driving LTP requires β1-containing integrin activity and the activation of its downstream cofilin signalling pathway, which is linked to actin polymerization.

    CAS  PubMed  Google Scholar 

  81. Bozdagi, O., Nagy, V., Kwei, K. T. & Huntley, G. W. In vivo roles for matrix metalloproteinase-9 in mature hippocampal synaptic physiology and plasticity. J. Neurophysiol. 98, 334–344 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Dziembowska, M. et al. Activity-dependent local translation of matrix metalloproteinase-9. J. Neurosci. 32, 14538–14547 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Babayan, A. H. et al. Integrin dynamics produce a delayed stage of long-term potentiation and memory consolidation. J. Neurosci. 32, 12854–12861 (2012). This study provides direct evidence that the effects of the temporal dynamics of β1-containing integrins and their downstream signalling pathway on LTP induction have a temporally discrete role in memory formation.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Lynch, G., Kramár, E. A. & Gall, C. M. Protein synthesis and consolidation of memory-related synaptic changes. Brain Res. 1621, 62–72 (2015).

    CAS  PubMed  Google Scholar 

  85. Bosch, M. et al. Structural and molecular remodeling of dendritic spine substructures during long-term potentiation. Neuron 82, 444–459 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Bi, X., Lynch, G., Zhou, J. & Gall, C. M. Polarized distribution of α5 integrin in dendrites of hippocampal and cortical neurons. J. Comp. Neurol. 435, 184–193 (2001).

    CAS  PubMed  Google Scholar 

  87. Varney, S., Polston, K. F., Jessen, T. & Carneiro, A. M. D. Mice lacking integrin β3 expression exhibit altered response to chronic stress. Neurobiol. Stress 2, 51–58 (2015).

    PubMed  PubMed Central  Google Scholar 

  88. Carter, M. D. et al. Absence of preference for social novelty and increased grooming in integrin β3 knockout mice: initial studies and future directions. Autism Res. 4, 57–67 (2011).

    PubMed  PubMed Central  Google Scholar 

  89. Moy, S. S. et al. Sociability and preference for social novelty in five inbred strains: an approach to assess autistic-like behavior in mice. Genes Brain Behav. 3, 287–302 (2004).

    CAS  PubMed  Google Scholar 

  90. Moy, S. S. et al. Mouse behavioral tasks relevant to autism: phenotypes of 10 inbred strains. Behav. Brain Res. 176, 4–20 (2007).

    PubMed  Google Scholar 

  91. Moy, S. S. et al. Development of a mouse test for repetitive, restricted behaviors: relevance to autism. Behav. Brain Res. 188, 178–194 (2008).

    CAS  PubMed  Google Scholar 

  92. Silverman, J. L., Yang, M., Lord, C. & Crawley, J. N. Behavioural phenotyping assays for mouse models of autism. Nat. Rev. Neurosci. 11, 490–502 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Nakatani, J. et al. Abnormal behavior in a chromosome-engineered mouse model for human 15q11-13 duplication seen in autism. Cell 137, 1235–1246 (2009).

    PubMed  PubMed Central  Google Scholar 

  94. Coutinho, A. M. et al. Evidence for epistasis between SLC6A4 and ITGB3 in autism etiology and in the determination of platelet serotonin levels. Hum. Genet. 121, 243–256 (2007).

    CAS  PubMed  Google Scholar 

  95. Weiss, L. A. et al. Variation in ITGB3 is associated with whole-blood serotonin level and autism susceptibility. Eur. J. Hum. Genet. 14, 923–931 (2006).

    CAS  PubMed  Google Scholar 

  96. Weiss, L. A., Ober, C. & Cook, E. H. Jr. ITGB3 shows genetic and expression interaction with SLC6A4. Hum. Genet. 120, 93–100 (2006).

    CAS  PubMed  Google Scholar 

  97. Abrahams, B. S. & Geschwind, D. H. Advances in autism genetics: on the threshold of a new neurobiology. Nat. Rev. Genet. 9, 341–355 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Cook, E. H. Jr et al. Platelet serotonin studies in hyperserotonemic relatives of children with autistic disorder. Life Sci. 52, 2005–2015 (1993).

    PubMed  Google Scholar 

  99. Coutinho, A. M. et al. Variants of the serotonin transporter gene (SLC6A4) significantly contribute to hyperserotonemia in autism. Mol. Psychiatry 9, 264–271 (2004).

    CAS  PubMed  Google Scholar 

  100. Altieri, S. C. et al. Perinatal versus genetic programming of serotonin states associated with anxiety. Neuropsychopharmacology 40, 1456–1470 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Rodríguez, J. J., Noristani, H. N. & Verkhratsky, A. The serotonergic system in ageing and Alzheimer's disease. Prog. Neurobiol. 99, 15–41 (2012).

    PubMed  Google Scholar 

  102. Stahl, S. M., Woo, D. J., Mefford, I. N., Berger, P. A. & Ciaranello, R. D. Hyperserotonemia and platelet serotonin uptake and release in schizophrenia and affective disorders. Am. J. Psychiatry 140, 26–30 (1983).

    CAS  PubMed  Google Scholar 

  103. Mestre, T. A., Zurowski, M. & Fox, S. H. 5-Hydroxytryptamine 2A receptor antagonists as potential treatment for psychiatric disorders. Expert Opin. Investig. Drugs 22, 411–421 (2013).

    CAS  PubMed  Google Scholar 

  104. de Angelis, L. 5-HT2A antagonists in psychiatric disorders. Curr. Opin. Investig. Drugs 3, 106–112 (2002).

    CAS  PubMed  Google Scholar 

  105. Brummelte, S., Mc Glanaghy, E., Bonnin, A. & Oberlander, T. F. Developmental changes in serotonin signaling: implications for early brain function, behavior and adaptation. Neuroscience http://dx.doi.org/10.1016/j.neuroscience.2016.02.037 (2016).

  106. Hanley, H. G., Stahl, S. M. & Freedman, D. X. Hyperserotonemia and amine metabolites in autistic and retarded children. Arch. Gen. Psychiatry 34, 521–531 (1977).

    CAS  PubMed  Google Scholar 

  107. Chen, Q. et al. Neural plasticity and addiction: integrin-linked kinase and cocaine behavioral sensitization. J. Neurochem. 107, 679–689 (2008).

    CAS  PubMed  Google Scholar 

  108. Quintero, G. C. Role of nucleus accumbens glutamatergic plasticity in drug addiction. Neuropsychiatr. Dis. Treat. 9, 1499–1512 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Pierce, R. C. & Wolf, M. E. Psychostimulant-induced neuroadaptations in nucleus accumbens AMPA receptor transmission. Cold Spring Harb. Perspect. Med. 3, a012021 (2013).

    PubMed  PubMed Central  Google Scholar 

  110. Lenz, J. D. & Lobo, M. K. Optogenetic insights into striatal function and behavior. Behav. Brain Res. 255, 44–54 (2013).

    CAS  PubMed  Google Scholar 

  111. Carlezon, W. A. Jr & Thomas, M. J. Biological substrates of reward and aversion: a nucleus accumbens activity hypothesis. Neuropharmacology 56, 122–132 (2009).

    CAS  PubMed  Google Scholar 

  112. Wiggins, A. T., Pacchioni, A. M. & Kalivas, P. W. Integrin expression is altered after acute and chronic cocaine. Neurosci. Lett. 450, 321–323 (2009).

    CAS  PubMed  Google Scholar 

  113. Wiggins, A. T., Smith, R. J., Shen, H.-W. & Kalivas, P. W. Integrins modulate relapse to cocaine-seeking. J. Neurosci. 31, 16177–16184 (2011). This study provides the first evidence for a correlation between temporal changes of β3-containing integrin level in the NAc during cocaine withdrawal and relapse to cocaine-seeking behaviour.

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Boudreau, A. C. & Wolf, M. E. Behavioral sensitization to cocaine is associated with increased AMPA receptor surface expression in the nucleus accumbens. J. Neurosci. 25, 9144–9151 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Boudreau, A. C., Reimers, J. M., Milovanovic, M. & Wolf, M. E. Cell surface AMPA receptors in the rat nucleus accumbens increase during cocaine withdrawal but internalize upon cocaine challenge in association with altered activation of mitogen-activated protein kinases. J. Neurosci. 27, 10621–10635 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Conrad, K. L. et al. Formation of accumbens GluR2-lacking AMPA receptors mediates incubation of cocaine craving. Nature 454, 118–121 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Famous, K. R. et al. Phosphorylation-dependent trafficking of GluR2-containing AMPA receptors in the nucleus accumbens plays a critical role in the reinstatement of cocaine seeking. J. Neurosci. 28, 11061–11070 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Graham, D. L. et al. Dynamic BDNF activity in nucleus accumbens with cocaine use increases self-administration and relapse. Nat. Neurosci. 10, 1029–1037 (2007).

    CAS  PubMed  Google Scholar 

  119. Grimm, J. W. et al. Time-dependent increases in brain-derived neurotrophic factor protein levels within the mesolimbic dopamine system after withdrawal from cocaine: implications for incubation of cocaine craving. J. Neurosci. 23, 742–747 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Li, X. et al. Different roles of BDNF in nucleus accumbens core versus shell during the incubation of cue-induced cocaine craving and its long-term maintenance. J. Neurosci. 33, 1130–1142 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Li, X. & Wolf, M. E. Multiple faces of BDNF in cocaine addiction. Behav. Brain Res. 279, 240–254 (2015).

    CAS  PubMed  Google Scholar 

  122. Brown, T. E. et al. Role of matrix metalloproteinases in the acquisition and reconsolidation of cocaine-induced conditioned place preference. Learn. Mem. 14, 214–223 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Smith, A. C. et al. Synaptic plasticity mediating cocaine relapse requires matrix metalloproteinases. Nat. Neurosci. 17, 1655–1657 (2014). This paper demonstrates the distinctive temporal dynamics of MMP2 and MMP9 activity and their roles during the course of drug addiction.

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Brown, T. E. et al. Increase in matrix metalloproteinase-9 levels in the rat medial prefrontal cortex after cocaine reinstatement of conditioned place preference. Synapse 62, 886–889 (2008).

    CAS  PubMed  Google Scholar 

  125. Ogier, C. et al. Matrix metalloproteinase-2 (MMP-2) regulates astrocyte motility in connection with the actin cytoskeleton and integrins. Glia 54, 272–284 (2006).

    PubMed  Google Scholar 

  126. Gourley, S. L., Olevska, A., Warren, M. S., Taylor, J. R. & Koleske, A. J. Arg kinase regulates prefrontal dendritic spine refinement and cocaine-induced plasticity. J. Neurosci. 32, 2314–2323 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Annaert, W. & De Strooper, B. A cell biological perspective on Alzheimer's disease. Annu. Rev. Cell Dev. Biol. 18, 25–51 (2002).

    CAS  PubMed  Google Scholar 

  128. Serrano-Pozo, A., Frosch, M. P., Masliah, E. & Hyman, B. T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 1, a006189 (2011).

    PubMed  PubMed Central  Google Scholar 

  129. De Strooper, B. & Karran, E. The cellular phase of Alzheimer's disease. Cell 164, 603–615 (2016).

    CAS  PubMed  Google Scholar 

  130. Liu-Seifert, H. et al. Cognitive impairment precedes and predicts functional impairment in mild Alzheimer's disease. J. Alzheimers Dis. 47, 205–214 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Morris, J. C. et al. Mild cognitive impairment represents early-stage Alzheimer disease. Arch. Neurol. 58, 397–405 (2001).

    CAS  PubMed  Google Scholar 

  132. Lambert, M. P. et al. Diffusible, nonfibrillar ligands derived from Aβ1–42 are potent central nervous system neurotoxins. Proc. Natl Acad. Sci. USA 95, 6448–6453 (1998).

    CAS  PubMed  Google Scholar 

  133. Walsh, D. M. et al. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo. Nature 416, 535–539 (2002).

    CAS  PubMed  Google Scholar 

  134. Shankar, G. M. et al. Amyloid-β protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat. Med. 14, 837–842 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Wang, Q. et al. αv integrins mediate β-amyloid induced inhibition of long-term potentiation. Neurobiol. Aging 29, 1485–1493 (2008).

    CAS  PubMed  Google Scholar 

  136. Rowan, M. J., Klyubin, I., Wang, Q., Hu, N. W. & Anwyl, R. Synaptic memory mechanisms: Alzheimer's disease amyloid β-peptide-induced dysfunction. Biochem. Soc. Trans. 35, 1219–1223 (2007).

    CAS  PubMed  Google Scholar 

  137. Wang, Q., Wu, J., Rowan, M. J. & Anwyl, R. β-amyloid inhibition of long-term potentiation is mediated via tumor necrosis factor. Eur. J. Neurosci. 22, 2827–2832 (2005).

    PubMed  Google Scholar 

  138. Wright, S. et al. α2β1 and αVβ1 integrin signaling pathways mediate amyloid-β-induced neurotoxicity. Neurobiol. Aging 28, 226–237 (2007).

    CAS  PubMed  Google Scholar 

  139. Mikuni, T., Nishiyama, J., Sun, Y., Kamasawa, N. & Yasuda, R. High-throughput, high-resolution mapping of protein localization in mammalian brain by in vivo genome editing. Cell 165, 1803–1817 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Rossier, O. et al. Integrins β1 and β3 exhibit distinct dynamic nanoscale organizations inside focal adhesions. Nat. Cell Biol. 14, 1057–1067 (2012).

    CAS  PubMed  Google Scholar 

  141. Choquet, D., Felsenfeld, D. P. & Sheetz, M. P. Extracellular matrix rigidity causes strengthening of integrin–cytoskeleton linkages. Cell 88, 39–48 (1997).

    CAS  PubMed  Google Scholar 

  142. Roca-Cusachs, P., Gauthier, N. C., Del Rio, A. & Sheetz, M. P. Clustering of α5β1 integrins determines adhesion strength whereas αvβ3 and talin enable mechanotransduction. Proc. Natl Acad. Sci. USA 106, 16245–16250 (2009).

    CAS  PubMed  Google Scholar 

  143. Yu, C. H. et al. Integrin-β3 clusters recruit clathrin-mediated endocytic machinery in the absence of traction force. Nat. Commun. 6, 8672 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Seo, D. et al. A mechanogenetic toolkit for interrogating cell signaling in space and time. Cell 165, 1507–1518 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Tadokoro, S. et al. Talin binding to integrin β tails: a final common step in integrin activation. Science 302, 103–106 (2003).

    CAS  PubMed  Google Scholar 

  146. Brown, R. E. & Milner, P. M. The legacy of Donald O. Hebb: more than the Hebb synapse. Nat. Rev. Neurosci. 4, 1013–1019 (2003).

    CAS  PubMed  Google Scholar 

  147. Collingridge, G. L., Isaac, J. T. & Wang, Y. T. Receptor trafficking and synaptic plasticity. Nat. Rev. Neurosci. 5, 952–962 (2004).

    CAS  PubMed  Google Scholar 

  148. Cooper, L. N. & Bear, M. F. The BCM theory of synapse modification at 30: interaction of theory with experiment. Nat. Rev. Neurosci. 13, 798–810 (2012).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  150. Pozo, K. & Goda, Y. Unraveling mechanisms of homeostatic synaptic plasticity. Neuron 66, 337–351 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Turrigiano, G. G. The self-tuning neuron: synaptic scaling of excitatory synapses. Cell 135, 422–435 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Sekine, K. et al. Reelin controls neuronal positioning by promoting cell-matrix adhesion via inside-out activation of integrin α5β1. Neuron 76, 353–369 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Graus-Porta, D. et al. β1-class integrins regulate the development of laminae and folia in the cerebral and cerebellar cortex. Neuron 31, 367–379 (2001).

    CAS  PubMed  Google Scholar 

  154. Barros, C. S. et al. β1 integrins are required for normal CNS myelination and promote AKT-dependent myelin outgrowth. Development 136, 2717–2724 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Câmara, J. et al. Integrin-mediated axoglial interactions initiate myelination in the central nervous system. J. Cell Biol. 185, 699–712 (2009).

    PubMed  PubMed Central  Google Scholar 

  156. Lee, K. K. et al. Dominant-negative β1 integrin mice have region-specific myelin defects accompanied by alterations in MAPK activity. Glia 53, 836–844 (2006).

    PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank T. Chater and P. Chipman for critical reading of the manuscript. Research in the authors' laboratory is supported by the RIKEN Brain Science Institute, the Japan Society for the Promotion of Science (JSPS) Core-to-Core Program, Grants-in-Aid for Scientific Research (15H04280) from the Ministry of Education, Culture, Sports, Science and Technology (MEXT), and the Brain Mapping by Integrated Neurotechnologies for Disease Studies (Brain/MINDS) from the Japan Agency for Medical Research and Development (AMED).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Yukiko Goda.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Counter receptors

Molecules that bind to cell surface receptors. Counter receptors for integrins include neural cell adhesion molecule L1 (NCAML1) and vascular cell adhesion protein 1 (VCAM1), which themselves are cell surface proteins.

Adaptor proteins

Molecules containing distinct modular domains that typically mediate protein–protein interactions and allow these proteins to form signal transduction complexes.

Perineuronal nets

Specialized extracellular matrix structures that surround the somata and proximal dendrites of certain neuron types. They are important for synaptic stabilization and have been suggested to be crucial for the closure of critical periods.

Synaptic dwell time

The amount of time during which a diffusible molecule remains inside the synapse.

Focal adhesion sites

Specialized sites of adhesive contact where integrins link the extracellular matrix to intracellular signalling complexes and to the actin cytoskeleton.

Long-term potentiation

(LTP). A long-lasting (hours or days) increase in the response of neurons to stimulation of their afferents following a brief patterned stimulus (for example, a 100 Hz stimulus train).

Long-term depression

(LTD). A long-lasting decrease in the response of neurons to stimulation of their afferents following a brief patterned stimulus (for example, a 1 Hz stimulus train).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Park, Y., Goda, Y. Integrins in synapse regulation. Nat Rev Neurosci 17, 745–756 (2016). https://doi.org/10.1038/nrn.2016.138

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn.2016.138

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