Skip to main content
Log in

Neuroprotective Actions of Glucagon-Like Peptide-1 (GLP-1) Analogues in Alzheimer’s and Parkinson’s Diseases

  • Leading Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

The current absence of effective treatments for Alzheimer’s disease (AD) and Parkinson’s disease (PD) reflects an incomplete knowledge of the underlying disease processes. Considerable efforts have been made to investigate the central pathological features of these diseases, giving rise to numerous attempts to develop compounds that interfere with such features. However, further characterization of the molecular targets within the interconnected AD and PD pathways is still required. Impaired brain insulin signaling has emerged as a feature that contributes to neuronal dysfunction in both AD and PD, leading to strategies aiming at restoring this pathway in the brain. Long-acting glucagon-like peptide-1 (GLP-1) analogues marketed for treatment of type 2 diabetes mellitus have been tested and have shown encouraging protective actions in experimental models of AD and PD as well as in initial clinical trials. We review studies revealing the neuroprotective actions of GLP-1 analogues in pre-clinical models of AD and PD and promising results from recent clinical trials.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. International Diabetes Federation. IDF Diabetes Atlas. 7th ed. Brussels: International Diabetes Federation; 2015.

    Google Scholar 

  2. United Nations, Department of Economic and Social Affairs, population division 2015. World population propects: the 2015 revision, key findings and advance tables.

  3. Nussbaum RL, Ellis CE. Alzheimer’s disease and Parkinson’s disease. N Engl J Med. 2003;348(14):1356–64.

    Article  CAS  PubMed  Google Scholar 

  4. Athauda D, Foltynie T. Challenges in detecting disease modification in Parkinson’s disease clinical trials. Parkinsonism Relat Disord. 2016;32:1–11.

    Article  PubMed  Google Scholar 

  5. Selkoe DJ. Resolving controversies on the path to Alzheimer’s therapeutics. Nat Med. 2011;17(9):1060–5.

    Article  CAS  PubMed  Google Scholar 

  6. Arnold SE, Arvanitakis Z, Macauley-Rambach SL, Koenig AM, Wang HY, Ahima RS, et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol. 2018;14(3):168–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Aviles-Olmos I, Limousin P, Lees A, Foltynie T. Parkinson’s disease, insulin resistance and novel agents of neuroprotection. Brain. 2013;136(Pt 2):374–84.

    Article  PubMed  Google Scholar 

  8. Foltynie T, Kahan J. Parkinson’s disease: an update on pathogenesis and treatment. J Neurol. 2013;260(5):1433–40.

    Article  CAS  PubMed  Google Scholar 

  9. Holscher C. Novel dual GLP-1/GIP receptor agonists show neuroprotective effects in Alzheimer’s and Parkinson’s disease models. Neuropharmacology. 2018;136(Pt B):251–259.

    Article  CAS  PubMed  Google Scholar 

  10. De Felice FG, Ferreira ST. Inflammation, defective insulin signaling, and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to Alzheimer disease. Diabetes. 2014;63(7):2262–72.

    Article  PubMed  Google Scholar 

  11. Ferreira ST, Clarke JR, Bomfim TR, De Felice FG. Inflammation, defective insulin signaling, and neuronal dysfunction in Alzheimer’s disease. Alzheimers Dement. 2014;10(1 Suppl):S76–83.

    Article  PubMed  Google Scholar 

  12. Saltiel AR, Kahn CR. Insulin signalling and the regulation of glucose and lipid metabolism. Nature. 2001;414(6865):799–806.

    Article  CAS  PubMed  Google Scholar 

  13. Ward CW, Menting JG, Lawrence MC. The insulin receptor changes conformation in unforeseen ways on ligand binding: sharpening the picture of insulin receptor activation. Bioessays. 2013;35(11):945–54. https://doi.org/10.1002/bies.201370111.

    Article  CAS  PubMed  Google Scholar 

  14. Menting JG, Whittaker J, Margetts MB, Whittaker LJ, Kong GK, Smith BJ, et al. How insulin engages its primary binding site on the insulin receptor. Nature. 2013;493(7431):241–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ghasemi R, Haeri A, Dargahi L, Mohamed Z, Ahmadiani A. Insulin in the brain: sources, localization and functions. Mol Neurobiol. 2013;47(1):145–71.

    Article  CAS  PubMed  Google Scholar 

  16. Haeusler RA, McGraw TE, Accili D. Biochemical and cellular properties of insulin receptor signalling. Nat Rev Mol Cell Biol. 2018;19(1):31–44.

    Article  CAS  PubMed  Google Scholar 

  17. Taniguchi CM, Emanuelli B, Kahn CR. Critical nodes in signalling pathways: insights into insulin action. Nat Rev Mol Cell Biol. 2006;7(2):85–96.

    Article  CAS  PubMed  Google Scholar 

  18. Banks WA, Jaspan JB, Huang W, Kastin AJ. Transport of insulin across the blood-brain barrier: saturability at euglycemic doses of insulin. Peptides. 1997;18(9):1423–9.

    Article  CAS  PubMed  Google Scholar 

  19. Banks WA, Owen JB, Erickson MA. Insulin in the brain: there and back again. Pharmacol Ther. 2012;136(1):82–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Ferrario CR, Reagan LP. Insulin-mediated synaptic plasticity in the CNS: anatomical, functional and temporal contexts. Neuropharmacology. 2017;Dec:5.

    Google Scholar 

  21. Gray SM, Meijer RI, Barrett EJ. Insulin regulates brain function, but how does it get there? Diabetes. 2014;63(12):3992–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Erbsloh F, Bernsmeier A, Hillesheim H. The glucose consumption of the brain and its dependence on the liver. Arch Psychiatr Nervenkr Z Gesamte Neurol Psychiatr. 1958;196(6):611–26.

    Article  CAS  PubMed  Google Scholar 

  23. Debons AF, Krimsky I, From A, Cloutier RJ. Rapid effects of insulin on the hypothalamic satiety center. Am J Physiol. 1969;217(4):1114–8.

    Article  CAS  PubMed  Google Scholar 

  24. Thon M, Hosoi T, Ozawa K. Possible integrative actions of leptin and insulin signaling in the hypothalamus targeting energy homeostasis. Front Endocrinol (Lausanne). 2016;7:138.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Havrankova J, Roth J, Brownstein M. Insulin receptors are widely distributed in the central nervous system of the rat. Nature. 1978;272(5656):827–9.

    Article  CAS  PubMed  Google Scholar 

  26. Werther GA, Hogg A, Oldfield BJ, McKinley MJ, Figdor R, Allen AM, et al. Localization and characterization of insulin receptors in rat brain and pituitary gland using in vitro autoradiography and computerized densitometry. Endocrinology. 1987;121(4):1562–70.

    Article  CAS  PubMed  Google Scholar 

  27. Zhao W, Chen H, Xu H, Moore E, Meiri N, Quon MJ, 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. 1999;274(49):34893–902.

    Article  CAS  PubMed  Google Scholar 

  28. Wickelgren I. Tracking insulin to the mind. Science. 1998;280(5363):517–9.

    Article  CAS  PubMed  Google Scholar 

  29. Biessels GJ, Reagan LP. Hippocampal insulin resistance and cognitive dysfunction. Nat Rev Neurosci. 2015;16(11):660–71.

    Article  CAS  PubMed  Google Scholar 

  30. Chiu SL, Chen CM, Cline HT. Insulin receptor signaling regulates synapse number, dendritic plasticity, and circuit function in vivo. Neuron. 2008;58(5):708–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Grillo CA, Piroli GG, Lawrence RC, Wrighten SA, Green AJ, Wilson SP, et al. Hippocampal insulin resistance impairs spatial learning and synaptic plasticity. Diabetes. 2015;64(11):3927–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Ott A, Stolk RP, van Harskamp F, Pols HA, Hofman A, Breteler MM. Diabetes mellitus and the risk of dementia: the Rotterdam Study. Neurology. 1999;53(9):1937–42.

    Article  CAS  PubMed  Google Scholar 

  33. Wang KC, Woung LC, Tsai MT, Liu CC, Su YH, Li CY. Risk of Alzheimer’s disease in relation to diabetes: a population-based cohort study. Neuroepidemiology. 2012;38(4):237–44.

    Article  PubMed  Google Scholar 

  34. Yang YW, Hsieh TF, Li CI, Liu CS, Lin WY, Chiang JH, et al. Increased risk of Parkinson disease with diabetes mellitus in a population-based study. Medicine (Baltimore). 2017;96(3):e5921.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Leibson CL, Rocca WA, Hanson VA, Cha R, Kokmen E, O’Brien PC, et al. Risk of dementia among persons with diabetes mellitus: a population-based cohort study. Am J Epidemiol. 1997;145(4):301–8.

    Article  CAS  PubMed  Google Scholar 

  36. Pagano G, Polychronis S, Wilson H, Giordano B, Ferrara N, Niccolini F, et al. Diabetes mellitus and Parkinson disease. Neurology. 2018;90(19):e1654–62.

    Article  PubMed  Google Scholar 

  37. De Pablo-Fernandez E, Goldacre R, Pakpoor J, Noyce AJ, Warner TT. Association between diabetes and subsequent Parkinson disease: a record-linkage cohort study. Neurology. 2018;91(2):e139–42.

    Article  PubMed  Google Scholar 

  38. Janson J, Laedtke T, Parisi JE, O’Brien P, Petersen RC, Butler PC. Increased risk of type 2 diabetes in Alzheimer disease. Diabetes. 2004;53(2):474–81.

    Article  CAS  PubMed  Google Scholar 

  39. Simon KC, Chen H, Schwarzschild M, Ascherio A. Hypertension, hypercholesterolemia, diabetes, and risk of Parkinson disease. Neurology. 2007;69(17):1688–95.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Becker C, Brobert GP, Johansson S, Jick SS, Meier CR. Diabetes in patients with idiopathic Parkinson’s disease. Diabetes Care. 2008;31(9):1808–12.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Reaven GM. Insulin resistance in noninsulin-dependent diabetes mellitus. Does it exist and can it be measured? Am J Med. 1983;74(1A):3–17.

    Article  CAS  PubMed  Google Scholar 

  42. Greenhill C. Diabetes: new mechanism for insulin resistance. Nat Rev Endocrinol. 2016;12(5):249.

    PubMed  Google Scholar 

  43. Mooradian AD. Central nervous system complications of diabetes mellitus—a perspective from the blood–brain barrier. Brain Res Brain Res Rev. 1997;23(3):210–8.

    Article  CAS  PubMed  Google Scholar 

  44. Acharya NK, Levin EC, Clifford PM, Han M, Tourtellotte R, Chamberlain D, et al. Diabetes and hypercholesterolemia increase blood-brain barrier permeability and brain amyloid deposition: beneficial effects of the LpPLA2 inhibitor darapladib. J Alzheimers Dis. 2013;35(1):179–98.

    Article  CAS  PubMed  Google Scholar 

  45. Sanna PP, Cammalleri M, Berton F, Simpson C, Lutjens R, Bloom FE, et al. Phosphatidylinositol 3-kinase is required for the expression but not for the induction or the maintenance of long-term potentiation in the hippocampal CA1 region. J Neurosci. 2002;22(9):3359–65.

    Article  CAS  PubMed  Google Scholar 

  46. Craft S. The role of metabolic disorders in Alzheimer disease and vascular dementia: two roads converged. Arch Neurol. 2009;66(3):300–5.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Luchsinger JA, Reitz C, Honig LS, Tang MX, Shea S, Mayeux R. Aggregation of vascular risk factors and risk of incident Alzheimer disease. Neurology. 2005;65(4):545–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Haan MN, Wallace R. Can dementia be prevented? Brain aging in a population-based context. Annu Rev Public Health. 2004;25:1–24.

    Article  PubMed  Google Scholar 

  49. Biessels GJ, Staekenborg S, Brunner E, Brayne C, Scheltens P. Risk of dementia in diabetes mellitus: a systematic review. Lancet Neurol. 2006;5(1):64–74.

    Article  PubMed  Google Scholar 

  50. Sperling RA, Aisen PS, Beckett LA, Bennett DA, Craft S, Fagan AM, et al. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7(3):280–92.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Small GW, Ercoli LM, Silverman DH, Huang SC, Komo S, Bookheimer SY, et al. Cerebral metabolic and cognitive decline in persons at genetic risk for Alzheimer’s disease. Proc Natl Acad Sci USA. 2000;97(11):6037–42.

    Article  CAS  PubMed  Google Scholar 

  52. Reiman EM, Caselli RJ, Yun LS, Chen K, Bandy D, Minoshima S, et al. Preclinical evidence of Alzheimer’s disease in persons homozygous for the epsilon 4 allele for apolipoprotein E. N Engl J Med. 1996;334(12):752–8.

    Article  CAS  PubMed  Google Scholar 

  53. Steen E, Terry BM, Rivera EJ, Cannon JL, Neely TR, Tavares R, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease–is this type 3 diabetes? J Alzheimers Dis. 2005;7(1):63–80.

    Article  CAS  PubMed  Google Scholar 

  54. Moloney AM, Griffin RJ, Timmons S, O’Connor R, Ravid R, O’Neill C. Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease indicate possible resistance to IGF-1 and insulin signalling. Neurobiol Aging. 2010;31(2):224–43.

    Article  CAS  PubMed  Google Scholar 

  55. Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, et al. Demonstrated brain insulin resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Investig. 2012;122(4):1316–38.

    Article  CAS  PubMed  Google Scholar 

  56. Matsuzaki T, Sasaki K, Tanizaki Y, Hata J, Fujimi K, Matsui Y, et al. Insulin resistance is associated with the pathology of Alzheimer disease: the Hisayama study. Neurology. 2010;75(9):764–70.

    Article  CAS  PubMed  Google Scholar 

  57. Lee S, Tong M, Hang S, Deochand C, de la Monte S. CSF and brain indices of insulin resistance, oxidative stress and neuro-inflammation in early versus late Alzheimer’s disease. J Alzheimers Dis Parkinsonism. 2013;31(3):128.

    Google Scholar 

  58. Gong Y, Chang L, Viola KL, Lacor PN, Lambert MP, Finch CE, et al. Alzheimer’s disease-affected brain: presence of oligomeric A beta ligands (ADDLs) suggests a molecular basis for reversible memory loss. Proc Natl Acad Sci USA. 2003;100(18):10417–22.

    Article  CAS  PubMed  Google Scholar 

  59. Ferreira ST, Klein WL. The Abeta oligomer hypothesis for synapse failure and memory loss in Alzheimer’s disease. Neurobiol Learn Mem. 2011;96(4):529–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med. 2016;8(6):595–608.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ferreira ST, Lourenco MV, Oliveira MM, De Felice FG. Soluble amyloid-beta oligomers as synaptotoxins leading to cognitive impairment in Alzheimer’s disease. Front Cell Neurosci. 2015;9:191.

    PubMed  PubMed Central  Google Scholar 

  62. De Felice FG, Vieira MN, Bomfim TR, Decker H, Velasco PT, Lambert MP, et al. Protection of synapses against Alzheimer’s-linked toxins: insulin signaling prevents the pathogenic binding of Abeta oligomers. Proc Natl Acad Sci USA. 2009;106(6):1971–6.

    Article  PubMed  Google Scholar 

  63. Zhao WQ, De Felice FG, Fernandez S, Chen H, Lambert MP, Quon MJ, et al. Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J. 2008;22(1):246–60.

    Article  CAS  PubMed  Google Scholar 

  64. Qiao LY, Goldberg JL, Russell JC, Sun XJ. Identification of enhanced serine kinase activity in insulin resistance. J Biol Chem. 1999;274(15):10625–32.

    Article  CAS  PubMed  Google Scholar 

  65. Paz K, Hemi R, LeRoith D, Karasik A, Elhanany E, Kanety H, et al. A molecular basis for insulin resistance. Elevated serine/threonine phosphorylation of IRS-1 and IRS-2 inhibits their binding to the juxtamembrane region of the insulin receptor and impairs their ability to undergo insulin-induced tyrosine phosphorylation. J Biol Chem. 1997;272(47):29911–8.

    Article  CAS  PubMed  Google Scholar 

  66. Lourenco MV, Clarke JR, Frozza RL, Bomfim TR, Forny-Germano L, Batista AF, et al. TNF-alpha mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s beta-amyloid oligomers in mice and monkeys. Cell Metab. 2013;18(6):831–43.

    Article  CAS  PubMed  Google Scholar 

  67. Bomfim TR, Forny-Germano L, Sathler LB, Brito-Moreira J, Houzel JC, Decker H, et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease- associated Abeta oligomers. J Clin Investig. 2012;122(4):1339–53.

    Article  CAS  PubMed  Google Scholar 

  68. Ma QL, Yang F, Rosario ER, Ubeda OJ, Beech W, Gant DJ, et al. Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: suppression by omega-3 fatty acids and curcumin. J Neurosci. 2009;29(28):9078–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science. 1996;271(5249):665–8.

    Article  CAS  PubMed  Google Scholar 

  70. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011;29:415–45.

    Article  CAS  PubMed  Google Scholar 

  71. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14(4):388–405.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542(7640):177–85.

    Article  CAS  PubMed  Google Scholar 

  73. De Felice FG, Lourenco MV, Ferreira ST. How does brain insulin resistance develop in Alzheimer’s disease? Alzheimers Dement. 2014;10(1 Suppl):S26–32.

    Article  PubMed  Google Scholar 

  74. Makki K, Froguel P, Wolowczuk I. Adipose tissue in obesity-related inflammation and insulin resistance: cells, cytokines, and chemokines. ISRN Inflamm. 2013;22(2013):139239.

    Google Scholar 

  75. Piya MK, McTernan PG, Kumar S. Adipokine inflammation and insulin resistance: the role of glucose, lipids and endotoxin. J Endocrinol. 2013;216(1):T1–15.

    Article  CAS  PubMed  Google Scholar 

  76. Roytblat L, Rachinsky M, Fisher A, Greemberg L, Shapira Y, Douvdevani A, et al. Raised interleukin-6 levels in obese patients. Obes Res. 2000;8(9):673–5.

    Article  CAS  PubMed  Google Scholar 

  77. Eder K, Baffy N, Falus A, Fulop AK. The major inflammatory mediator interleukin-6 and obesity. Inflamm Res. 2009;58(11):727–36.

    Article  CAS  PubMed  Google Scholar 

  78. Di Renzo L, Bigioni M, Del Gobbo V, Premrov MG, Barbini U, Di Lorenzo N, et al. Interleukin-1 (IL-1) receptor antagonist gene polymorphism in normal weight obese syndrome: relationship to body composition and IL-1 alpha and beta plasma levels. Pharmacol Res. 2007;55(2):131–8.

    Article  CAS  PubMed  Google Scholar 

  79. Hoene M, Weigert C. The role of interleukin-6 in insulin resistance, body fat distribution and energy balance. Obes Rev. 2008;9(1):20–9.

    CAS  PubMed  Google Scholar 

  80. Jager J, Gremeaux T, Cormont M, Le Marchand-Brustel Y, Tanti JF. Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression. Endocrinology. 2007;148(1):241–51.

    Article  CAS  PubMed  Google Scholar 

  81. Rehman K, Akash MS. Mechanisms of inflammatory responses and development of insulin resistance: how are they interlinked? J Biomed Sci. 2016;23(1):87.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Cnop M, Foufelle F, Velloso LA. Endoplasmic reticulum stress, obesity and diabetes. Trends Mol Med. 2012;18(1):59–68.

    Article  CAS  PubMed  Google Scholar 

  83. Ozcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi NN, Ozdelen E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science. 2004;306(5695):457–61.

    Article  CAS  PubMed  Google Scholar 

  84. Swardfager W, Lanctot K, Rothenburg L, Wong A, Cappell J, Herrmann N. A meta-analysis of cytokines in Alzheimer’s disease. Biol Psychiatry. 2010;68(10):930–41.

    Article  CAS  PubMed  Google Scholar 

  85. Brosseron F, Traschutz A, Widmann CN, Kummer MP, Tacik P, Santarelli F, et al. Characterization and clinical use of inflammatory cerebrospinal fluid protein markers in Alzheimer’s disease. Alzheimers Res Ther. 2018;10(1):25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Hoozemans JJ, van Haastert ES, Nijholt DA, Rozemuller AJ, Eikelenboom P, Scheper W. The unfolded protein response is activated in pretangle neurons in Alzheimer’s disease hippocampus. Am J Pathol. 2009;174(4):1241–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Ledo JH, Azevedo EP, Beckman D, Ribeiro FC, Santos LE, Razolli DS, et al. Cross talk between brain innate immunity and serotonin signaling underlies depressive-like behavior induced by Alzheimer’s amyloid-beta oligomers in mice. J Neurosci. 2016;36(48):12106–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Ledo JH, Azevedo EP, Clarke JR, Ribeiro FC, Figueiredo CP, Foguel D, et al. Amyloid-beta oligomers link depressive-like behavior and cognitive deficits in mice. Mol Psychiatry. 2013;18(10):1053–4.

    Article  CAS  PubMed  Google Scholar 

  89. Abisambra JF, Jinwal UK, Blair LJ, O’Leary JC 3rd, Li Q, Brady S, et al. Tau accumulation activates the unfolded protein response by impairing endoplasmic reticulum-associated degradation. J Neurosci. 2013;33(22):9498–507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Scheper W, Hoozemans JJ. Endoplasmic reticulum protein quality control in neurodegenerative disease: the good, the bad and the therapy. Curr Med Chem. 2009;16(5):615–26.

    Article  CAS  PubMed  Google Scholar 

  91. Shoji M, Iwakami N, Takeuchi S, Waragai M, Suzuki M, Kanazawa I, et al. JNK activation is associated with intracellular beta-amyloid accumulation. Brain Res Mol Brain Res. 2000;85(1–2):221–33.

    Article  CAS  PubMed  Google Scholar 

  92. Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015;386(9996):896–912.

    Article  CAS  PubMed  Google Scholar 

  93. Goldman JG, Williams-Gray C, Barker RA, Duda JE, Galvin JE. The spectrum of cognitive impairment in Lewy body diseases. Mov Disord. 2014;29(5):608–21.

    Article  PubMed  PubMed Central  Google Scholar 

  94. Broeders M, Velseboer DC, de Bie R, Speelman JD, Muslimovic D, Post B, et al. Cognitive change in newly-diagnosed patients with Parkinson’s disease: a 5-year follow-up study. J Int Neuropsychol Soc. 2013;19(6):695–708.

    Article  PubMed  Google Scholar 

  95. Hoogland J, Boel JA, de Bie RMA, Geskus RB, Schmand BA, Dalrymple-Alford JC, et al. Mild cognitive impairment as a risk factor for Parkinson’s disease dementia. Mov Disord. 2017;32(7):1056–65.

    Article  PubMed  Google Scholar 

  96. Athauda D, Foltynie T. Protective effects of the GLP-1 mimetic exendin-4 in Parkinson’s disease. Neuropharmacology. 2017;Sep:18.

    Google Scholar 

  97. Pressley JC, Louis ED, Tang MX, Cote L, Cohen PD, Glied S, et al. The impact of comorbid disease and injuries on resource use and expenditures in parkinsonism. Neurology. 2003;60(1):87–93.

    Article  CAS  PubMed  Google Scholar 

  98. Xu Q, Park Y, Huang X, Hollenbeck A, Blair A, Schatzkin A, et al. Diabetes and risk of Parkinson’s disease. Diabetes Care. 2011;34(4):910–5.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Chalmanov V, Vurbanova M. Diabetes mellitus in parkinsonism patients. Vutr Boles. 1987;26(1):68–73.

    CAS  PubMed  Google Scholar 

  100. Schwab RS. Progression and prognosis in Parkinson’s disease. J Nerv Ment Dis. 1960;130:556–66.

    Article  CAS  PubMed  Google Scholar 

  101. Van Woert MH, Mueller PS. Glucose, insulin, and free fatty acid metabolism in Parkinson’s disease treated with levodopa. Clin Pharmacol Ther. 1971;12(2):360–7.

    Article  PubMed  Google Scholar 

  102. Sirtori CR, Bolme P, Azarnoff DL. Metabolic responses to acute and chronic L-dopa administration in patients with parkinsonism. N Engl J Med. 1972;287(15):729–33.

    Article  CAS  PubMed  Google Scholar 

  103. Van Woert MH, Mueller PS, Ambani LM, Rathey U. Abnormal insulin secretion in Parkinson’s disease before and during L-dihydroxyphenylalanine (L-dopa) therapy. J Endocrinol. 1973;59(3):523–34.

    Article  PubMed  Google Scholar 

  104. Levy G, Tang MX, Cote LJ, Louis ED, Alfaro B, Mejia H, et al. Do risk factors for Alzheimer’s disease predict dementia in Parkinson’s disease? An exploratory study. Mov Disord. 2002;17(2):250–7.

    Article  PubMed  Google Scholar 

  105. Liepelt I, Reimold M, Maetzler W, Godau J, Reischl G, Gaenslen A, et al. Cortical hypometabolism assessed by a metabolic ratio in Parkinson’s disease primarily reflects cognitive deterioration-[18F]FDG-PET. Mov Disord. 2009;24(10):1504–11.

    Article  PubMed  Google Scholar 

  106. Dunn L, Allen GF, Mamais A, Ling H, Li A, Duberley KE, et al. Dysregulation of glucose metabolism is an early event in sporadic Parkinson’s disease. Neurobiol Aging. 2014;35(5):1111–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Unger JW, Moss AM, Livingston JN. Immunohistochemical localization of insulin receptors and phosphotyrosine in the brainstem of the adult rat. Neuroscience. 1991;42(3):853–61.

    Article  CAS  PubMed  Google Scholar 

  108. Moroo I, Yamada T, Makino H, Tooyama I, McGeer PL, McGeer EG, et al. Loss of insulin receptor immunoreactivity from the substantia nigra pars compacta neurons in Parkinson’s disease. Acta Neuropathol. 1994;87(4):343–8.

    Article  CAS  PubMed  Google Scholar 

  109. Liu S, Labouebe G, Karunakaran S, Clee SM, Borgland SL. Effect of insulin on excitatory synaptic transmission onto dopamine neurons of the ventral tegmental area in a mouse model of hyperinsulinemia. Nutr Diabetes. 2013;16(3):e97.

    Article  CAS  Google Scholar 

  110. Godau J, Knauel K, Weber K, Brockmann K, Maetzler W, Binder G, et al. Serum insulinlike growth factor 1 as possible marker for risk and early diagnosis of Parkinson disease. Arch Neurol. 2011;68(7):925–31.

    Article  PubMed  Google Scholar 

  111. Ma J, Jiang Q, Xu J, Sun Q, Qiao Y, Chen W, et al. Plasma insulin-like growth factor 1 is associated with cognitive impairment in Parkinson’s disease. Dement Geriatr Cogn Disord. 2015;39(5–6):251–6.

    Article  CAS  PubMed  Google Scholar 

  112. Mashayekhi F, Mirzajani E, Naji M, Azari M. Expression of insulin-like growth factor-1 and insulin-like growth factor binding proteins in the serum and cerebrospinal fluid of patients with Parkinson’s disease. J Clin Neurosci. 2010;17(5):623–7.

    Article  CAS  PubMed  Google Scholar 

  113. Morris JK, Bomhoff GL, Gorres BK, Davis VA, Kim J, Lee PP, et al. Insulin resistance impairs nigrostriatal dopamine function. Exp Neurol. 2011;231(1):171–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Morris JK, Bomhoff GL, Stanford JA, Geiger PC. Neurodegeneration in an animal model of Parkinson’s disease is exacerbated by a high-fat diet. Am J Physiol Regul Integr Comp Physiol. 2010;299(4):R1082–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Torres-Aleman I. Toward a comprehensive neurobiology of IGF-I. Dev Neurobiol. 2010;70(5):384–96.

    CAS  PubMed  Google Scholar 

  116. Skow MA, Bergmann NC, Knop FK. Diabetes and obesity treatment based on dual incretin receptor activation: ‘twincretins’. Diabetes Obes Metab. 2016;18(9):847–54.

    Article  CAS  PubMed  Google Scholar 

  117. Nauck M. Incretin therapies: highlighting common features and differences in the modes of action of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Diabetes Obes Metab. 2016;18(3):203–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Varndell IM, Bishop AE, Sikri KL, Uttenthal LO, Bloom SR, Polak JM. Localization of glucagon-like peptide (GLP) immunoreactants in human gut and pancreas using light and electron microscopic immunocytochemistry. J Histochem Cytochem. 1985;33(10):1080–6.

    Article  CAS  PubMed  Google Scholar 

  119. Deacon CF, Nauck MA, Toft-Nielsen M, Pridal L, Willms B, Holst JJ. Both subcutaneously and intravenously administered glucagon-like peptide I are rapidly degraded from the NH2-terminus in type II diabetic patients and in healthy subjects. Diabetes. 1995;44(9):1126–31.

    Article  CAS  PubMed  Google Scholar 

  120. Deacon CF, Johnsen AH, Holst JJ. Degradation of glucagon-like peptide-1 by human plasma in vitro yields an N-terminally truncated peptide that is a major endogenous metabolite in vivo. J Clin Endocrinol Metab. 1995;80(3):952–7.

    CAS  PubMed  Google Scholar 

  121. Holst JJ. The physiology of glucagon-like peptide 1. Physiol Rev. 2007;87(4):1409–39.

    Article  CAS  PubMed  Google Scholar 

  122. Kjems LL, Holst JJ, Volund A, Madsbad S. The influence of GLP-1 on glucose-stimulated insulin secretion: effects on beta-cell sensitivity in type 2 and nondiabetic subjects. Diabetes. 2003;52(2):380–6.

    Article  CAS  PubMed  Google Scholar 

  123. Larsen PJ, Tang-Christensen M, Holst JJ, Orskov C. Distribution of glucagon-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuroscience. 1997;77(1):257–70.

    Article  CAS  PubMed  Google Scholar 

  124. Llewellyn-Smith IJ, Reimann F, Gribble FM, Trapp S. Preproglucagon neurons project widely to autonomic control areas in the mouse brain. Neuroscience. 2011;28(180):111–21.

    Article  CAS  Google Scholar 

  125. Merchenthaler I, Lane M, Shughrue P. Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system. J Comp Neurol. 1999;403(2):261–80.

    Article  CAS  PubMed  Google Scholar 

  126. Cork SC, Richards JE, Holt MK, Gribble FM, Reimann F, Trapp S. Distribution and characterisation of Glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol Metab. 2015;4(10):718–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Heppner KM, Kirigiti M, Secher A, Paulsen SJ, Buckingham R, Pyke C, et al. Expression and distribution of glucagon-like peptide-1 receptor mRNA, protein and binding in the male nonhuman primate (Macaca mulatta) brain. Endocrinology. 2015;156(1):255–67.

    Article  CAS  PubMed  Google Scholar 

  128. Farr OM, Sofopoulos M, Tsoukas MA, Dincer F, Thakkar B, Sahin-Efe A, et al. GLP-1 receptors exist in the parietal cortex, hypothalamus and medulla of human brains and the GLP-1 analogue liraglutide alters brain activity related to highly desirable food cues in individuals with diabetes: a crossover, randomised, placebo-controlled trial. Diabetologia. 2016;59(5):954–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Llewellyn-Smith IJ, Gnanamanickam GJ, Reimann F, Gribble FM, Trapp S. Preproglucagon (PPG) neurons innervate neurochemically identified autonomic neurons in the mouse brainstem. Neuroscience. 2013;15(229):130–43.

    Article  CAS  Google Scholar 

  130. Vrang N, Hansen M, Larsen PJ, Tang-Christensen M. Characterization of brainstem preproglucagon projections to the paraventricular and dorsomedial hypothalamic nuclei. Brain Res. 2007;29(1149):118–26.

    Article  CAS  Google Scholar 

  131. During MJ, Cao L, Zuzga DS, Francis JS, Fitzsimons HL, Jiao X, et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med. 2003;9(9):1173–9.

    Article  CAS  PubMed  Google Scholar 

  132. 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. 2009;205(1):265–71.

    Article  CAS  PubMed  Google Scholar 

  133. Mattson MP, Perry T, Greig NH. Learning from the gut. Nat Med. 2003;9(9):1113–5.

    Article  CAS  PubMed  Google Scholar 

  134. Ritzel U, Leonhardt U, Ottleben M, Ruhmann A, Eckart K, Spiess J, et al. A synthetic glucagon-like peptide-1 analog with improved plasma stability. J Endocrinol. 1998;159(1):93–102.

    Article  CAS  PubMed  Google Scholar 

  135. Werner U, Haschke G, Herling AW, Kramer W. Pharmacological profile of lixisenatide: A new GLP-1 receptor agonist for the treatment of type 2 diabetes. Regul Pept. 2010;164(2–3):58–64.

    Article  CAS  PubMed  Google Scholar 

  136. Uccellatore A, Genovese S, Dicembrini I, Mannucci E, Ceriello A. Comparison review of short-acting and long-acting glucagon-like peptide-1 receptor agonists. Diabetes Ther. 2015;6(3):239–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Andersen A, Lund A, Knop FK, Vilsboll T. Glucagon-like peptide 1 in health and disease. Nat Rev Endocrinol. 2018;14(7):390–403.

    Article  CAS  PubMed  Google Scholar 

  138. Christensen M, Sparre-Ulrich AH, Hartmann B, Grevstad U, Rosenkilde MM, Holst JJ, et al. Transfer of liraglutide from blood to cerebrospinal fluid is minimal in patients with type 2 diabetes. Int J Obes (Lond). 2015;39(11):1651–4.

    Article  CAS  Google Scholar 

  139. Hunter K, Holscher C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neurosci. 2012;23(13):33.

    Article  CAS  Google Scholar 

  140. Kastin AJ, Akerstrom V. Entry of exendin-4 into brain is rapid but may be limited at high doses. Int J Obes Relat Metab Disord. 2003;27(3):313–8.

    Article  CAS  PubMed  Google Scholar 

  141. Bhavsar S, Mudaliar S, Cherrington A. Evolution of exenatide as a diabetes therapeutic. Curr Diabetes Rev. 2013;9(2):161–93.

    PubMed  PubMed Central  Google Scholar 

  142. Perry T, Lahiri DK, Sambamurti K, Chen D, Mattson MP, Egan JM, 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. 2003;72(5):603–12.

    Article  CAS  PubMed  Google Scholar 

  143. Li Y, Duffy KB, Ottinger MA, Ray B, Bailey JA, Holloway HW, et al. GLP-1 receptor stimulation reduces amyloid-beta peptide accumulation and cytotoxicity in cellular and animal models of Alzheimer’s disease. J Alzheimers Dis. 2010;19(4):1205–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. McClean PL, Holscher C. Liraglutide can reverse memory impairment, synaptic loss and reduce plaque load in aged APP/PS1 mice, a model of Alzheimer’s disease. Neuropharmacology. 2014;76(Pt A):57–67.

    Article  CAS  PubMed  Google Scholar 

  145. McClean PL, Parthsarathy V, Faivre E, Holscher C. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J Neurosci. 2011;31(17):6587–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Liu XY, Wang LX, Chen Z, Liu LB. Liraglutide prevents beta-amyloid-induced neurotoxicity in SH-SY5Y cells via a PI3K-dependent signaling pathway. Neurol Res. 2016;38(4):313–9.

    Article  CAS  PubMed  Google Scholar 

  147. Sharma MK, Jalewa J, Holscher C. Neuroprotective and anti-apoptotic effects of liraglutide on SH-SY5Y cells exposed to methylglyoxal stress. J Neurochem. 2014;128(3):459–71.

    Article  CAS  PubMed  Google Scholar 

  148. Qi L, Chen Z, Wang Y, Liu X, Liu X, Ke L, et al. Subcutaneous liraglutide ameliorates methylglyoxal-induced Alzheimer-like tau pathology and cognitive impairment by modulating tau hyperphosphorylation and glycogen synthase kinase-3beta. Am J Transl Res. 2017;9(2):247–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Qi L, Ke L, Liu X, Liao L, Ke S, Liu X, et al. Subcutaneous administration of liraglutide ameliorates learning and memory impairment by modulating tau hyperphosphorylation via the glycogen synthase kinase-3beta pathway in an amyloid beta protein induced alzheimer disease mouse model. Eur J Pharmacol. 2016;15(783):23–32.

    Article  CAS  Google Scholar 

  150. Hansen HH, Fabricius K, Barkholt P, Niehoff ML, Morley JE, Jelsing J, et al. The GLP-1 receptor agonist liraglutide improves memory function and increases hippocampal CA1 neuronal numbers in a senescence-accelerated mouse model of Alzheimer’s disease. J Alzheimers Dis. 2015;46(4):877–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Yang Y, Zhang J, Ma D, Zhang M, Hu S, Shao S, et al. Subcutaneous administration of liraglutide ameliorates Alzheimer-associated tau hyperphosphorylation in rats with type 2 diabetes. J Alzheimers Dis. 2013;37(3):637–48.

    Article  CAS  PubMed  Google Scholar 

  152. Chen S, Sun J, Zhao G, Guo A, Chen Y, Fu R, et al. Liraglutide improves water maze learning and memory performance while reduces hyperphosphorylation of tau and neurofilaments in APP/PS1/tau triple transgenic mice. Neurochem Res. 2017;42(8):2326–35.

    Article  CAS  PubMed  Google Scholar 

  153. McClean PL, Jalewa J, Holscher C. Prophylactic liraglutide treatment prevents amyloid plaque deposition, chronic inflammation and memory impairment in APP/PS1 mice. Behav Brain Res. 2015;15(293):96–106.

    Article  CAS  Google Scholar 

  154. Hansen HH, Fabricius K, Barkholt P, Kongsbak-Wismann P, Schlumberger C, Jelsing J, et al. Long-term treatment with liraglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, has no effect on beta-amyloid plaque load in two transgenic APP/PS1 mouse models of Alzheimer’s disease. PLoS One. 2016;11(7):e0158205.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Batista AF, Forny-Germano L, Clarke JR, Lyra ESNM, Brito-Moreira J, Boehnke SE, et al. The diabetes drug liraglutide reverses cognitive impairment in mice and attenuates insulin receptor and synaptic pathology in a non-human primate model of Alzheimer’s disease. J Pathol. 2018;245(1):85–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. McClean PL, Holscher C. Lixisenatide, a drug developed to treat type 2 diabetes, shows neuroprotective effects in a mouse model of Alzheimer’s disease. Neuropharmacology. 2014;86:241–58.

    Article  CAS  PubMed  Google Scholar 

  157. Cai HY, Yang JT, Wang ZJ, Zhang J, Yang W, Wu MN, et al. Lixisenatide reduces amyloid plaques, neurofibrillary tangles and neuroinflammation in an APP/PS1/tau mouse model of Alzheimer’s disease. Biochem Biophys Res Commun. 2018;495(1):1034–40.

    Article  CAS  PubMed  Google Scholar 

  158. Li Y, Perry T, Kindy MS, Harvey BK, Tweedie D, Holloway HW, et al. GLP-1 receptor stimulation preserves primary cortical and dopaminergic neurons in cellular and rodent models of stroke and Parkinsonism. Proc Natl Acad Sci USA. 2009;106(4):1285–90.

    Article  PubMed  Google Scholar 

  159. Kim S, Moon M, Park S. Exendin-4 protects dopaminergic neurons by inhibition of microglial activation and matrix metalloproteinase-3 expression in an animal model of Parkinson’s disease. J Endocrinol. 2009;202(3):431–9.

    Article  CAS  PubMed  Google Scholar 

  160. Harkavyi A, Abuirmeileh A, Lever R, Kingsbury AE, Biggs CS, Whitton PS. Glucagon-like peptide 1 receptor stimulation reverses key deficits in distinct rodent models of Parkinson’s disease. J Neuroinflamm. 2008;21(5):19.

    Article  CAS  Google Scholar 

  161. Bertilsson G, Patrone C, Zachrisson O, Andersson A, Dannaeus K, Heidrich J, et al. Peptide hormone exendin-4 stimulates subventricular zone neurogenesis in the adult rodent brain and induces recovery in an animal model of Parkinson’s disease. J Neurosci Res. 2008;86(2):326–38.

    Article  CAS  PubMed  Google Scholar 

  162. Liu W, Jalewa J, Sharma M, Li G, Li L, Holscher C. Neuroprotective effects of lixisenatide and liraglutide in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson’s disease. Neuroscience. 2015;10(303):42–50.

    Article  CAS  Google Scholar 

  163. Zhang L, Zhang L, Li L, Holscher C. Neuroprotective effects of the novel GLP-1 long acting analogue semaglutide in the MPTP Parkinson’s disease mouse model. Neuropeptides. 2018;71:70–80.

    Article  CAS  PubMed  Google Scholar 

  164. Yun SP, Kam TI, Panicker N, Kim S, Oh Y, Park JS, et al. Block of A1 astrocyte conversion by microglia is neuroprotective in models of Parkinson’s disease. Nat Med. 2018;24(7):931–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. McManus RM, Heneka MT. Role of neuroinflammation in neurodegeneration: new insights. Alzheimers Res Ther. 2017;9(1):14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38(8):1285–91.

    Article  CAS  PubMed  Google Scholar 

  167. Duke DC, Moran LB, Pearce RK, Graeber MB. The medial and lateral substantia nigra in Parkinson’s disease: mRNA profiles associated with higher brain tissue vulnerability. Neurogenetics. 2007;8(2):83–94.

    Article  CAS  PubMed  Google Scholar 

  168. Sekiyama K, Sugama S, Fujita M, Sekigawa A, Takamatsu Y, Waragai M, et al. Neuroinflammation in Parkinson’s disease and related disorders: a lesson from genetically manipulated mouse models of alpha-synucleinopathies. Parkinsons Dis. 2012;2012:271732.

    PubMed  PubMed Central  Google Scholar 

  169. Brodacki B, Staszewski J, Toczylowska B, Kozlowska E, Drela N, Chalimoniuk M, et al. Serum interleukin (IL-2, IL-10, IL-6, IL-4), TNFalpha, and INFgamma concentrations are elevated in patients with atypical and idiopathic parkinsonism. Neurosci Lett. 2008;441(2):158–62.

    Article  CAS  PubMed  Google Scholar 

  170. Reale M, Greig NH, Kamal MA. Peripheral chemo-cytokine profiles in Alzheimer’s and Parkinson’s diseases. Mini Rev Med Chem. 2009;9(10):1229–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Song IU, Kim JS, Chung SW, Lee KS. Is there an association between the level of high-sensitivity C-reactive protein and idiopathic Parkinson’s disease? A comparison of Parkinson’s disease patients, disease controls and healthy individuals. Eur Neurol. 2009;62(2):99–104.

    Article  CAS  PubMed  Google Scholar 

  172. Brosseron F, Krauthausen M, Kummer M, Heneka MT. Body fluid cytokine levels in mild cognitive impairment and Alzheimer’s disease: a comparative overview. Mol Neurobiol. 2014;50(2):534–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Motta M, Imbesi R, Di Rosa M, Stivala F, Malaguarnera L. Altered plasma cytokine levels in Alzheimer’s disease: correlation with the disease progression. Immunol Lett. 2007;114(1):46–51.

    Article  CAS  PubMed  Google Scholar 

  174. Forloni G, Balducci C. Alzheimer’s disease, oligomers, and inflammation. J Alzheimers Dis. 2018;62(3):1261–76.

    Article  PubMed  PubMed Central  Google Scholar 

  175. Tai J, Liu W, Li Y, Li L, Holscher C. Neuroprotective effects of a triple GLP-1/GIP/glucagon receptor agonist in the APP/PS1 transgenic mouse model of Alzheimer’s disease. Brain Res. 2018;1(1678):64–74.

    Article  CAS  Google Scholar 

  176. Feng P, Zhang X, Li D, Ji C, Yuan Z, Wang R, et al. Two novel dual GLP-1/GIP receptor agonists are neuroprotective in the MPTP mouse model of Parkinson’s disease. Neuropharmacology. 2018;1(133):385–94.

    Article  CAS  Google Scholar 

  177. Yuan Z, Li D, Feng P, Xue G, Ji C, Li G, et al. A novel GLP-1/GIP dual agonist is more effective than liraglutide in reducing inflammation and enhancing GDNF release in the MPTP mouse model of Parkinson’s disease. Eur J Pharmacol. 2017;5(812):82–90.

    Article  CAS  Google Scholar 

  178. Liddelow SA, Barres BA. Reactive astrocytes: production, function, and therapeutic potential. Immunity. 2017;46(6):957–67.

    Article  CAS  PubMed  Google Scholar 

  179. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541(7638):481–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Li Y, Liu W, Li L, Holscher C. D-Ala2-GIP-glu-PAL is neuroprotective in a chronic Parkinson’s disease mouse model and increases BNDF expression while reducing neuroinflammation and lipid peroxidation. Eur J Pharmacol. 2017;15(797):162–72.

    Article  CAS  Google Scholar 

  181. Li Y, Liu W, Li L, Holscher C. Neuroprotective effects of a GIP analogue in the MPTP Parkinson’s disease mouse model. Neuropharmacology. 2016;101:255–63.

    Article  CAS  PubMed  Google Scholar 

  182. Duffy AM, Holscher C. The incretin analogue D-Ala2GIP reduces plaque load, astrogliosis and oxidative stress in an APP/PS1 mouse model of Alzheimer’s disease. Neuroscience. 2013;3(228):294–300.

    Article  CAS  Google Scholar 

  183. Faivre E, Holscher C. D-Ala2GIP facilitated synaptic plasticity and reduces plaque load in aged wild type mice and in an Alzheimer’s disease mouse model. J Alzheimers Dis. 2013;35(2):267–83.

    Article  CAS  PubMed  Google Scholar 

  184. Faivre E, Hamilton A, Holscher C. Effects of acute and chronic administration of GIP analogues on cognition, synaptic plasticity and neurogenesis in mice. Eur J Pharmacol. 2012;674(2–3):294–306.

    Article  CAS  PubMed  Google Scholar 

  185. Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab. 2013;17(6):819–37.

    Article  CAS  PubMed  Google Scholar 

  186. Pilkis SJ, Granner DK. Molecular physiology of the regulation of hepatic gluconeogenesis and glycolysis. Annu Rev Physiol. 1992;54:885–909.

    Article  CAS  PubMed  Google Scholar 

  187. Sandoval DA, D’Alessio DA. Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease. Physiol Rev. 2015;95(2):513–48.

    Article  CAS  PubMed  Google Scholar 

  188. Finan B, Ma T, Ottaway N, Muller TD, Habegger KM, Heppner KM, et al. Unimolecular dual incretins maximize metabolic benefits in rodents, monkeys, and humans. Sci Transl Med. 2013;5(209):209ra151.

    Article  CAS  PubMed  Google Scholar 

  189. Frias JP, Bastyr EJ 3rd, Vignati L, Tschop MH, Schmitt C, Owen K, et al. The sustained effects of a dual GIP/GLP-1 receptor agonist, NNC0090-2746, in patients with type 2 diabetes. Cell Metab. 2017;26(2):343–52.

    Article  CAS  PubMed  Google Scholar 

  190. Zhou J, Cai X, Huang X, Dai Y, Sun L, Zhang B, et al. A novel glucagon-like peptide-1/glucagon receptor dual agonist exhibits weight-lowering and diabetes-protective effects. Eur J Med Chem. 2017;29(138):1158–69.

    Article  CAS  Google Scholar 

  191. Jall S, Sachs S, Clemmensen C, Finan B, Neff F, DiMarchi RD, et al. Monomeric GLP-1/GIP/glucagon triagonism corrects obesity, hepatosteatosis, and dyslipidemia in female mice. Mol Metab. 2017;6(5):440–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Jalewa J, Sharma MK, Gengler S, Holscher C. A novel GLP-1/GIP dual receptor agonist protects from 6-OHDA lesion in a rat model of Parkinson’s disease. Neuropharmacology. 2017;1(117):238–48.

    Article  CAS  Google Scholar 

  193. Cao Y, Holscher C, Hu MM, Wang T, Zhao F, Bai Y, et al. DA5-CH, a novel GLP-1/GIP dual agonist, effectively ameliorates the cognitive impairments and pathology in the APP/PS1 mouse model of Alzheimer’s disease. Eur J Pharmacol. 2018;15(827):215–26.

    Article  CAS  Google Scholar 

  194. Aviles-Olmos I, Dickson J, Kefalopoulou Z, Djamshidian A, Ell P, Soderlund T, et al. Exenatide and the treatment of patients with Parkinson’s disease. J Clin Investig. 2013;123(6):2730–6.

    Article  CAS  PubMed  Google Scholar 

  195. Aviles-Olmos I, Dickson J, Kefalopoulou Z, Djamshidian A, Kahan J, Ell P, et al. Motor and cognitive advantages persist 12 months after exenatide exposure in Parkinson’s disease. J Parkinsons Dis. 2014;4(3):337–44.

    Article  CAS  PubMed  Google Scholar 

  196. Athauda D, Maclagan K, Skene SS, Bajwa-Joseph M, Letchford D, Chowdhury K, et al. Exenatide once weekly versus placebo in Parkinson’s disease: a randomised, double-blind, placebo-controlled trial. Lancet. 2017;390(10103):1664–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Mansur RB, Ahmed J, Cha DS, Woldeyohannes HO, Subramaniapillai M, Lovshin J, et al. Liraglutide promotes improvements in objective measures of cognitive dysfunction in individuals with mood disorders: a pilot, open-label study. J Affect Disord. 2017;1(207):114–20.

    Article  CAS  Google Scholar 

  198. Egefjord L, Gejl M, Moller A, Braendgaard H, Gottrup H, Antropova O, et al. Effects of liraglutide on neurodegeneration, blood flow and cognition in Alzheimer s disease—protocol for a controlled, randomized double-blinded trial. Dan Med J. 2012;59(10):A4519.

    PubMed  Google Scholar 

  199. Gejl M, Gjedde A, Egefjord L, Moller A, Hansen SB, Vang K, et al. In Alzheimer’s disease, 6-month treatment with GLP-1 analog prevents decline of brain glucose metabolism: randomized, placebo-controlled, double-blind clinical trial. Front Aging Neurosci. 2016;8:108.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the use of Mind the Graph (http://www.mindthegraph.com) for the design of the figure in this work.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Fernanda G. De Felice or Sergio T. Ferreira.

Ethics declarations

Conflict of interest

AFB, VBS, FGDF and STF have no conflicts of interest that are directly relevant to the content of this article.

Funding

Work in the authors’ groups has been supported by the National Institute for Translational Neuroscience (Brazil), the Brazilian agencies Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq) and Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ) (to STF and FDF), and Alzheimer Society Canada and Weston Brain Institute (to FDF). AFB and VBS are supported by post-doctoral and pre-doctoral fellowships, respectively, from CNPq.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Batista, A.F., Bodart-Santos, V., De Felice, F.G. et al. Neuroprotective Actions of Glucagon-Like Peptide-1 (GLP-1) Analogues in Alzheimer’s and Parkinson’s Diseases. CNS Drugs 33, 209–223 (2019). https://doi.org/10.1007/s40263-018-0593-6

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-018-0593-6

Navigation