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:

The role of G protein-coupled receptors in the pathology of Alzheimer's disease

Key Points

  • Alzheimer's disease (AD), the most common neurodegenerative disorder, is characterized by massive neuronal cell and synapse loss, senile plaques composed primarily of the amyloid-β peptide, and tau-containing neurofibrillary lesions.

  • Current AD therapies mainly target acetylcholinesterase, which broadly stimulates cholinergic neurons. However, neurodegeneration is not limited to a specific neurotransmitter system. Glutamatergic, serotonergic, adrenergic and peptidergic neurotransmitter systems are also deregulated in AD.

  • G protein-coupled receptors (GPCRs), also known as seven-transmembrane receptors (7TMRs), are the largest class of transmembrane receptors and a common therapeutic target. Several studies have presented compelling evidence implicating GPCRs in the pathogenesis of AD and in multiple stages of the proteolysis of amyloid precursor protein (APP).

  • Amyloid-β is produced from sequential proteolysis of the APP by β- and γ-secretase. By contrast, sequential cleavage by α- and γ-secretase precludes amyloid-β generation. α-, β- and γ-secretases are regulated by GPCRs, and amyloid-β itself can directly or indirectly affect GPCR function.

  • Activation of muscarinic acetycholine receptors (mAChRs), group I metabotropic glutamate receptors (mGluRs), and 5-hydroxytryptamine (5-HT; also known as serotonin) receptors have been shown to increase the non-amyloidogenic processing of APP through activation of α-secretase. For example, the M1 and M3 mAChR agonist AF267B reduces amyloid- and tau-related pathologies and rescues hippocampus-dependent learning and memory impairments in an AD mouse model.

  • The opioid receptors are involved in learning and memory and are deregulated in the AD brain. Recent studies suggest that these GPCRs and their ligand, enkephalin, are involved in modulation of the β-secretase and subsequent amyloid-β generation.

  • The β2 adrenergic receptor (β2-AR) and G protein-coupled receptor 3 (GPR3) are involved in regulation of the γ-secretase-mediated cleavage of APP. The β2-AR influences the localization of the γ-secretase complex and thereby regulates the amyloidogenic processing of APP and exacerbates the amyloid pathology in an AD mouse model. GPR3 regulates the in vitro and in vivo amyloidogenic proteolysis of APP through modulation of the localization and/or activity of the γ-secretase complex in the absence of an effect on Notch processing.

  • Although the mechanism of amyloid-β-mediated toxicity is not clearly understood, recent studies suggest that amyloid-β accumulation is involved in the oligomerization of the angiotensin type 2 receptor and sequestration of the Gαq/11 family of G proteins. Sequestration of Gαq/11 results in dysfunctional M1 mAChR–Gαq/11 coupling and signalling, and accompanies hippocampal neurodegeneration, tau phosphorylation and neuronal loss in an AD transgenic mouse model.

  • The chemokine receptors CC-chemokine receptor 2 (CCR2) and CX3C-chemokine receptor 1 (CX3CR1) have a putative role in Alzheimer's disease pathology. These receptors are expressed by microglia, which have been shown to surround amyloid plaques both in patients with AD and in AD transgenic mouse models, but whether they have beneficial or detrimental effects on plaque formation remains unclear.

  • Promoting amyloid-β clearance from the brain is a possible therapeutic strategy for inhibition of amyloid-β generation. However, stimulation of GPCRs, in particular the somatostatin receptor, could represent an interesting alternative approach, as these GPCRs induce expression of amyloid-β-degrading enzymes, such as neprilysin, in the brain. A combination of memory enhancement, neuroprotection and anti-amyloid-β activity makes this an attractive therapeutic approach for AD.

Abstract

G protein-coupled receptors (GPCRs) are involved in numerous key neurotransmitter systems in the brain that are disrupted in Alzheimer's disease (AD). GPCRs also directly influence the amyloid cascade through modulation of the α-, β- and γ-secretases, proteolysis of the amyloid precursor protein (APP), and regulation of amyloid-β degradation. Additionally, amyloid-β has been shown to perturb GPCR function. Emerging insights into the mechanistic link between GPCRs and AD highlight the potential of this class of receptors as a therapeutic target for AD.

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: Modulation of APP processing by GPCRs.
Figure 2: GPCR signalling and the α-secretase pathway.
Figure 3: Amyloid-β toxicity and deregulation of AT2R and M1 mAChR signalling.
Figure 4: Adenosine A2A receptor and amyloid-β-mediated toxicity.

Similar content being viewed by others

References

  1. De Strooper, B. Proteases and proteolysis in Alzheimer disease: a multifactorial view on the disease process. Physiol. Rev. 90, 465–494 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Furukawa, K. et al. Increased activity-regulating and neuroprotective efficacy of alpha-secretase-derived secreted amyloid precursor protein conferred by a C-terminal heparin-binding domain. J. Neurochem. 67, 1882–1896 (1996).

    Article  CAS  PubMed  Google Scholar 

  3. Small, D. H. et al. A heparin-binding domain in the amyloid protein precursor of Alzheimer's disease is involved in the regulation of neurite outgrowth. J. Neurosci. 14, 2117–2127 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ishida, A., Furukawa, K., Keller, J. N. & Mattson, M. P. Secreted form of beta-amyloid precursor protein shifts the frequency dependency for induction of LTD, and enhances LTP in hippocampal slices. Neuroreport 8, 2133–2137 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Sennvik, K. et al. Levels of alpha- and beta-secretase cleaved amyloid precursor protein in the cerebrospinal fluid of Alzheimer's disease patients. Neurosci. Lett. 278, 169–172 (2000).

    Article  CAS  PubMed  Google Scholar 

  6. Jang, H. et al. Truncated beta-amyloid peptide channels provide an alternative mechanism for Alzheimer's Disease and Down syndrome. Proc. Natl Acad. Sci. USA 107, 6538–6543 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Buxbaum, J. D. et al. Processing of Alzheimer beta/A4 amyloid precursor protein: modulation by agents that regulate protein phosphorylation. Proc. Natl Acad. Sci. USA 87, 6003–6006 (1990). This is the first study to demonstrate that proteolytic processing of APP involves a signal transduction cascade via activation of PKC.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Caporaso, G. L., Gandy, S. E., Buxbaum, J. D., Ramabhadran, T. V. & Greengard, P. Protein phosphorylation regulates secretion of Alzheimer beta/A4 amyloid precursor protein. Proc. Natl Acad. Sci. USA 89, 3055–3059 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Efthimiopoulos, S. et al. Intracellular cyclic AMP inhibits constitutive and phorbol ester-stimulated secretory cleavage of amyloid precursor protein. J. Neurochem. 67, 872–875 (1996).

    Article  CAS  PubMed  Google Scholar 

  10. Robert, S. J., Zugaza, J. L., Fischmeister, R., Gardier, A. M. & Lezoualc'h, F. The human serotonin 5-HT4 receptor regulates secretion of non-amyloidogenic precursor protein. J. Biol. Chem. 276, 44881–44888 (2001).

    Article  CAS  PubMed  Google Scholar 

  11. Xu, H., Sweeney, D., Greengard, P. & Gandy, S. Metabolism of Alzheimer beta-amyloid precursor protein: regulation by protein kinase A in intact cells and in a cell-free system. Proc. Natl Acad. Sci. USA 93, 4081–4084 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mills, J. et al. Regulation of amyloid precursor protein catabolism involves the mitogen-activated protein kinase signal transduction pathway. J. Neurosci. 17, 9415–9422 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Solano, D. C. et al. Insulin regulates soluble amyloid precursor protein release via phosphatidyl inositol 3 kinase-dependent pathway. FASEB J. 14, 1015–1022 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Buxbaum, J. D., Koo, E. H. & Greengard, P. Protein phosphorylation inhibits production of Alzheimer amyloid beta/A4 peptide. Proc. Natl Acad. Sci. USA 90, 9195–9198 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hung, A. Y. et al. Activation of protein kinase C inhibits cellular production of the amyloid beta-protein. J. Biol. Chem. 268, 22959–22962 (1993).

    CAS  PubMed  Google Scholar 

  16. da Cruz e Silva, O. A. et al. Enhanced generation of Alzheimer's amyloid-beta following chronic exposure to phorbol ester correlates with differential effects on alpha and epsilon isozymes of protein kinase C. J. Neurochem. 108, 319–330 (2009).

    Article  CAS  PubMed  Google Scholar 

  17. Levey, A. I., Kitt, C. A., Simonds, W. F., Price, D. L. & Brann, M. R. Identification and localization of muscarinic acetylcholine receptor proteins in brain with subtype-specific antibodies. J. Neurosci. 11, 3218–3226 (1991).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Wei, J., Walton, E. A., Milici, A. & Buccafusco, J. J. m1-m5 muscarinic receptor distribution in rat CNS by RT-PCR and HPLC. J. Neurochem. 63, 815–821 (1994).

    Article  CAS  PubMed  Google Scholar 

  19. Wess, J., Eglen, R. M. & Gautam, D. Muscarinic acetylcholine receptors: mutant mice provide new insights for drug development. Nature Rev. Drug Discov. 6, 721–733 (2007).

    Article  CAS  Google Scholar 

  20. Nitsch, R. M., Slack, B. E., Wurtman, R. J. & Growdon, J. H. Release of Alzheimer amyloid precursor derivatives stimulated by activation of muscarinic acetylcholine receptors. Science 258, 304–307 (1992). This study and reference 21 were the first to demonstrate the effect of neurotransmitter receptor activation on the proteolysis of APP. Stimulation of the M1 mAChR and the M3 mAChR increases the PKC-mediated release of sAPP.

    Article  CAS  PubMed  Google Scholar 

  21. Buxbaum, J. D. et al. Cholinergic agonists and interleukin 1 regulate processing and secretion of the Alzheimer beta/A4 amyloid protein precursor. Proc. Natl Acad. Sci. USA 89, 10075–10078 (1992). Along with reference 20, this report demonstrates that activation of the M1 mAChR stimulates the release of sAPP.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Flynn, D. D., Ferrari-DiLeo, G., Mash, D. C. & Levey, A. I. Differential regulation of molecular subtypes of muscarinic receptors in Alzheimer's disease. J. Neurochem. 64, 1888–1891 (1995).

    Article  CAS  PubMed  Google Scholar 

  23. Anagnostaras, S. G. et al. Selective cognitive dysfunction in acetylcholine M1 muscarinic receptor mutant mice. Nature Neurosci. 6, 51–58 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Levey, A. I. Muscarinic acetylcholine receptor expression in memory circuits: implications for treatment of Alzheimer disease. Proc. Natl Acad. Sci. USA 93, 13541–13546 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Messer, W. S. Jr, Bohnett, M. & Stibbe, J. Evidence for a preferential involvement of M1 muscarinic receptors in representational memory. Neurosci. Lett. 116, 184–189 (1990).

    Article  CAS  PubMed  Google Scholar 

  26. Wall, S. J. et al. Production of antisera selective for m1 muscarinic receptors using fusion proteins: distribution of m1 receptors in rat brain. Mol. Pharmacol. 39, 643–649 (1991).

    CAS  PubMed  Google Scholar 

  27. Caccamo, A. et al. M1 receptors play a central role in modulating AD-like pathology in transgenic mice. Neuron 49, 671–682 (2006). This study demonstrates that the selective M1 mAChR agonist AF267B reduces the cellular and learning and memory impairments in an AD mouse model. It also demonstrates that the underlying mechanism involves activation of ADAM17.

    Article  CAS  PubMed  Google Scholar 

  28. Davis, A. A., Fritz, J. J., Wess, J., Lah, J. J. & Levey, A. I. Deletion of M1 muscarinic acetylcholine receptors increases amyloid pathology in vitro and in vivo. J. Neurosci. 30, 4190–4196 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jones, C. K. et al. Novel selective allosteric activator of the M1 muscarinic acetylcholine receptor regulates amyloid processing and produces antipsychotic-like activity in rats. J. Neurosci. 28, 10422–10433 (2008). This study identifies the first specific allosteric M1 mAChR agonist, TBPD, which increases the non-amyloidogenic processing of APP and decreases amyloid-β generation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Farber, S. A., Nitsch, R. M., Schulz, J. G. & Wurtman, R. J. Regulated secretion of beta-amyloid precursor protein in rat brain. J. Neurosci. 15, 7442–7451 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lee, R. K., Wurtman, R. J., Cox, A. J. & Nitsch, R. M. Amyloid precursor protein processing is stimulated by metabotropic glutamate receptors. Proc. Natl Acad. Sci. USA 92, 8083–8087 (1995). This is the first study to demonstrate that the metabotropic glutamate receptors are involved in the proteolysis of APP and sAPP release.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Conn, P. J. & Pin, J. P. Pharmacology and functions of metabotropic glutamate receptors. Annu. Rev. Pharmacol. Toxicol. 37, 205–237 (1997).

    Article  CAS  PubMed  Google Scholar 

  33. Schoepp, D. D., Jane, D. E. & Monn, J. A. Pharmacological agents acting at subtypes of metabotropic glutamate receptors. Neuropharmacology 38, 1431–1476 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Pinheiro, P. S. & Mulle, C. Presynaptic glutamate receptors: physiological functions and mechanisms of action. Nature Rev. Neurosci. 9, 423–436 (2008).

    Article  CAS  Google Scholar 

  35. Schoepp, D. D. Unveiling the functions of presynaptic metabotropic glutamate receptors in the central nervous system. J. Pharmacol. Exp. Ther. 299, 12–20 (2001).

    CAS  PubMed  Google Scholar 

  36. Ferraguti, F., Baldani-Guerra, B., Corsi, M., Nakanishi, S. & Corti, C. Activation of the extracellular signal-regulated kinase 2 by metabotropic glutamate receptors. Eur. J. Neurosci. 11, 2073–2082 (1999).

    Article  CAS  PubMed  Google Scholar 

  37. Phillips, T., Barnes, A., Scott, S., Emson, P. & Rees, S. Human metabotropic glutamate receptor 2 couples to the MAP kinase cascade in chinese hamster ovary cells. Neuroreport 9, 2335–2339 (1998).

    Article  CAS  PubMed  Google Scholar 

  38. Albasanz, J. L., Dalfo, E., Ferrer, I. & Martin, M. Impaired metabotropic glutamate receptor/phospholipase C signaling pathway in the cerebral cortex in Alzheimer's disease and dementia with Lewy bodies correlates with stage of Alzheimer's-disease-related changes. Neurobiol. Dis. 20, 685–693 (2005).

    Article  CAS  PubMed  Google Scholar 

  39. Westmark, C. J., Westmark, P. R. & Malter, J. S. MPEP reduces seizure severity in Fmr-1 KO mice over expressing human Abeta. Int. J. Clin. Exp. Pathol. 3, 56–68 (2009).

    PubMed  Google Scholar 

  40. Lee, H. G. et al. Aberrant expression of metabotropic glutamate receptor 2 in the vulnerable neurons of Alzheimer's disease. Acta Neuropathol. 107, 365–371 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Lee, H. G. et al. The effect of mGluR2 activation on signal transduction pathways and neuronal cell survival. Brain Res. 1249, 244–250 (2009).

    Article  CAS  PubMed  Google Scholar 

  42. Kim, S. H. et al. Group II metabotropic glutamate receptor stimulation triggers production and release of Alzheimer's amyloid b42 from isolated intact nerve terminals. J. Neurosci. 30, 3870–3875 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kim, J. et al. Abeta40 inhibits amyloid deposition in vivo. J. Neurosci. 27, 627–633 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Caughey, B. & Lansbury, P. T. Protofibrils, pores, fibrils, and neurodegeneration: separating the responsible protein aggregates from the innocent bystanders. Annu. Rev. Neurosci. 26, 267–298 (2003).

    Article  CAS  PubMed  Google Scholar 

  45. Younkin, S. G. The role of A beta 42 in Alzheimer's disease. J. Physiol. Paris 92, 289–292 (1998).

    Article  CAS  PubMed  Google Scholar 

  46. Fryer, J. D. & Holtzman, D. M. The bad seed in Alzheimer's disease. Neuron 47, 167–168 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Higgins, G. A. et al. Pharmacological manipulation of mGlu2 receptors influences cognitive performance in the rodent. Neuropharmacology 46, 907–917 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Blin, J. et al. Loss of brain 5-HT2 receptors in Alzheimer's disease. In vivo assessment with positron emission tomography and [18F]setoperone. Brain 116, 497–510 (1993).

    Article  PubMed  Google Scholar 

  49. Holmes, C., Arranz, M. J., Powell, J. F., Collier, D. A. & Lovestone, S. 5-HT2A and 5-HT2C receptor polymorphisms and psychopathology in late onset Alzheimer's disease. Hum. Mol. Genet. 7, 1507–1509 (1998).

    Article  CAS  PubMed  Google Scholar 

  50. Assal, F. et al. Association of the serotonin transporter and receptor gene polymorphisms in neuropsychiatric symptoms in Alzheimer disease. Arch. Neurol. 61, 1249–1253 (2004).

    Article  PubMed  Google Scholar 

  51. Holmes, C., Arranz, M., Collier, D., Powell, J. & Lovestone, S. Depression in Alzheimer's disease: the effect of serotonin receptor gene variation. Am. J. Med. Genet. B Neuropsychiatr. Genet. 119B, 40–43 (2003).

    Article  PubMed  Google Scholar 

  52. Nitsch, R. M., Deng, M., Growdon, J. H. & Wurtman, R. J. Serotonin 5-HT2a and 5-HT2c receptors stimulate amyloid precursor protein ectodomain secretion. J. Biol. Chem. 271, 4188–4194 (1996).

    Article  CAS  PubMed  Google Scholar 

  53. Arjona, A. A., Pooler, A. M., Lee, R. K. & Wurtman, R. J. Effect of a 5-HT(2C) serotonin agonist, dexnorfenfluramine, on amyloid precursor protein metabolism in guinea pigs. Brain Res. 951, 135–140 (2002).

    Article  CAS  PubMed  Google Scholar 

  54. Medhurst, A. D., Lezoualc'h, F., Fischmeister, R., Middlemiss, D. N. & Sanger, G. J. Quantitative mRNA analysis of five C-terminal splice variants of the human 5-HT4 receptor in the central nervous system by TaqMan real time RT-PCR. Brain Res. Mol. Brain Res. 90, 125–134 (2001).

    Article  CAS  PubMed  Google Scholar 

  55. Cachard-Chastel, M. et al. 5-HT4 receptor agonists increase sAPPalpha levels in the cortex and hippocampus of male C57BL/6j mice. Br. J. Pharmacol. 150, 883–892 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Consolo, S., Arnaboldi, S., Giorgi, S., Russi, G. & Ladinsky, H. 5-HT4 receptor stimulation facilitates acetylcholine release in rat frontal cortex. Neuroreport 5, 1230–1232 (1994).

    Article  CAS  PubMed  Google Scholar 

  57. Cho, S. & Hu, Y. Activation of 5-HT4 receptors inhibits secretion of beta-amyloid peptides and increases neuronal survival. Exp. Neurol. 203, 274–278 (2007).

    Article  CAS  PubMed  Google Scholar 

  58. Reynolds, G. P. et al. 5-Hydroxytryptamine (5-HT)4 receptors in post mortem human brain tissue: distribution, pharmacology and effects of neurodegenerative diseases. Br. J. Pharmacol. 114, 993–998 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ruat, M. et al. A novel rat serotonin (5-HT6) receptor: molecular cloning, localization and stimulation of cAMP accumulation. Biochem. Biophys. Res. Commun. 193, 268–276 (1993).

    Article  CAS  PubMed  Google Scholar 

  60. Kohen, R. et al. Cloning, characterization, and chromosomal localization of a human 5-HT6 serotonin receptor. J. Neurochem. 66, 47–56 (1996).

    Article  CAS  PubMed  Google Scholar 

  61. Foley, A. G. et al. The 5-HT(6) receptor antagonist SB-271046 reverses scopolamine-disrupted consolidation of a passive avoidance task and ameliorates spatial task deficits in aged rats. Neuropsychopharmacology 29, 93–100 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. Mitchell, E. S. & Neumaier, J. F. 5-HT6 receptors: a novel target for cognitive enhancement. Pharmacol. Ther. 108, 320–333 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Geldenhuys, W. J. & Van der Schyf, C. J. The serotonin 5-HT6 receptor: a viable drug target for treating cognitive deficits in Alzheimer's disease. Expert Rev. Neurother. 9, 1073–1085 (2009).

    Article  CAS  PubMed  Google Scholar 

  64. Upton, N., Chuang, T. T., Hunter, A. J. & Virley, D. J. 5-HT6 receptor antagonists as novel cognitive enhancing agents for Alzheimer's disease. Neurotherapeutics 5, 458–469 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Tsai, S. J., Liu, H. C., Liu, T. Y., Wang, Y. C. & Hong, C. J. Association analysis of the 5-HT6 receptor polymorphism C267T in Alzheimer's disease. Neurosci. Lett. 276, 138–139 (1999).

    Article  CAS  PubMed  Google Scholar 

  66. Lorke, D. E., Lu, G., Cho, E. & Yew, D. T. Serotonin 5-HT2A and 5-HT6 receptors in the prefrontal cortex of Alzheimer and normal aging patients. BMC Neurosci. 7, 36 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Lezoualc'h, F., Engert, S., Berning, B. & Behl, C. Corticotropin-releasing hormone-mediated neuroprotection against oxidative stress is associated with the increased release of non-amyloidogenic amyloid beta precursor protein and with the suppression of nuclear factor-kappaB. Mol. Endocrinol. 14, 147–159 (2000).

    Article  CAS  PubMed  Google Scholar 

  68. Bissette, G., Reynolds, G. P., Kilts, C. D., Widerlov, E. & Nemeroff, C. B. Corticotropin-releasing factor-like immunoreactivity in senile dementia of the Alzheimer type. Reduced cortical and striatal concentrations. JAMA 254, 3067–3069 (1985).

    Article  CAS  PubMed  Google Scholar 

  69. De Souza, E. B., Whitehouse, P. J., Kuhar, M. J., Price, D. L. & Vale, W. W. Reciprocal changes in corticotropin-releasing factor (CRF)-like immunoreactivity and CRF receptors in cerebral cortex of Alzheimer's disease. Nature 319, 593–595 (1986).

    Article  CAS  PubMed  Google Scholar 

  70. Pomara, N. et al. CSF corticotropin-releasing factor (CRF) in Alzheimer's disease: its relationship to severity of dementia and monoamine metabolites. Biol. Psychiatry 26, 500–504 (1989).

    Article  CAS  PubMed  Google Scholar 

  71. Behan, D. P. et al. Displacement of corticotropin releasing factor from its binding protein as a possible treatment for Alzheimer's disease. Nature 378, 284–287 (1995).

    Article  CAS  PubMed  Google Scholar 

  72. Joo, K. M. et al. Distribution of vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide receptors (VPAC1, VPAC2, and PAC1 receptor) in the rat brain. J. Comp. Neurol. 476, 388–413 (2004).

    Article  CAS  PubMed  Google Scholar 

  73. Sacchetti, B. et al. Pituitary adenylate cyclase-activating polypeptide hormone (PACAP) at very low dosages improves memory in the rat. Neurobiol. Learn. Mem. 76, 1–6 (2001).

    Article  CAS  PubMed  Google Scholar 

  74. Kojro, E. et al. The neuropeptide PACAP promotes the alpha-secretase pathway for processing the Alzheimer amyloid precursor protein. FASEB J. 20, 512–514 (2006).

    Article  CAS  PubMed  Google Scholar 

  75. Dogrukol-Ak, D., Tore, F. & Tuncel, N. Passage of VIP/PACAP/secretin family across the blood-brain barrier: therapeutic effects. Curr. Pharm. Des 10, 1325–1340 (2004).

    Article  CAS  PubMed  Google Scholar 

  76. Vassar, R., Kovacs, D. M., Yan, R. & Wong, P. C. The beta-secretase enzyme BACE in health and Alzheimer's disease: regulation, cell biology, function, and therapeutic potential. J. Neurosci. 29, 12787–12794 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. De Strooper, B., Vassar, R. & Golde, T. The secretases: enzymes with therapeutic potential in Alzheimer disease. Nature Rev. Neurol. 6, 99–107 (2010).

    Article  CAS  Google Scholar 

  78. Teng, L., Zhao, J., Wang, F., Ma, L. & Pei, G. A GPCR/secretase complex regulates beta- and gamma-secretase specificity for Abeta production and contributes to AD pathogenesis. Cell Res. 20, 138–153 (2010).

    Article  CAS  PubMed  Google Scholar 

  79. Mathieu-Kia, A. M., Fan, L. Q., Kreek, M. J., Simon, E. J. & Hiller, J. M. Mu-, delta- and kappa-opioid receptor populations are differentially altered in distinct areas of postmortem brains of Alzheimer's disease patients. Brain Res. 893, 121–134 (2001).

    Article  CAS  PubMed  Google Scholar 

  80. Ni, Y. et al. Activation of beta2-adrenergic receptor stimulates gamma-secretase activity and accelerates amyloid plaque formation. Nature Med. 12, 1390–1396 (2006). This study demonstrates that the β 2 -AR regulates the localization of the γ-secretase complex, thereby regulating the amyloidogenic processing of APP and exacerbating the amyloid pathology in an AD mouse model.

    Article  CAS  PubMed  Google Scholar 

  81. Meilandt, W. J. et al. Enkephalin elevations contribute to neuronal and behavioral impairments in a transgenic mouse model of Alzheimer's disease. J. Neurosci. 28, 5007–5017 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Diez, M. et al. Neuropeptide alterations in the hippocampal formation and cortex of transgenic mice overexpressing beta-amyloid precursor protein (APP) with the Swedish double mutation (APP23). Neurobiol. Dis. 14, 579–594 (2003).

    Article  CAS  PubMed  Google Scholar 

  83. Williams, J. T., Christie, M. J. & Manzoni, O. Cellular and synaptic adaptations mediating opioid dependence. Physiol. Rev. 81, 299–343 (2001).

    Article  CAS  PubMed  Google Scholar 

  84. von Zastrow, M., Svingos, A., Haberstock-Debic, H. & Evans, C. Regulated endocytosis of opioid receptors: cellular mechanisms and proposed roles in physiological adaptation to opiate drugs. Curr. Opin. Neurobiol. 13, 348–353 (2003).

    Article  CAS  PubMed  Google Scholar 

  85. Jansen, K. L., Faull, R. L., Dragunow, M. & Synek, B. L. Alzheimer's disease: changes in hippocampal N-methyl-D-aspartate, quisqualate, neurotensin, adenosine, benzodiazepine, serotonin and opioid receptors — an autoradiographic study. Neuroscience 39, 613–627 (1990).

    Article  CAS  PubMed  Google Scholar 

  86. De Strooper, B. Aph-1, Pen-2, and Nicastrin with Presenilin generate an active gamma-Secretase complex. Neuron 38, 9–12 (2003).

    Article  CAS  PubMed  Google Scholar 

  87. Langui, D. et al. Subcellular topography of neuronal Abeta peptide in APPxPS1 transgenic mice. Am. J. Pathol. 165, 1465–1477 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Pasternak, S. H. et al. Presenilin-1, nicastrin, amyloid precursor protein, and gamma-secretase activity are co-localized in the lysosomal membrane. J. Biol. Chem. 278, 26687–26694 (2003).

    Article  CAS  PubMed  Google Scholar 

  89. Kalaria, R. N. et al. Adrenergic receptors in aging and Alzheimer's disease: increased beta 2-receptors in prefrontal cortex and hippocampus. J. Neurochem. 53, 1772–1781 (1989).

    Article  CAS  PubMed  Google Scholar 

  90. Yu, J. T. et al. Polymorphisms at the beta2-adrenergic receptor gene influence Alzheimer's disease susceptibility. Brain Res. 1210, 216–222 (2008).

    Article  CAS  PubMed  Google Scholar 

  91. Uhlenbrock, K., Gassenhuber, H. & Kostenis, E. Sphingosine 1-phosphate is a ligand of the human gpr3, gpr6 and gpr12 family of constitutively active G protein-coupled receptors. Cell Signal 14, 941–953 (2002).

    Article  CAS  PubMed  Google Scholar 

  92. Thathiah, A. et al. The orphan G protein-coupled receptor 3 modulates amyloid-beta peptide generation in neurons. Science 323, 946–951 (2009). This study demonstrates that the orphan GPCR GPR3 regulates the in vitro and in vivo amyloidogenic proteolysis of APP through modulation of the localization and/or activity of the γ-secretase complex in the absence of an effect on Notch processing.

    Article  CAS  PubMed  Google Scholar 

  93. Valverde, O. et al. GPR3 receptor, a novel actor in the emotional-like responses. PLoS One 4, e4704 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Iismaa, T. P. et al. Isolation and chromosomal localization of a novel human G-protein-coupled receptor (GPR3) expressed predominantly in the central nervous system. Genomics 24, 391–394 (1994).

    Article  CAS  PubMed  Google Scholar 

  95. Tanaka, S., Ishii, K., Kasai, K., Yoon, S. O. & Saeki, Y. Neural Expression of G. Protein-coupled Receptors GPR3, GPR6, and GPR12 Up-regulates Cyclic AMP Levels and Promotes Neurite Outgrowth. J. Biol. Chem. 282, 10506–10515 (2007).

    Article  CAS  PubMed  Google Scholar 

  96. Radde, R. et al. Abeta42-driven cerebral amyloidosis in transgenic mice reveals early and robust pathology. EMBO Rep. 7, 940–946 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Horuk, R. et al. Expression of chemokine receptors by subsets of neurons in the central nervous system. J. Immunol. 158, 2882–2890 (1997).

    CAS  PubMed  Google Scholar 

  98. Xia, M., Qin, S., McNamara, M., Mackay, C. & Hyman, B. T. Interleukin-8 receptor B immunoreactivity in brain and neuritic plaques of Alzheimer's disease. Am. J. Pathol. 150, 1267–1274 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Bakshi, P., Margenthaler, E., Laporte, V., Crawford, F. & Mullan, M. Novel role of CXCR2 in regulation of gamma-secretase activity. ACS Chem. Biol. 3, 777–789 (2008).

    Article  CAS  PubMed  Google Scholar 

  100. Bakshi, P., Margenthaler, E., Reed, J., Crawford, F. & Mullan, M. Depletion of CXCR2 inhibits gamma-secretase activity and amyloid-beta production in a murine model of Alzheimer's disease. Cytokine 15 Nov 2010 (doi:10.1016/j.cyto.2010.10.008) [epub ahead of print].

    Article  CAS  PubMed  Google Scholar 

  101. Liao, Y. F., Wang, B. J., Cheng, H. T., Kuo, L. H. & Wolfe, M. S. Tumor necrosis factor-alpha, interleukin-1beta, and interferon-gamma stimulate gamma-secretase-mediated cleavage of amyloid precursor protein through a JNK-dependent MAPK pathway. J. Biol. Chem. 279, 49523–49532 (2004).

    Article  CAS  PubMed  Google Scholar 

  102. Kuo, L. H. et al. Tumor necrosis factor-alpha-elicited stimulation of gamma-secretase is mediated by c-Jun N-terminal kinase-dependent phosphorylation of presenilin and nicastrin. Mol. Biol. Cell 19, 4201–4212 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Lambert, M. P. et al. Diffusible, nonfibrillar ligands derived from Abeta1–42 are potent central nervous system neurotoxins. Proc. Natl Acad. Sci. USA 95, 6448–6453 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Shenoy, U. V., Richards, E. M., Huang, X. C. & Sumners, C. Angiotensin II type 2 receptor-mediated apoptosis of cultured neurons from newborn rat brain. Endocrinology 140, 500–509 (1999).

    Article  CAS  PubMed  Google Scholar 

  105. Ichiki, T. et al. Effects on blood pressure and exploratory behaviour of mice lacking angiotensin II type-2 receptor. Nature 377, 748–750 (1995).

    Article  CAS  PubMed  Google Scholar 

  106. Vervoort, V. S. et al. AGTR2 mutations in X-linked mental retardation. Science 296, 2401–2403 (2002).

    CAS  PubMed  Google Scholar 

  107. AbdAlla, S., Lother, H., Abdel-tawab, A. M. & Quitterer, U. The angiotensin II AT2 receptor is an AT1 receptor antagonist. J. Biol. Chem. 276, 39721–39726 (2001).

    Article  CAS  PubMed  Google Scholar 

  108. Ferrari-DiLeo, G. & Flynn, D. D. Diminished muscarinic receptor-stimulated [3H]-PIP2 hydrolysis in Alzheimer's disease. Life Sci. 53, PL439–444 (1993).

    Article  CAS  PubMed  Google Scholar 

  109. Tsang, S. W. et al. Impaired coupling of muscarinic M1 receptors to G-proteins in the neocortex is associated with severity of dementia in Alzheimer's disease. Neurobiol. Aging 27, 1216–1223 (2006).

    Article  CAS  PubMed  Google Scholar 

  110. AbdAlla, S. et al. Angiotensin II AT2 receptor oligomers mediate G-protein dysfunction in an animal model of Alzheimer disease. J. Biol. Chem. 284, 6554–6565 (2009).

    Article  CAS  PubMed  Google Scholar 

  111. Thathiah, A. & De Strooper, B. G protein-coupled receptors, cholinergic dysfunction, and Abeta toxicity in Alzheimer's disease. Sci. Signal 2, re8 (2009).

    Article  PubMed  Google Scholar 

  112. Fredholm, B. B., AP, I. J., Jacobson, K. A., Klotz, K. N. & Linden, J. International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol. Rev. 53, 527–552 (2001).

    CAS  PubMed  Google Scholar 

  113. Angulo, E. et al. A1 adenosine receptors accumulate in neurodegenerative structures in Alzheimer disease and mediate both amyloid precursor protein processing and tau phosphorylation and translocation. Brain Pathol. 13, 440–451 (2003).

    Article  CAS  PubMed  Google Scholar 

  114. Albasanz, J. L., Perez, S., Barrachina, M., Ferrer, I. & Martin, M. Up-regulation of adenosine receptors in the frontal cortex in Alzheimer's disease. Brain Pathol. 18, 211–219 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Wang, J. H., Ma, Y. Y. & van den Buuse, M. Improved spatial recognition memory in mice lacking adenosine A2A receptors. Exp. Neurol. 199, 438–445 (2006).

    Article  CAS  PubMed  Google Scholar 

  116. Gimenez-Llort, L. et al. Working memory deficits in transgenic rats overexpressing human adenosine A2A receptors in the brain. Neurobiol. Learn. Mem. 87, 42–56 (2007).

    Article  CAS  PubMed  Google Scholar 

  117. Dall'Igna, O. P., Porciuncula, L. O., Souza, D. O., Cunha, R. A. & Lara, D. R. Neuroprotection by caffeine and adenosine A2A receptor blockade of beta-amyloid neurotoxicity. Br. J. Pharmacol. 138, 1207–1209 (2003).

    Article  CAS  PubMed  Google Scholar 

  118. Dall'Igna, O. P. et al. Caffeine and adenosine A(2a) receptor antagonists prevent beta-amyloid (25–35)-induced cognitive deficits in mice. Exp. Neurol. 203, 241–245 (2007).

    Article  CAS  PubMed  Google Scholar 

  119. Arendash, G. W. et al. Caffeine protects Alzheimer's mice against cognitive impairment and reduces brain beta-amyloid production. Neuroscience 142, 941–952 (2006).

    Article  CAS  PubMed  Google Scholar 

  120. Canas, P. M. et al. Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by beta-amyloid peptides via p38 mitogen-activated protein kinase pathway. J. Neurosci. 29, 14741–14751 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Querfurth, H. W., Jiang, J., Geiger, J. D. & Selkoe, D. J. Caffeine stimulates amyloid beta-peptide release from beta-amyloid precursor protein-transfected HEK293 cells. J. Neurochem. 69, 1580–1591 (1997).

    Article  CAS  PubMed  Google Scholar 

  122. Broad, R. M. & Fredholm, B. B. A1, but not A2A, adenosine receptors modulate electrically stimulated [14C]acetylcholine release from rat cortex. J. Pharmacol. Exp. Ther. 277, 193–197 (1996).

    CAS  PubMed  Google Scholar 

  123. Carter, A. J., O'Connor, W. T., Carter, M. J. & Ungerstedt, U. Caffeine enhances acetylcholine release in the hippocampus in vivo by a selective interaction with adenosine A1 receptors. J. Pharmacol. Exp. Ther. 273, 637–642 (1995).

    CAS  PubMed  Google Scholar 

  124. Frautschy, S. A. et al. Microglial response to amyloid plaques in APPsw transgenic mice. Am. J. Pathol. 152, 307–317 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Babcock, A. A., Kuziel, W. A., Rivest, S. & Owens, T. Chemokine expression by glial cells directs leukocytes to sites of axonal injury in the CNS. J. Neurosci. 23, 7922–7930 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Ishizuka, K. et al. Identification of monocyte chemoattractant protein-1 in senile plaques and reactive microglia of Alzheimer's disease. Psychiatry Clin. Neurosci. 51, 135–138 (1997).

    Article  CAS  PubMed  Google Scholar 

  127. Smits, H. A. et al. Amyloid-beta-induced chemokine production in primary human macrophages and astrocytes. J. Neuroimmunol 127, 160–168 (2002).

    Article  CAS  PubMed  Google Scholar 

  128. El Khoury, J. et al. Ccr2 deficiency impairs microglial accumulation and accelerates progression of Alzheimer-like disease. Nature Med. 13, 432–438 (2007).

    Article  CAS  PubMed  Google Scholar 

  129. Chapman, G. A. et al. Fractalkine cleavage from neuronal membranes represents an acute event in the inflammatory response to excitotoxic brain damage. J. Neurosci. 20, RC87 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Harrison, J. K. et al. Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proc. Natl Acad. Sci. USA 95, 10896–10901 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Fuhrmann, M. et al. Microglial Cx3cr1 knockout prevents neuron loss in a mouse model of Alzheimer's disease. Nature Neurosci. 13, 411–413 (2010).

    Article  CAS  PubMed  Google Scholar 

  132. Grathwohl, S. A. et al. Formation and maintenance of Alzheimer's disease beta-amyloid plaques in the absence of microglia. Nature Neurosci. 12, 1361–1363 (2009).

    Article  CAS  PubMed  Google Scholar 

  133. Epelbaum, J., Dournaud, P., Fodor, M. & Viollet, C. The neurobiology of somatostatin. Crit. Rev. Neurobiol. 8, 25–44 (1994).

    CAS  PubMed  Google Scholar 

  134. Patel, Y. C. Somatostatin and its receptor family. Front. Neuroendocrinol. 20, 157–198 (1999).

    Article  CAS  PubMed  Google Scholar 

  135. Kumar, U. Expression of somatostatin receptor subtypes (SSTR1–5) in Alzheimer's disease brain: an immunohistochemical analysis. Neuroscience 134, 525–538 (2005).

    Article  CAS  PubMed  Google Scholar 

  136. Tamminga, C. A., Foster, N. L., Fedio, P., Bird, E. D. & Chase, T. N. Alzheimer's disease: low cerebral somatostatin levels correlate with impaired cognitive function and cortical metabolism. Neurology 37, 161–165 (1987).

    Article  CAS  PubMed  Google Scholar 

  137. Dournaud, P., Delaere, P., Hauw, J. J. & Epelbaum, J. Differential correlation between neurochemical deficits, neuropathology, and cognitive status in Alzheimer's disease. Neurobiol. Aging 16, 817–823 (1995).

    Article  CAS  PubMed  Google Scholar 

  138. Davies, P., Katzman, R. & Terry, R. D. Reduced somatostatin-like immunoreactivity in cerebral cortex from cases of Alzheimer disease and Alzheimer senile dementa. Nature 288, 279–280 (1980).

    Article  CAS  PubMed  Google Scholar 

  139. Saito, T. et al. Somatostatin regulates brain amyloid beta peptide Abeta42 through modulation of proteolytic degradation. Nature Med. 11, 434–439 (2005). This study demonstrates that somatostatin, a neuropeptide that binds to the SSTRs in the brain, regulates the activity of neprilysin, one of the major amyloid-β-degrading enzymes.

    Article  CAS  PubMed  Google Scholar 

  140. Iwata, N. et al. Metabolic regulation of brain Abeta by neprilysin. Science 292, 1550–1552 (2001).

    Article  CAS  PubMed  Google Scholar 

  141. Ramos, B. et al. Early neuropathology of somatostatin/NPY GABAergic cells in the hippocampus of a PS1xAPP transgenic model of Alzheimer's disease. Neurobiol. Aging 27, 1658–1672 (2006).

    Article  CAS  PubMed  Google Scholar 

  142. Horgan, J., Miguel-Hidalgo, J. J., Thrasher, M. & Bissette, G. Longitudinal brain corticotropin releasing factor and somatostatin in a transgenic mouse (TG2576) model of Alzheimer's disease. J. Alzheimers Dis. 12, 115–127 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Slack, B. E. & Wurtman, R. J. in Research Progress in Alzheimer's Disease and Dementia (ed. Sun, M.-K.) 1–25 (Nova Science Publishers, New York, 2006). This is an excellent review that examines the effects of neurotransmitters, growth factors and cytokines on the synthesis and metabolism of APP.

    Google Scholar 

  144. Lemere, C. A. & Masliah, E. Can Alzheimer disease be prevented by amyloid-beta immunotherapy? Nature Rev. Neurol. 6, 108–119 (2010).

    Article  CAS  Google Scholar 

  145. Selkoe, D. J. The molecular pathology of Alzheimer's disease. Neuron 6, 487–498 (1991).

    Article  CAS  PubMed  Google Scholar 

  146. Hardy, J. A. & Higgins, G. A. Alzheimer's disease: the amyloid cascade hypothesis. Science 256, 184–185 (1992).

    Article  CAS  PubMed  Google Scholar 

  147. Bartus, R. T., Dean, R. L. 3rd, Beer, B. & Lippa, A. S. The cholinergic hypothesis of geriatric memory dysfunction. Science 217, 408–414 (1982).

    Article  CAS  PubMed  Google Scholar 

  148. Davies, P. & Maloney, A. J. Selective loss of central cholinergic neurons in Alzheimer's disease. Lancet 2, 1403 (1976).

    Article  CAS  PubMed  Google Scholar 

  149. Whitehouse, P. J. et al. Alzheimer's disease and senile dementia: loss of neurons in the basal forebrain. Science 215, 1237–1239 (1982).

    Article  CAS  PubMed  Google Scholar 

  150. Woolf, N. J. The critical role of cholinergic basal forebrain neurons in morphological change and memory encoding: a hypothesis. Neurobiol. Learn. Mem. 66, 258–266 (1996).

    Article  CAS  PubMed  Google Scholar 

  151. Tzavara, E. T. et al. Dysregulated hippocampal acetylcholine neurotransmission and impaired cognition in M2, M4 and M2/M4 muscarinic receptor knockout mice. Mol. Psychiatry 8, 673–679 (2003).

    Article  CAS  PubMed  Google Scholar 

  152. Zhang, W. et al. Characterization of central inhibitory muscarinic autoreceptors by the use of muscarinic acetylcholine receptor knock-out mice. J. Neurosci. 22, 1709–1717 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Sadot, E. et al. Activation of m1 muscarinic acetylcholine receptor regulates tau phosphorylation in transfected PC12 cells. J. Neurochem. 66, 877–880 (1996).

    Article  CAS  PubMed  Google Scholar 

  154. Oddo, S. et al. Chronic nicotine administration exacerbates tau pathology in a transgenic model of Alzheimer's disease. Proc. Natl Acad. Sci. USA 102, 3046–3051 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Haan, M. N. Therapy Insight: type 2 diabetes mellitus and the risk of late-onset Alzheimer's disease. Nature Clin. Pract. Neurol. 2, 159–166 (2006).

    Article  CAS  Google Scholar 

  156. Biessels, G. J. et al. Place learning and hippocampal synaptic plasticity in streptozotocin-induced diabetic rats. Diabetes 45, 1259–1266 (1996).

    Article  CAS  PubMed  Google Scholar 

  157. Zhao, W. et al. Brain insulin receptors and spatial memory. Correlated changes in gene expression, tyrosine phosphorylation, and signaling molecules in the hippocampus of water maze trained rats. J. Biol. Chem. 274, 34893–34902 (1999).

    Article  CAS  PubMed  Google Scholar 

  158. Zhao, W. Q., Chen, H., Quon, M. J. & Alkon, D. L. Insulin and the insulin receptor in experimental models of learning and memory. Eur. J. Pharmacol. 490, 71–81 (2004).

    Article  CAS  PubMed  Google Scholar 

  159. Carro, E. & Torres-Aleman, I. The role of insulin and insulin-like growth factor I in the molecular and cellular mechanisms underlying the pathology of Alzheimer's disease. Eur. J. Pharmacol. 490, 127–133 (2004).

    Article  CAS  PubMed  Google Scholar 

  160. Beeri, M. S. et al. Insulin in combination with other diabetes medication is associated with less Alzheimer neuropathology. Neurology 71, 750–757 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Plastino, M. et al. Effects of insulinic therapy on cognitive impairment in patients with Alzheimer disease and diabetes mellitus type-2. J. Neurol. Sci. 288, 112–116 (2010).

    Article  CAS  PubMed  Google Scholar 

  162. Li, L. & Holscher, C. Common pathological processes in Alzheimer disease and type 2 diabetes: a review. Brain Res. Rev. 56, 384–402 (2007).

    Article  CAS  PubMed  Google Scholar 

  163. Abbas, T., Faivre, E. & Holscher, C. Impairment of synaptic plasticity and memory formation in GLP-1 receptor KO mice: Interaction between type 2 diabetes and Alzheimer's disease. Behav. Brain Res. 205, 265–271 (2009).

    Article  CAS  PubMed  Google Scholar 

  164. Perry, T. et al. Glucagon-like peptide-1 decreases endogenous amyloid-beta peptide (Abeta) levels and protects hippocampal neurons from death induced by Abeta and iron. J. Neurosci. Res. 72, 603–612 (2003).

    Article  CAS  PubMed  Google Scholar 

  165. Gault, V. A. & Holscher, C. GLP-1 agonists facilitate hippocampal LTP and reverse the impairment of LTP induced by beta-amyloid. Eur. J. Pharmacol. 587, 112–117 (2008).

    Article  CAS  PubMed  Google Scholar 

  166. Wang, X. H. et al. Val8-glucagon-like peptide-1 protects against Abeta1-40-induced impairment of hippocampal late-phase long-term potentiation and spatial learning in rats. Neuroscience 170, 1239–1248 (2010).

    Article  CAS  PubMed  Google Scholar 

  167. Gao, H. et al. GLP-1 amplifies insulin signaling by up-regulation of IRbeta, IRS-1 and Glut4 in 3T3-L1 adipocytes. Endocrine 32, 90–95 (2007).

    Article  CAS  PubMed  Google Scholar 

  168. Wheeler, M. B. et al. Functional expression of the rat glucagon-like peptide-I receptor, evidence for coupling to both adenylyl cyclase and phospholipase-C. Endocrinology 133, 57–62 (1993).

    Article  CAS  PubMed  Google Scholar 

  169. Montrose-Rafizadeh, C. et al. Pancreatic glucagon-like peptide-1 receptor couples to multiple G proteins and activates mitogen-activated protein kinase pathways in Chinese hamster ovary cells. Endocrinology 140, 1132–1140 (1999).

    Article  CAS  PubMed  Google Scholar 

  170. Buteau, J., Roduit, R., Susini, S. & Prentki, M. Glucagon-like peptide-1 promotes DNA synthesis, activates phosphatidylinositol 3-kinase and increases transcription factor pancreatic and duodenal homeobox gene 1 (PDX-1) DNA binding activity in beta (INS-1)-cells. Diabetologia 42, 856–864 (1999).

    Article  CAS  PubMed  Google Scholar 

  171. Holz, G. G., Leech, C. A. & Habener, J. F. Activation of a cAMP-regulated Ca2+-signaling pathway in pancreatic beta-cells by the insulinotropic hormone glucagon-like peptide-1. J. Biol. Chem. 270, 17749–17757 (1995).

    Article  CAS  PubMed  Google Scholar 

  172. During, M. J. et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nature Med. 9, 1173–1179 (2003).

    Article  CAS  PubMed  Google Scholar 

  173. Cooper, G. J. et al. Purification and characterization of a peptide from amyloid-rich pancreases of type 2 diabetic patients. Proc. Natl Acad. Sci. USA 84, 8628–8632 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Dore, S., Kar, S. & Quirion, R. Insulin-like growth factor I protects and rescues hippocampal neurons against beta-amyloid- and human amylin-induced toxicity. Proc. Natl Acad. Sci. USA 94, 4772–4777 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  175. Jhamandas, J. H. & MacTavish, D. Antagonist of the amylin receptor blocks beta-amyloid toxicity in rat cholinergic basal forebrain neurons. J. Neurosci. 24, 5579–5584 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Takeda, S. et al. Diabetes-accelerated memory dysfunction via cerebrovascular inflammation and Abeta deposition in an Alzheimer mouse model with diabetes. Proc. Natl Acad. Sci. USA 107, 7036–7041 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Nitsch, R. M., Deng, M., Tennis, M., Schoenfeld, D. & Growdon, J. H. The selective muscarinic M1 agonist AF102B decreases levels of total Abeta in cerebrospinal fluid of patients with Alzheimer's disease. Ann. Neurol. 48, 913–918 (2000).

    Article  CAS  PubMed  Google Scholar 

  178. Beach, T. G., Walker, D. G., Potter, P. E., Sue, L. I. & Fisher, A. Reduction of cerebrospinal fluid amyloid beta after systemic administration of M1 muscarinic agonists. Brain Res. 905, 220–223 (2001).

    Article  CAS  PubMed  Google Scholar 

  179. Kirazov, L., Loffler, T., Schliebs, R. & Bigl, V. Glutamate-stimulated secretion of amyloid precursor protein from cortical rat brain slices. Neurochem. Int. 30, 557–563 (1997).

    Article  CAS  PubMed  Google Scholar 

  180. Nitsch, R. M., Deng, A., Wurtman, R. J. & Growdon, J. H. Metabotropic glutamate receptor subtype mGluR1alpha stimulates the secretion of the amyloid beta-protein precursor ectodomain. J. Neurochem. 69, 704–712 (1997).

    Article  CAS  PubMed  Google Scholar 

  181. Maillet, M. et al. Crosstalk between Rap1 and Rac regulates secretion of sAPPalpha. Nature Cell Biol. 5, 633–639 (2003).

    Article  CAS  PubMed  Google Scholar 

  182. Robert, S. J. & Lezoualc'h, F. Distinct functional effects of human 5-HT4 receptor isoforms on beta-amyloid secretion. Neurodegener Dis. 5, 163–165 (2008).

    Article  CAS  PubMed  Google Scholar 

  183. Xia, M. & Hyman, B. T. GROalpha/KC, a chemokine receptor CXCR2 ligand, can be a potent trigger for neuronal ERK1/2 and PI-3 kinase pathways and for tau hyperphosphorylation-a role in Alzheimer's disease? J. Neuroimmunol. 122, 55–64 (2002).

    Article  CAS  PubMed  Google Scholar 

  184. AbdAlla, S. et al. Dominant negative AT2 receptor oligomers induce G-protein arrest and symptoms of neurodegeneration. J. Biol. Chem. 284, 6566–6574 (2009).

    Article  CAS  PubMed  Google Scholar 

  185. Cunha, G. M. et al. Adenosine A2A receptor blockade prevents memory dysfunction caused by beta-amyloid peptides but not by scopolamine or MK-801. Exp. Neurol. 210, 776–781 (2008).

    Article  CAS  PubMed  Google Scholar 

  186. Nordberg, A. et al. Chronic nicotine treatment reduces beta-amyloidosis in the brain of a mouse model of Alzheimer's disease (APPsw). J. Neurochem. 81, 655–658 (2002).

    Article  CAS  PubMed  Google Scholar 

  187. Hellstrom-Lindahl, E. et al. Nicotine reduces A beta in the brain and cerebral vessels of APPsw mice. Eur. J. Neurosci. 19, 2703–2710 (2004).

    Article  PubMed  Google Scholar 

  188. Wang, H. Y., Li, W., Benedetti, N. J. & Lee, D. H. Alpha 7 nicotinic acetylcholine receptors mediate beta-amyloid peptide-induced tau protein phosphorylation. J. Biol. Chem. 278, 31547–31553 (2003).

    Article  CAS  PubMed  Google Scholar 

  189. Dineley, K. T. et al. Beta-amyloid activates the mitogen-activated protein kinase cascade via hippocampal alpha7 nicotinic acetylcholine receptors: in vitro and in vivo mechanisms related to Alzheimer's disease. J. Neurosci. 21, 4125–4133 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Unger, C., Svedberg, M. M., Yu, W. F., Hedberg, M. M. & Nordberg, A. Effect of subchronic treatment of memantine, galantamine, and nicotine in the brain of Tg2576 (APPswe) transgenic mice. J. Pharmacol. Exp. Ther. 317, 30–36 (2006).

    Article  CAS  PubMed  Google Scholar 

  191. Pettit, D. L., Shao, Z. & Yakel, J. L. beta-Amyloid(1–42) peptide directly modulates nicotinic receptors in the rat hippocampal slice. J. Neurosci. 21, RC120 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Dziewczapolski, G., Glogowski, C. M., Masliah, E. & Heinemann, S. F. Deletion of the alpha 7 nicotinic acetylcholine receptor gene improves cognitive deficits and synaptic pathology in a mouse model of Alzheimer's disease. J. Neurosci. 29, 8805–8815 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Ray, B., Banerjee, P. K., Greig, N. H. & Lahiri, D. K. Memantine treatment decreases levels of secreted Alzheimer's amyloid precursor protein (APP) and amyloid beta (A beta) peptide in the human neuroblastoma cells. Neurosci. Lett. 470, 1–5 (2010).

    Article  CAS  PubMed  Google Scholar 

  194. Alley, G. M. et al. Memantine lowers amyloid-beta peptide levels in neuronal cultures and in APP/PS1 transgenic mice. J. Neurosci. Res. 88, 143–154 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  195. Minkeviciene, R., Banerjee, P. & Tanila, H. Memantine improves spatial learning in a transgenic mouse model of Alzheimer's disease. J. Pharmacol. Exp. Ther. 311, 677–682 (2004).

    Article  CAS  PubMed  Google Scholar 

  196. Scholtzova, H. et al. Memantine leads to behavioral improvement and amyloid reduction in Alzheimer's-disease-model transgenic mice shown as by micromagnetic resonance imaging. J. Neurosci. Res. 86, 2784–2791 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Brandenburg, L. O. et al. Involvement of formyl-peptide-receptor-like-1 and phospholipase D in the internalization and signal transduction of amyloid beta 1–42 in glial cells. Neuroscience 156, 266–276 (2008).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Work in the laboratory was supported by the Flanders Institute for Biotechnology (VIB), Fonds voor Wetenschappelijk onderzoek (FWO) and Stichting Alzheimer Onderzoek-Fondation pour la recherche de la maladie d'Alzheimer (SAO-FRMA) (grant cycle 2008–2009), the Federal Office for Scientific Affairs, Belgium (IUAP P6/43/), a Methusalem grant of the Catholic University of Leuven (KUL) and the Flemish Government, and Memosad (FZ-2007-200611) of the European Union. B.D.S. is supported by an Arthur Bax and Anna Vanluffelen Chair for Alzheimer's Disease.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Amantha Thathiah or Bart De Strooper.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Glossary

Synaptoneurosome

A purified synapse, containing a presynaptic sac (synaptosome) attached to a resealed postsynaptic sac (neurosome), that is modestly enriched for synaptic proteins.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Thathiah, A., De Strooper, B. The role of G protein-coupled receptors in the pathology of Alzheimer's disease. Nat Rev Neurosci 12, 73–87 (2011). https://doi.org/10.1038/nrn2977

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn2977

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research