Skip to main content
Top
Published in: NeuroMolecular Medicine 4/2012

01-12-2012 | Original Paper

Insulin Receptor β-Subunit Haploinsufficiency Impairs Hippocampal Late-Phase LTP and Recognition Memory

Authors: Robert Nisticò, Virve Cavallucci, Sonia Piccinin, Simone Macrì, Marco Pignatelli, Bisan Mehdawy, Fabio Blandini, Giovanni Laviola, Davide Lauro, Nicola B. Mercuri, Marcello D’Amelio

Published in: NeuroMolecular Medicine | Issue 4/2012

Login to get access

Abstract

The insulin receptor (IR) is a protein tyrosine kinase playing a pivotal role in the regulation of peripheral glucose metabolism and energy homoeostasis. IRs are also abundantly distributed in the cerebral cortex and hippocampus, where they regulate synaptic activity required for learning and memory. As the major anabolic hormone in mammals, insulin stimulates protein synthesis partially through the activation of the PI3K/Akt/mTOR pathway, playing fundamental roles in neuronal development, synaptic plasticity and memory. Here, by means of a multidisciplinary approach, we report that long-term synaptic plasticity and recognition memory are impaired in IR β-subunit heterozygous mice. Since IR expression is diminished in type-2 diabetes as well as in Alzheimer’s disease (AD) patients, these data may provide a mechanistic link between insulin resistance, impaired synaptic transmission and cognitive decline in humans with metabolic disorders.
Literature
go back to reference Accili, D., Drago, J., Lee, E. J., Johnson, M. D., Cool, M. H., Salvatore, P., et al. (1996). Early neonatal death in mice homozygous for a null allele of the insulin receptor gene. Nature Genetics, 12, 106–109.PubMedCrossRef Accili, D., Drago, J., Lee, E. J., Johnson, M. D., Cool, M. H., Salvatore, P., et al. (1996). Early neonatal death in mice homozygous for a null allele of the insulin receptor gene. Nature Genetics, 12, 106–109.PubMedCrossRef
go back to reference Atkins, C. M., Davare, M. A., Oh, M. C., Derkach, V., & Soderling, T. R. (2005). Bidirectional regulation of cytoplasmic polyadenylation element-binding protein phosphorylation by Ca2+/calmodulin-dependent protein kinase II and protein phosphatase 1 during hippocampal long-term potentiation. Journal of Neuroscience, 25, 5604–5610.PubMedCrossRef Atkins, C. M., Davare, M. A., Oh, M. C., Derkach, V., & Soderling, T. R. (2005). Bidirectional regulation of cytoplasmic polyadenylation element-binding protein phosphorylation by Ca2+/calmodulin-dependent protein kinase II and protein phosphatase 1 during hippocampal long-term potentiation. Journal of Neuroscience, 25, 5604–5610.PubMedCrossRef
go back to reference Baura, G. D., Foster, D. M., Porte, D., Jr, Kahn, S. E., Bergman, R. N., Cobelli, C., et al. (1993). Saturable transport of insulin from plasma into the central nervous system of dogs in vivo. A mechanism for regulated insulin delivery to the brain. Journal of Clinical Investigation, 92, 1824–1830.PubMedCrossRef Baura, G. D., Foster, D. M., Porte, D., Jr, Kahn, S. E., Bergman, R. N., Cobelli, C., et al. (1993). Saturable transport of insulin from plasma into the central nervous system of dogs in vivo. A mechanism for regulated insulin delivery to the brain. Journal of Clinical Investigation, 92, 1824–1830.PubMedCrossRef
go back to reference Beattie, E. C., Carroll, R. C., Yu, X., Morishita, W., Yasuda, H., von Zastrow, M., et al. (2000). Regulation of AMPA receptor endocytosis by a signaling mechanism shared with LTD. Nature Neuroscience, 3, 1291–1300.PubMedCrossRef Beattie, E. C., Carroll, R. C., Yu, X., Morishita, W., Yasuda, H., von Zastrow, M., et al. (2000). Regulation of AMPA receptor endocytosis by a signaling mechanism shared with LTD. Nature Neuroscience, 3, 1291–1300.PubMedCrossRef
go back to reference Cammalleri, M., Lütjens, R., Berton, F., King, A. R., Simpson, C., Francesconi, W., et al. (2003). Time-restricted role for dendritic activation of the mTOR-p70S6K pathway in the induction of late-phase long-term potentiation in the CA1. Proceedings of the National academy of Sciences of the United States of America, 100, 14368–14373.PubMedCrossRef Cammalleri, M., Lütjens, R., Berton, F., King, A. R., Simpson, C., Francesconi, W., et al. (2003). Time-restricted role for dendritic activation of the mTOR-p70S6K pathway in the induction of late-phase long-term potentiation in the CA1. Proceedings of the National academy of Sciences of the United States of America, 100, 14368–14373.PubMedCrossRef
go back to reference Clarke, J. R., Cammarota, M., Gruart, A., Izquierdo, I., & Delgado-García, J. M. (2010). Plastic modifications induced by object recognition memory processing. Proceedings of the National Academy of Sciences USA, 107, 2652–2657.CrossRef Clarke, J. R., Cammarota, M., Gruart, A., Izquierdo, I., & Delgado-García, J. M. (2010). Plastic modifications induced by object recognition memory processing. Proceedings of the National Academy of Sciences USA, 107, 2652–2657.CrossRef
go back to reference Clarke, D. W., Mudd, L., Boyd, F. T., Jr, Fields, M., & Raizada, M. K. (1986). Insulin is released from rat brain neuronal cells in culture. Journal of Neurochemistry, 47, 831–836.PubMedCrossRef Clarke, D. W., Mudd, L., Boyd, F. T., Jr, Fields, M., & Raizada, M. K. (1986). Insulin is released from rat brain neuronal cells in culture. Journal of Neurochemistry, 47, 831–836.PubMedCrossRef
go back to reference Coutellier, L., & Würbel, H. (2009). Early environmental cues affect object recognition memory in adult female but not male C57BL/6 mice. Behavioural Brain Research, 203, 312–315.PubMedCrossRef Coutellier, L., & Würbel, H. (2009). Early environmental cues affect object recognition memory in adult female but not male C57BL/6 mice. Behavioural Brain Research, 203, 312–315.PubMedCrossRef
go back to reference Craft, S. (2007). Insulin resistance and Alzheimer’s disease pathogenesis: Potential mechanisms and implications for treatment. Current Alzheimer Research, 4, 147–152.PubMedCrossRef Craft, S. (2007). Insulin resistance and Alzheimer’s disease pathogenesis: Potential mechanisms and implications for treatment. Current Alzheimer Research, 4, 147–152.PubMedCrossRef
go back to reference D’Amelio, M., Cavallucci, V., Middei, S., Marchetti, C., Pacioni, S., Ferri, A., et al. (2011). Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer’s disease. Nature Neuroscience, 14, 69–76.PubMedCrossRef D’Amelio, M., Cavallucci, V., Middei, S., Marchetti, C., Pacioni, S., Ferri, A., et al. (2011). Caspase-3 triggers early synaptic dysfunction in a mouse model of Alzheimer’s disease. Nature Neuroscience, 14, 69–76.PubMedCrossRef
go back to reference Dou, J. T., Chen, M., Dufour, F., Alkon, D. L., & Zhao, W. Q. (2005). Insulin receptor signaling in long-term memory consolidation following spatial learning. Learning & Memory, 12, 646–655.CrossRef Dou, J. T., Chen, M., Dufour, F., Alkon, D. L., & Zhao, W. Q. (2005). Insulin receptor signaling in long-term memory consolidation following spatial learning. Learning & Memory, 12, 646–655.CrossRef
go back to reference Ennaceur, A., & Delacour, J. (1988). A new one-trial test for neurobiological studies of memory in rats. 1: Behavioral data. Behavioural Brain Research, 31, 47–59.PubMedCrossRef Ennaceur, A., & Delacour, J. (1988). A new one-trial test for neurobiological studies of memory in rats. 1: Behavioral data. Behavioural Brain Research, 31, 47–59.PubMedCrossRef
go back to reference Feng, W., & Zhang, M. (2009). Organization and dynamics of PDZ-domain-related supramodules in the postsynaptic density. Nature Reviews Neuroscience, 10, 87–99.PubMedCrossRef Feng, W., & Zhang, M. (2009). Organization and dynamics of PDZ-domain-related supramodules in the postsynaptic density. Nature Reviews Neuroscience, 10, 87–99.PubMedCrossRef
go back to reference Havrankova, J., Roth, J., & Brownstein, M. (1978). Insulin receptors are widely distributed in the central nervous system of the rat. Nature, 272, 827–829.PubMedCrossRef Havrankova, J., Roth, J., & Brownstein, M. (1978). Insulin receptors are widely distributed in the central nervous system of the rat. Nature, 272, 827–829.PubMedCrossRef
go back to reference Hering, H., & Sheng, M. (2001). Dendritic spines: Structure, dynamics and regulation. Nature Reviews Neuroscience, 2, 880–888.PubMedCrossRef Hering, H., & Sheng, M. (2001). Dendritic spines: Structure, dynamics and regulation. Nature Reviews Neuroscience, 2, 880–888.PubMedCrossRef
go back to reference Kern, W., Peters, A., Fruehwald-Schultes, B., Deininger, E., Born, J., & Fehm, H. L. (2001). Improving influence of insulin on cognitive functions in humans. Neuroendocrinology, 74, 270–280.PubMedCrossRef Kern, W., Peters, A., Fruehwald-Schultes, B., Deininger, E., Born, J., & Fehm, H. L. (2001). Improving influence of insulin on cognitive functions in humans. Neuroendocrinology, 74, 270–280.PubMedCrossRef
go back to reference Kim, E., & Sheng, M. (2004). PDZ domain proteins of synapses. Nature Reviews Neuroscience, 5, 771–781.PubMedCrossRef Kim, E., & Sheng, M. (2004). PDZ domain proteins of synapses. Nature Reviews Neuroscience, 5, 771–781.PubMedCrossRef
go back to reference Lee, C. C., Huang, C. C., Wu, M. Y., & Hsu, K. S. (2005). Insulin stimulates postsynaptic density-95 protein translation via the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin signaling pathway. Journal of Biological Chemistry, 280, 18543–18550.PubMedCrossRef Lee, C. C., Huang, C. C., Wu, M. Y., & Hsu, K. S. (2005). Insulin stimulates postsynaptic density-95 protein translation via the phosphoinositide 3-kinase-Akt-mammalian target of rapamycin signaling pathway. Journal of Biological Chemistry, 280, 18543–18550.PubMedCrossRef
go back to reference Li, X. L., Aou, S., Oomura, Y., Hori, N., Fukunaga, K., & Hori, T. (2002). Impairment of long-term potentiation and spatial memory in leptin receptor-deficient rodents. Neuroscience, 113, 607–615.PubMedCrossRef Li, X. L., Aou, S., Oomura, Y., Hori, N., Fukunaga, K., & Hori, T. (2002). Impairment of long-term potentiation and spatial memory in leptin receptor-deficient rodents. Neuroscience, 113, 607–615.PubMedCrossRef
go back to reference Lin, J. W., Ju, W., Foster, K., Lee, S. H., Ahmadian, G., Wyszynski, M., et al. (2000). Distinct molecular mechanisms and divergent endocytotic pathways of AMPA receptor internalization. Nature Neuroscience, 3, 1282–1290.PubMedCrossRef Lin, J. W., Ju, W., Foster, K., Lee, S. H., Ahmadian, G., Wyszynski, M., et al. (2000). Distinct molecular mechanisms and divergent endocytotic pathways of AMPA receptor internalization. Nature Neuroscience, 3, 1282–1290.PubMedCrossRef
go back to reference Liu, L., Brown, J. C., 3rd, Webster, W. W., Morrisett, R. A., & Monaghan, D. T. (1995). Insulin potentiates N-methyl-D-aspartate receptor activity in Xenopus oocytes and rat hippocampus. Neuroscience Letters, 192, 5–8.PubMedCrossRef Liu, L., Brown, J. C., 3rd, Webster, W. W., Morrisett, R. A., & Monaghan, D. T. (1995). Insulin potentiates N-methyl-D-aspartate receptor activity in Xenopus oocytes and rat hippocampus. Neuroscience Letters, 192, 5–8.PubMedCrossRef
go back to reference Luchsinger, J. A., Tang, M. X., Shea, S., & Mayeux, R. (2004). Hyperinsulinemia and risk of Alzheimer disease. Neurology, 63, 1187–1192.PubMedCrossRef Luchsinger, J. A., Tang, M. X., Shea, S., & Mayeux, R. (2004). Hyperinsulinemia and risk of Alzheimer disease. Neurology, 63, 1187–1192.PubMedCrossRef
go back to reference Moloney, A. M., Griffin, R. J., Timmons, S., O’Connor, R., Ravid, R., & O’Neill, C. (2010). Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiology of Aging, 31, 224–243.PubMedCrossRef Moloney, A. M., Griffin, R. J., Timmons, S., O’Connor, R., Ravid, R., & O’Neill, C. (2010). Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiology of Aging, 31, 224–243.PubMedCrossRef
go back to reference Opazo, P., Watabe, A. M., Grant, S. G., & O’Dell, T. J. (2003). Phosphatidylinositol 3-kinase regulates the induction of long-term potentiation through extracellular signal-related kinase-independent mechanisms. Journal of Neuroscience, 23, 3679–3688.PubMed Opazo, P., Watabe, A. M., Grant, S. G., & O’Dell, T. J. (2003). Phosphatidylinositol 3-kinase regulates the induction of long-term potentiation through extracellular signal-related kinase-independent mechanisms. Journal of Neuroscience, 23, 3679–3688.PubMed
go back to reference Reger, M. A., Watson, G. S., Green, P. S., Wilkinson, C. W., Baker, L. D., Cholerton, B., et al. (2008). Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology, 70, 440–448.PubMedCrossRef Reger, M. A., Watson, G. S., Green, P. S., Wilkinson, C. W., Baker, L. D., Cholerton, B., et al. (2008). Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology, 70, 440–448.PubMedCrossRef
go back to reference Rivera, E. J., Goldin, A., Fulmer, N., Tavares, R., Wands, J. R., & de la Monte, S. M. (2005). Insulin and insulin-like growth factor expression and function deteriorate with progression of Alzheimer’s disease: Link to brain reductions in acetylcholine. Journal Alzheimers Disease, 8, 247–268. Rivera, E. J., Goldin, A., Fulmer, N., Tavares, R., Wands, J. R., & de la Monte, S. M. (2005). Insulin and insulin-like growth factor expression and function deteriorate with progression of Alzheimer’s disease: Link to brain reductions in acetylcholine. Journal Alzheimers Disease, 8, 247–268.
go back to reference Rodriguez-Rivera, J., Denner, L., & Dineley, K. T. (2011). Rosiglitazone reversal of Tg2576 cognitive deficits is independent of peripheral gluco-regulatory status. Behavioural Brain Research, 216, 255–261.PubMedCrossRef Rodriguez-Rivera, J., Denner, L., & Dineley, K. T. (2011). Rosiglitazone reversal of Tg2576 cognitive deficits is independent of peripheral gluco-regulatory status. Behavioural Brain Research, 216, 255–261.PubMedCrossRef
go back to reference Skeberdis, V. A., Lan, J., Zheng, X., Zukin, R. S., & Bennett, M. V. (2001). Insulin promotes rapid delivery of N-methyl-D-aspartate receptors to the cell surface by exocytosis. Proceedings of the National academy of Sciences of the United States of America, 98, 3561–3566.PubMedCrossRef Skeberdis, V. A., Lan, J., Zheng, X., Zukin, R. S., & Bennett, M. V. (2001). Insulin promotes rapid delivery of N-methyl-D-aspartate receptors to the cell surface by exocytosis. Proceedings of the National academy of Sciences of the United States of America, 98, 3561–3566.PubMedCrossRef
go back to reference Stranahan, A. M., Arumugam, T. V., Cutler, R. G., Lee, K., Egan, J. M., & Mattson, M. P. (2008). Diabetes impairs hippocampal function through glucocorticoid-mediated effects on new and mature neurons. Nature Neuroscience, 11, 309–317.PubMedCrossRef Stranahan, A. M., Arumugam, T. V., Cutler, R. G., Lee, K., Egan, J. M., & Mattson, M. P. (2008). Diabetes impairs hippocampal function through glucocorticoid-mediated effects on new and mature neurons. Nature Neuroscience, 11, 309–317.PubMedCrossRef
go back to reference Sun, X. J., Rothenberg, P., Kahn, C. R., Backer, J. M., Araki, E., Wilden, P. A., et al. (1991). Structure of the insulin receptor substrate IRS-1 defines a unique signal transduction protein. Nature, 352, 73–77.PubMedCrossRef Sun, X. J., Rothenberg, P., Kahn, C. R., Backer, J. M., Araki, E., Wilden, P. A., et al. (1991). Structure of the insulin receptor substrate IRS-1 defines a unique signal transduction protein. Nature, 352, 73–77.PubMedCrossRef
go back to reference Swiech, L., Perycz, M., Malik, A., & Jaworski, J. (2008). Role of mTOR in physiology and pathology of the nervous system. Biochimica et Biophysica Acta, 1784, 116–132.PubMedCrossRef Swiech, L., Perycz, M., Malik, A., & Jaworski, J. (2008). Role of mTOR in physiology and pathology of the nervous system. Biochimica et Biophysica Acta, 1784, 116–132.PubMedCrossRef
go back to reference Takei, N., Inamura, N., Kawamura, M., Namba, H., Hara, K., Yonezawa, K., et al. (2004). Brain-derived neurotrophic factor induces mammalian target of rapamycin-dependent local activation of translation machinery and protein synthesis in neuronal dendrites. Journal of Neuroscience, 24, 9760–9769.PubMedCrossRef Takei, N., Inamura, N., Kawamura, M., Namba, H., Hara, K., Yonezawa, K., et al. (2004). Brain-derived neurotrophic factor induces mammalian target of rapamycin-dependent local activation of translation machinery and protein synthesis in neuronal dendrites. Journal of Neuroscience, 24, 9760–9769.PubMedCrossRef
go back to reference Tang, S. J., Reis, G., Kang, H., Gingras, A. C., Sonenberg, N., & Schuman, E. M. (2002). A rapamycin-sensitive signaling pathway contributes to long-term synaptic plasticity in the hippocampus. Proceedings of the National academy of Sciences of the United States of America, 99, 467–472.PubMedCrossRef Tang, S. J., Reis, G., Kang, H., Gingras, A. C., Sonenberg, N., & Schuman, E. M. (2002). A rapamycin-sensitive signaling pathway contributes to long-term synaptic plasticity in the hippocampus. Proceedings of the National academy of Sciences of the United States of America, 99, 467–472.PubMedCrossRef
go back to reference Taylor, S. I., Cama, A., Accili, D., Barbetti, F., Quon, M. J., de la Luz Sierra, M., et al. (1992). Mutations in the insulin receptor gene. Endocrine Reviews, 13, 566–595.PubMed Taylor, S. I., Cama, A., Accili, D., Barbetti, F., Quon, M. J., de la Luz Sierra, M., et al. (1992). Mutations in the insulin receptor gene. Endocrine Reviews, 13, 566–595.PubMed
go back to reference Tzimopoulou, S., Cunningham, V. J., Nichols, T. E., Searle, G., Bird, N. P., Mistry, P., et al. (2010). A multi-center randomized proof-of-concept clinical trial applying [18F]FDG-PET for evaluation of metabolic therapy with rosiglitazone XR in mild to moderate Alzheimer’s disease. Journal of Alzheimer’s Disease, 22, 1241–1256.PubMed Tzimopoulou, S., Cunningham, V. J., Nichols, T. E., Searle, G., Bird, N. P., Mistry, P., et al. (2010). A multi-center randomized proof-of-concept clinical trial applying [18F]FDG-PET for evaluation of metabolic therapy with rosiglitazone XR in mild to moderate Alzheimer’s disease. Journal of Alzheimer’s Disease, 22, 1241–1256.PubMed
go back to reference Van der Heide, L. P., Kamal, A., Artola, A., Gispen, W. H., & Ramakers, G. M. (2005). Insulin modulates hippocampal activity-dependent synaptic plasticity in a N-methyl-d-aspartate receptor and phosphatidyl-inositol-3-kinase-dependent manner. Journal of Neurochemistry, 94, 1158–1166.PubMedCrossRef Van der Heide, L. P., Kamal, A., Artola, A., Gispen, W. H., & Ramakers, G. M. (2005). Insulin modulates hippocampal activity-dependent synaptic plasticity in a N-methyl-d-aspartate receptor and phosphatidyl-inositol-3-kinase-dependent manner. Journal of Neurochemistry, 94, 1158–1166.PubMedCrossRef
go back to reference Vetiska, S. M., Ahmadian, G., Ju, W., Liu, L., Wymann, M. P., & Wang, Y. T. (2007). GABAA receptor-associated phosphoinositide 3-kinase is required for insulin-induced recruitment of postsynaptic GABAA receptors. Neuropharmacology, 52, 146–155.PubMedCrossRef Vetiska, S. M., Ahmadian, G., Ju, W., Liu, L., Wymann, M. P., & Wang, Y. T. (2007). GABAA receptor-associated phosphoinositide 3-kinase is required for insulin-induced recruitment of postsynaptic GABAA receptors. Neuropharmacology, 52, 146–155.PubMedCrossRef
go back to reference Wan, Q., Xiong, Z. G., Man, H. Y., Ackerley, C. A., Braunton, J., Lu, W. Y., et al. (1997). Recruitment of functional GABA(A) receptors to postsynaptic domains by insulin. Nature, 388, 686–690.PubMedCrossRef Wan, Q., Xiong, Z. G., Man, H. Y., Ackerley, C. A., Braunton, J., Lu, W. Y., et al. (1997). Recruitment of functional GABA(A) receptors to postsynaptic domains by insulin. Nature, 388, 686–690.PubMedCrossRef
go back to reference Yamato, T., Misumi, Y., Yamasaki, S., Kino, M., & Aomine, M. (2004). Diabetes mellitus decreases hippocampal release of neurotransmitters: An in vivo microdialysis study of awake, freely moving rats. Diabetes, Nutrition & Metabolism, 17, 128–136. Yamato, T., Misumi, Y., Yamasaki, S., Kino, M., & Aomine, M. (2004). Diabetes mellitus decreases hippocampal release of neurotransmitters: An in vivo microdialysis study of awake, freely moving rats. Diabetes, Nutrition & Metabolism, 17, 128–136.
go back to reference Zhao, W., Chen, H., Xu, H., Moore, E., Meiri, N., Quon, M. J., et al. (1999). Brain insulin receptors and spatial memory. Correlated changes in gene expression, tyrosine phosphorylation, and signaling molecules in the hippocampus of water maze trained rats. Journal of Biological Chemistry, 274, 34893–34902.PubMedCrossRef Zhao, W., Chen, H., Xu, H., Moore, E., Meiri, N., Quon, M. J., et al. (1999). Brain insulin receptors and spatial memory. Correlated changes in gene expression, tyrosine phosphorylation, and signaling molecules in the hippocampus of water maze trained rats. Journal of Biological Chemistry, 274, 34893–34902.PubMedCrossRef
Metadata
Title
Insulin Receptor β-Subunit Haploinsufficiency Impairs Hippocampal Late-Phase LTP and Recognition Memory
Authors
Robert Nisticò
Virve Cavallucci
Sonia Piccinin
Simone Macrì
Marco Pignatelli
Bisan Mehdawy
Fabio Blandini
Giovanni Laviola
Davide Lauro
Nicola B. Mercuri
Marcello D’Amelio
Publication date
01-12-2012
Publisher
Humana Press Inc
Published in
NeuroMolecular Medicine / Issue 4/2012
Print ISSN: 1535-1084
Electronic ISSN: 1559-1174
DOI
https://doi.org/10.1007/s12017-012-8184-z

Other articles of this Issue 4/2012

NeuroMolecular Medicine 4/2012 Go to the issue