939
Views
26
CrossRef citations to date
0
Altmetric
Review

Modulation of GLP-1 signaling as a novel therapeutic approach in the treatment of Alzheimer’s disease pathology

, , , &
Pages 59-75 | Received 25 Jul 2016, Accepted 05 Oct 2016, Published online: 20 Oct 2016

References

  • Alzheimer’s Disease International. World Alzheimer Report 2015. The global impact of dementia. Ed. London: Alzheimer’s Disease International (ADI); 2015.
  • Albanese E, Acosta D, Guerra M, et al. Dementia in the developing world: risk, impact, and prevention. Neurobiol Aging. 2014;35:S1–S2.
  • Profenno LA, Porsteinsson AP, Faraone SV. Meta-analysis of Alzheimer’s disease risk with obesity, diabetes, and related disorders. Biol Psychiatry. 2010;67(6):505–512.
  • Tolppanen AM, Lavikainen P, Solomon A, et al. History of medically treated diabetes and risk of Alzheimer disease in a nationwide case-control study. Diabetes Care. 2013;36(7):2015–2019.
  • Biessels GJ, Strachan MW, Visseren FL, et al. Dementia and cognitive decline in type 2 diabetes and prediabetic stages: towards targeted interventions. Lancet Diabetes Endocrinol. 2014;2(3):246–255.
  • Cheng G, Huang C, Deng H, et al. Diabetes as a risk factor for dementia and mild cognitive impairment: a meta-analysis of longitudinal studies. Intern Med J. 2012;42(5):484–491.
  • Butterfield DA, Di Domenico F, Barone E. Elevated risk of type 2 diabetes for development of Alzheimer disease: a key role for oxidative stress in brain. Biochim Biophys Acta. 2014;1842(9):1693–1706.
  • Bedse G, Di Domenico F, Serviddio G, et al. Aberrant insulin signaling in Alzheimer’s disease: current knowledge. Front Neurosci. 2015;9:204.
  • Craft S, Cholerton B, Baker LD. Insulin and Alzheimer’s disease: untangling the web. J Alzheimers Dis. 2013;33(Suppl 1):S263–275.
  • Lebovitz HE. Insulin resistance: definition and consequences. Exp Clin Endocrinol Diabetes. 2001;109(Suppl 2):S135–148.
  • Grundy SM, Brewer HB, Cleeman JI, et al. Definition of metabolic syndrome - report of the national heart, lung, and blood institute/American heart association conference on scientific issues related to definition. Circulation. 2004;109(3):433–438.
  • Watson GS, Craft S. Modulation of memory by insulin and glucose: neuropsychological observations in Alzheimer’s disease. Eur J Pharmacol. 2004;490(1–3):97–113.
  • Schwartz MW, Figlewicz DF, Kahn SE, et al. Insulin binding to brain capillaries is reduced in genetically obese, hyperinsulinemic zucker rats. Peptides. 1990;11(3):467–472.
  • Wallum BJ, Taborsky GJ Jr., Porte D Jr., et al. Cerebrospinal fluid insulin levels increase during intravenous insulin infusions in man. J Clin Endocrinol Metab. 1987;64(1):190–194.
  • Barone E, Di Domenico F, Cassano T, et al. Impairment of biliverdin reductase-A promotes brain insulin resistance in Alzheimer disease: a new paradigm. Free Radic Biol Med. 2016;91:127–142.
  • Ferreira ST, Clarke JR, Bomfim TR, et al. Inflammation, defective insulin signaling, and neuronal dysfunction in Alzheimer’s disease. Alzheimers Dement. 2014;10(1 Suppl):S76–S83.
  • Biessels GJ, Reagan LP. Hippocampal insulin resistance and cognitive dysfunction. Nat Rev Neurosci. 2015;16(11):660–671.
  • 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):2262–2272.
  • De Felice FG, Vieira MN, Bomfim TR, et al. Protection of synapses against Alzheimer’s-linked toxins: insulin signaling prevents the pathogenic binding of abeta oligomers. Proc Natl Acad Sci U S A. 2009;106(6):1971–1976.
  • Bomfim TR, Forny-Germano L, Sathler LB, et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease-associated abeta oligomers. J Clin Invest. 2012;122(4):1339–1353.
  • Lourenco MV, Clarke JR, Frozza RL, 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):831–843.
  • Yamagishi S, Ueda S, Okuda S. Food-derived advanced glycation end products (AGEs): a novel therapeutic target for various disorders. Curr Pharm Des. 2007;13(27):2832–2836.
  • Jia X, Olson DJ, Ross AR, et al. Structural and functional changes in human insulin induced by methylglyoxal. Faseb J. 2006;20(9):1555–1557.
  • Luchsinger JA, Tang MX, Shea S, et al. Hyperinsulinemia and risk of Alzheimer disease. Neurology. 2004;63(7):1187–1192.
  • Qiu WQ, Folstein MF. Insulin, insulin-degrading enzyme and amyloid-beta peptide in Alzheimer’s disease: review and hypothesis. Neurobiol Aging. 2006;27(2):190–198.
  • Schrijvers EM, Witteman JC, Sijbrands EJ, et al. Insulin metabolism and the risk of Alzheimer disease: the rotterdam study. Neurology. 2010;75(22):1982–1987.
  • Plum L, Schubert M, Bruning JC. The role of insulin receptor signaling in the brain. Trends Endocrinol Metab. 2005;16(2):59–65.
  • Ghasemi R, Haeri A, Dargahi L, et al. Insulin in the brain: sources, localization and functions. Mol Neurobiol. 2013;47(1):145–171.
  • Belanger M, Allaman I, Magistretti PJ. Brain energy metabolism: focus on astrocyte-neuron metabolic cooperation. Cell Metab. 2011;14(6):724–738.
  • Jurcovicova J. Glucose transport in brain - effect of inflammation. Endocr Regul. 2014;48(1):35–48.
  • Holst JJ, Burcelin R, Nathanson E. Neuroprotective properties of GLP-1: theoretical and practical applications. Curr Med Res Opin. 2011;27(3):547–558.
  • Mojsov S, Weir GC, Habener JF. Insulinotropin: glucagon-like peptide I (7-37) co-encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas. J Clin Invest. 1987;79(2):616–619.
  • Kreymann B, Yiangou Y, Kanse S, et al. Isolation and characterisation of GLP-1 7-36 amide from rat intestine. Elevated levels in diabetic rats. FEBS Lett. 1988;242(1):167–170.
  • Kreymann B, Williams G, Ghatei MA, et al. Glucagon-like peptide-1 7-36: a physiological incretin in man. Lancet. 1987;2(8571):1300–1304.
  • Tolhurst G, Reimann F, Gribble FM. Nutritional regulation of glucagon-like peptide-1 secretion. J Physiol. 2009;587(1):27–32.
  • Korner M, Stockli M, Waser B, et al. GLP-1 receptor expression in human tumors and human normal tissues: potential for in vivo targeting. J Nucl Med. 2007;48(5):736–743.
  • Nadkarni P, Chepurny OG, Holz GG. Regulation of glucose homeostasis by GLP-1. Prog Mol Biol Transl Sci. 2014;121:23–65.
  • Koole C, Pabreja K, Savage EE, et al. Recent advances in understanding GLP-1R (glucagon-like peptide-1 receptor) function. Biochem Soc Trans. 2013;41(1):172–179.
  • Hallbrink M, Holmqvist T, Olsson M, et al. Different domains in the third intracellular loop of the GLP-1 receptor are responsible for Galpha(s) and Galpha(i)/Galpha(o) activation. Biochim Biophys Acta. 2001;1546(1):79–86.
  • Montrose-Rafizadeh C, Avdonin P, Garant MJ, 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. 1999;140(3):1132–1140.
  • Hay CW, Sinclair EM, Bermano G, et al. Glucagon-like peptide-1 stimulates human insulin promoter activity in part through cAMP-responsive elements that lie upstream and downstream of the transcription start site. J Endocrinol. 2005;186(2):353–365.
  • Drucker DJ. Glucagon-like peptides: regulators of cell proliferation, differentiation, and apoptosis. Mol Endocrinol. 2003;17(2):161–171.
  • Buteau J. GLP-1 receptor signaling: effects on pancreatic beta-cell proliferation and survival. Diabetes Metab. 2008;34(Suppl 2):S73–77.
  • McIntosh CH, Widenmaier S, Kim SJ. Pleiotropic actions of the incretin hormones. Vitam Horm. 2010;84:21–79.
  • Portha B, Tourrel-Cuzin C, Movassat J. Activation of the GLP-1 receptor signalling pathway: a relevant strategy to repair a deficient beta-cell mass. Exp Diabetes Res. 2011;376509:2011.
  • Yabe D, Seino Y. Two incretin hormones GLP-1 and GIP: comparison of their actions in insulin secretion and beta cell preservation. Prog Biophys Mol Biol. 2011;107(2):248–256.
  • Trapp S, Richards JE. The gut hormone glucagon-like peptide-1 produced in brain: is this physiologically relevant? Curr Opin Pharmacol. 2013;13(6):964–969.
  • Baggio LL, Drucker DJ. Glucagon-like peptide-1 receptors in the brain: controlling food intake and body weight. J Clin Invest. 2014;124(10):4223–4226.
  • Parthsarathy V, Holscher C. The type 2 diabetes drug liraglutide reduces chronic inflammation induced by irradiation in the mouse brain. Eur J Pharmacol. 2013;700(1–3):42–50.
  • Fischer AJ, Omar G, Walton NA, et al. Glucagon-expressing neurons within the retina regulate the proliferation of neural progenitors in the circumferential marginal zone of the avian eye. J Neurosci. 2005;25(44):10157–10166.
  • Furman B, Ong WK, Pyne NJ. Cyclic AMP signaling in pancreatic islets. Adv Exp Med Biol. 2010;654:281–304.
  • Song WJ, Seshadri M, Ashraf U, et al. Snapin mediates incretin action and augments glucose-dependent insulin secretion. Cell Metab. 2011;13(3):308–319.
  • Jhala US, Canettieri G, Screaton RA, et al. cAMP promotes pancreatic beta-cell survival via CREB-mediated induction of IRS2. Genes Dev. 2003;17(13):1575–1580.
  • Ortega-Martinez S. A new perspective on the role of the CREB family of transcription factors in memory consolidation via adult hippocampal neurogenesis. Front Mol Neurosci. 2015;8:46.
  • Leech CA, Chepurny OG, Holz GG. Epac2-dependent rap1 activation and the control of islet insulin secretion by glucagon-like peptide-1. Vitam Horm. 2010;84:279–302.
  • Koole C, Wootten D, Simms J, et al. Second extracellular loop of human glucagon-like peptide-1 receptor (GLP-1R) has a critical role in GLP-1 peptide binding and receptor activation. J Biol Chem. 2012;287(6):3642–3658.
  • Holz GG. Epac: A new cAMP-binding protein in support of glucagon-like peptide-1 receptor-mediated signal transduction in the pancreatic beta-cell. Diabetes. 2004;53(1):5–13.
  • McClean PL, Gault VA, Harriott P, et al. Glucagon-like peptide-1 analogues enhance synaptic plasticity in the brain: a link between diabetes and Alzheimer’s disease. Eur J Pharmacol. 2010;630(1–3):158–162.
  • Holscher C, Fung K, McCurtin R, et al. Novel GLP-1 analogues cross the blood brain barrier and enhance synaptic plasticity in the brain: a link between diabetes and Alzheimer’s disease. Diabetologia. 2009;52:S311–S311.
  • McClean PL, Parthasarathy V, Gault VA, et al. Liraglutide, a novel glp-1 analogue, prevents the impairment of learning and ltp and plaque formation in an app/ps-1 mouse model of Alzheimer’s disease. Irish J Med Sci. 2011;180:24–24.
  • Scrocchi LA, Brown TJ, MaClusky N, et al. Glucose intolerance but normal satiety in mice with a null mutation in the glucagon-like peptide 1 receptor gene. Nat Med. 1996;2(11):1254–1258.
  • Perry T, Lahiri DK, Chen D, et al. A novel neurotrophic property of glucagon-like peptide 1: a promoter of nerve growth factor-mediated differentiation in PC12 cells. J Pharmacol Exp Ther. 2002;300(3):958–966.
  • Bertilsson G, Patrone C, Zachrisson O, 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):326–338.
  • Belsham DD, Fick LJ, Dalvi PS, et al. Ciliary neurotrophic factor recruitment of glucagon-like peptide-1 mediates neurogenesis, allowing immortalization of adult murine hypothalamic neurons. Faseb J. 2009;23(12):4256–4265.
  • Perry T, Haughey NJ, Mattson MP, et al. Protection and reversal of excitotoxic neuronal damage by glucagon-like peptide-1 and exendin-4. J Pharmacol Exp Ther. 2002;302(3):881–888.
  • Perry T, Lahiri DK, Sambamurti K, 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):603–612.
  • Li Y, Tweedie D, Mattson MP, et al. Enhancing the GLP-1 receptor signaling pathway leads to proliferation and neuroprotection in human neuroblastoma cells. J Neurochem. 2010;113(6):1621–1631.
  • Fukuda S, Nakagawa S, Tatsumi R, et al. Glucagon-like peptide-1 strengthens the barrier integrity in primary cultures of rat brain endothelial cells under basal and hyperglycemia conditions. J Mol Neurosci. 2016;59(2):211–219.
  • Turton MD, O’Shea D, Gunn I, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature. 1996;379(6560):69–72.
  • Knauf C, Cani PD, Perrin C, et al. Brain glucagon-like peptide-1 increases insulin secretion and muscle insulin resistance to favor hepatic glycogen storage. J Clin Invest. 2005;115(12):3554–3563.
  • Flint A, Raben A, Astrup A, et al. Glucagon-like peptide 1 promotes satiety and suppresses energy intake in humans. J Clin Invest. 1998;101(3):515–520.
  • Buteau J, Spatz ML, Accili D. Transcription factor FoxO1 mediates glucagon-like peptide-1 effects on pancreatic beta-cell mass. Diabetes. 2006;55(5):1190–1196.
  • Li Y, Cao X, Li LX, et al. beta-cell Pdx1 expression is essential for the glucoregulatory, proliferative, and cytoprotective actions of glucagon-like peptide-1. Diabetes. 2005;54(2):482–491.
  • Cozar-Castellano I, Fiaschi-Taesch N, Bigatel TA, et al. Molecular control of cell cycle progression in the pancreatic beta-cell. Endocr Rev. 2006;27(4):356–370.
  • Athauda D, Foltynie T. The glucagon-like peptide 1 (GLP) receptor as a therapeutic target in Parkinson’s disease: mechanisms of action. Drug Discov Today. 2016;21(5):802–818.
  • Kimura R, Okouchi M, Fujioka H, et al. Glucagon-like peptide-1 (GLP-1) protects against methylglyoxal-induced PC12 cell apoptosis through the PI3K/akt/mTOR/GCLc/redox signaling pathway. Neuroscience. 2009;162(4):1212–1219.
  • Farilla L, Bulotta A, Hirshberg B, et al. Glucagon-like peptide 1 inhibits cell apoptosis and improves glucose responsiveness of freshly isolated human islets. Endocrinology. 2003;144(12):5149–5158.
  • Holscher C. The role of GLP-1 in neuronal activity and neurodegeneration. Vitam Horm. 2010;84:331–354.
  • Velmurugan K, Bouchard R, Mahaffey G, et al. Neuroprotective actions of Glucagon-like peptide-1 in differentiated human neuroprogenitor cells. J Neurochem. 2012;123(6):919–931.
  • Bao Y, Jiang L, Chen HY, et al. The neuroprotective effect of liraglutide is mediated by glucagon-like peptide 1 receptor-mediated activation of cAMP/PKA/CREB pathway. Cell Physiol Biochem. 2015;36(6):2366–2378.
  • Kieffer TJ, McIntosh CH, Pederson RA. Degradation of glucose-dependent insulinotropic polypeptide and truncated glucagon-like peptide 1 in vitro and in vivo by dipeptidyl peptidase IV. Endocrinology. 1995;136(8):3585–3596.
  • Shigeto M, Katsura M, Matsuda M, et al. but physiological, concentration of GLP-1 stimulates insulin secretion independent of the cAMP-dependent protein kinase pathway. J Pharmacol Sci. 2008;108(3):274–279.
  • Gautier JF, Choukem SP, Girard J. Physiology of incretins (GIP and GLP-1) and abnormalities in type 2 diabetes. Diabetes Metab. 2008;34(Suppl 2):S65–72.
  • Hansen L, Deacon CF, Orskov C, et al. Glucagon-like peptide-1-(7-36)amide is transformed to glucagon-like peptide-1-(9-36)amide by dipeptidyl peptidase IV in the capillaries supplying the L cells of the porcine intestine. Endocrinology. 1999;140(11):5356–5363.
  • Mentlein R. Dipeptidyl-peptidase IV (CD26)–role in the inactivation of regulatory peptides. Regul Pept. 1999;85(1):9–24.
  • Cork SC, Richards JE, Holt MK, et al. Distribution and c haracterisation of glucagon-like peptide-1 receptor expressing cells in the mouse brain. Mol Metab. 2015;4(10):718–731.
  • 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):261–280.
  • Iwai T, Ito S, Tanimitsu K, et al. Glucagon-like peptide-1 inhibits LPS-induced IL-1beta production in cultured rat astrocytes. Neurosci Res. 2006;55(4):352–360.
  • Hamilton A, Holscher C. Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuroreport. 2009;20(13):1161–1166.
  • Ghosal S, Myers B, Herman JP. Role of central glucagon-like peptide-1 in stress regulation. Physiol Behav. 2013;122:201–207.
  • Vrang N, Larsen PJ. Preproglucagon derived peptides GLP-1, GLP-2 and oxyntomodulin in the CNS: role of peripherally secreted and centrally produced peptides. Prog Neurobiol. 2010;92(3):442–462.
  • Barrera JG, Sandoval DA, D’Alessio DA, et al. GLP-1and energy balance: an integrated model of short-term and long-term control. Nat Rev Endocrinol. 2011;7(9):507–516.
  • Trapp S, Hisadome K. Glucagon-like peptide 1 and the brain: central actions-central sources? Auton Neurosci. 2011;161(1–2):14–19.
  • Talbot K, Wang HY. The nature, significance, and glucagon-like peptide-1 analog treatment of brain insulin resistance in Alzheimer’s disease. Alzheimers Dement. 2014;10(1 Suppl):S12–S25.
  • Holscher C, Li L. New roles for insulin-like hormones in neuronal signalling and protection: new hopes for novel treatments of Alzheimer’s disease? Neurobiol Aging. 2010;31(9):1495–1502.
  • Li H, Lee CH, Yoo KY, et al. Chronic treatment of exendin-4 affects cell proliferation and neuroblast differentiation in the adult mouse hippocampal dentate gyrus. Neurosci Lett. 2010;486(1):38–42.
  • Hamilton A, Patterson S, Porter D, et al. Novel GLP-1 mimetics developed to treat type 2 diabetes promote progenitor cell proliferation in the brain. J Neurosci Res. 2011;89(4):481–489.
  • Isacson R, Nielsen E, Dannaeus K, et al. The glucagon-like peptide 1 receptor agonist exendin-4 improves reference memory performance and decreases immobility in the forced swim test. Eur J Pharmacol. 2011;650(1):249–255.
  • McClean PL, Parthsarathy V, Faivre E, et al. The diabetes drug liraglutide prevents degenerative processes in a mouse model of Alzheimer’s disease. J Neurosci. 2011;31(17):6587–6594.
  • Perry T, Holloway HW, Weerasuriya A, et al. Evidence of GLP-1-mediated neuroprotection in an animal model of pyridoxine-induced peripheral sensory neuropathy. Exp Neurol. 2007;203(2):293–301.
  • Luciani P, Deledda C, Benvenuti S, et al. Differentiating effects of the glucagon-like peptide-1 analogue exendin-4 in a human neuronal cell model. Cell Mol Life Sci. 2010;67(21):3711–3723.
  • 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):265–271.
  • During MJ, Cao L, Zuzga DS, et al. Glucagon-like peptide-1 receptor is involved in learning and neuroprotection. Nat Med. 2003;9(9):1173–1179.
  • Holscher C. Central effects of GLP-1: new opportunities for treatments of neurodegenerative diseases. J Endocrinol. 2014;221(1):T31–41.
  • Gonzalez C, Beruto V, Keller G, et al. Investigational treatments for type 2 diabetes mellitus: exenatide and liraglutide. Expert Opin Investig Drugs. 2006;15(8):887–895.
  • Ahren B, Landin-Olsson M, Jansson PA, et al. Inhibition of dipeptidyl peptidase-4 reduces glycemia, sustains insulin levels, and reduces glucagon levels in type 2 diabetes. J Clin Endocrinol Metab. 2004;89(5):2078–2084.
  • Yoshino Y, Ishisaka M, Tsujii S, et al. Glucagon-like peptide-1 protects the murine hippocampus against stressors via akt and ERK1/2 signaling. Biochem Biophys Res Commun. 2015;458(2):274–279.
  • Li Y, Duffy KB, Ottinger MA, 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):1205–1219.
  • Chen S, Liu AR, An FM, et al. Amelioration of neurodegenerative changes in cellular and rat models of diabetes-related Alzheimer’s disease by exendin-4. Age (Dordr). 2012;34(5):1211–1224.
  • Xu W, Yang Y, Yuan G, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist, reduces Alzheimer disease-associated tau hyperphosphorylation in the hippocampus of rats with type 2 diabetes. J Investig Med. 2015;63(2):267–272.
  • Solmaz V, Cinar BP, Yigitturk G, et al. Exenatide reduces TNF-alpha expression and improves hippocampal neuron numbers and memory in streptozotocin treated rats. Eur J Pharmacol. 2015;765:482–487.
  • Takach O, Gill TB, Silverman MA. Modulation of insulin signaling rescues BDNF transport defects independent of tau in amyloid-beta oligomer-treated hippocampal neurons. Neurobiol Aging. 2015;36(3):1378–1382.
  • Tweedie D, Rachmany L, Rubovitch V, et al. Exendin-4, a glucagon-like peptide-1 receptor agonist prevents mTBI-induced changes in hippocampus gene expression and memory deficits in mice. Exp Neurol. 2013;239:170–182.
  • Bomba M, Ciavardelli D, Silvestri E, et al. Exenatide promotes cognitive enhancement and positive brain metabolic changes in PS1-KI mice but has no effects in 3xTg-AD animals. Cell Death Dis. 2013;4:e612.
  • 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;86:241–258.
  • Liu J, Zheng X, Yin F, et al. Neurotrophic property of geniposide for inducing the neuronal differentiation of PC12 cells. Int J Dev Neurosci. 2006;24(7):419–424.
  • Liu J, Yin F, Zheng X, et al. Geniposide, a novel agonist for GLP-1 receptor, prevents PC12 cells from oxidative damage via MAP kinase pathway. Neurochem Int. 2007;51(6–7):361–369.
  • Gao C, Liu Y, Jiang Y, et al. Geniposide ameliorates learning memory deficits, reduces tau phosphorylation and decreases apoptosis via GSK3beta pathway in streptozotocin-induced alzheimer rat model. Brain Pathol. 2014;24(3):261–269.
  • Wang XH, Li L, Holscher C, 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. 2010;170(4):1239–1248.
  • Gengler S, McClean PL, McCurtin R, et al. Val(8)GLP-1 rescues synaptic plasticity and reduces dense core plaques in APP/PS1 mice. Neurobiol Aging. 2012;33(2):265–276.
  • Lennox R, Porter DW, Flatt PR, et al. Comparison of the independent and combined effects of sub-chronic therapy with metformin and a stable GLP-1 receptor agonist on cognitive function, hippocampal synaptic plasticity and metabolic control in high-fat fed mice. Neuropharmacology. 2014;86:22–30.
  • Li L, Zhang ZF, Holscher C, et al. (Val(8)) glucagon-like peptide-1 prevents tau hyperphosphorylation, impairment of spatial learning and ultra-structural cellular damage induced by streptozotocin in rat brains. Eur J Pharmacol. 2012;674(2–3):280–286.
  • Ma T, Du X, Pick JE, et al. Glucagon-like peptide-1 cleavage product GLP-1 (9-36)amide rescues synaptic plasticity and memory deficits in Alzheimer’s disease model mice. J Neurosci. 2012;32(40):13701–13708.
  • Iwai T, Sawabe T, Tanimitsu K, et al. Glucagon-like peptide-1 protects synaptic and learning functions from neuroinflammation in rodents. J Neurosci Res. 2014;92(4):446–454.
  • 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.
  • Parthsarathy V, Holscher C. Chronic treatment with the GLP1 analogue liraglutide increases cell proliferation and differentiation into neurons in an AD mouse model. PLoS One. 2013;8(3):e58784.
  • 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;293:96–106.
  • Hou J, Manaenko A, Hakon J, et al. Liraglutide, a long-acting GLP-1 mimetic, and its metabolite attenuate inflammation after intracerebral hemorrhage. J Cereb Blood Flow Metab. 2012;32(12):2201–2210.
  • Hakon J, Ruscher K, Romner B, et al. Preservation of the blood brain barrier and cortical neuronal tissue by liraglutide, a long acting glucagon-like-1 analogue, after experimental traumatic brain injury. PLoS One. 2015;10(3):e0120074.
  • Hansen HH, Barkholt P, Fabricius K, et al. The GLP-1 receptor agonist liraglutide reduces pathology-specific tau phosphorylation and improves motor function in a transgenic hTauP301L mouse model of tauopathy. Brain Res. 1634;158-170:2016.
  • Xiong H, Zheng C, Wang J, et al. The neuroprotection of liraglutide on Alzheimer-like learning and memory impairment by modulating the hyperphosphorylation of tau and neurofilament proteins and insulin signaling pathways in mice. J Alzheimers Dis. 2013;37(3):623–635.
  • Qi L, Ke L, 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;783:23–32.
  • Yang Y, Zhang J, Ma D, et al. Subcutaneous administration of liraglutide ameliorates Alzheimer-associated tau hyperphosphorylation in rats with type 2 diabetes. J Alzheimers Dis. 2013;37(3):637–648.
  • Spolcova A, Mikulaskova B, Holubova M, et al. Anorexigenic lipopeptides ameliorate central insulin signaling and attenuate tau phosphorylation in hippocampi of mice with monosodium glutamate-induced obesity. J Alzheimers Dis. 2015;45(3):823–835.
  • Long-Smith CM, Manning S, McClean PL, et al. The diabetes drug liraglutide ameliorates aberrant insulin receptor localisation and signalling in parallel with decreasing both amyloid-beta plaque and glial pathology in a mouse model of Alzheimer’s disease. Neuromolecular Med. 2013;15(1):102–114.
  • Gejl M, Gjedde A, Egefjord L, 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). doi:10.3389/fnagi.2016.00108.
  • D’Amico M, Di Filippo C, Marfella R, et al. Long-term inhibition of dipeptidyl peptidase-4 in Alzheimer’s prone mice. Exp Gerontol. 2010;45(3):202–207.
  • Tsai TH, Sun CK, Su CH, et al. Sitagliptin attenuated brain damage and cognitive impairment in mice with chronic cerebral hypo-perfusion through suppressing oxidative stress and inflammatory reaction. J Hypertens. 2015;33(5):1001–1013.
  • Pintana H, Apaijai N, Chattipakorn N, et al. DPP-4 inhibitors improve cognition and brain mitochondrial function of insulin-resistant rats. J Endocrinol. 2013;218(1):1–11.
  • Gault VA, Lennox R, Flatt PR. Sitagliptin, a dipeptidyl peptidase-4 inhibitor, improves recognition memory, oxidative stress and hippocampal neurogenesis and upregulates key genes involved in cognitive decline. Diabetes Obes Metab. 2015;17(4):403–413.
  • Kosaraju J, Gali CC, Khatwal RB, et al. Saxagliptin: a dipeptidyl peptidase-4 inhibitor ameliorates streptozotocin induced Alzheimer’s disease. Neuropharmacology. 2013;72:291–300.
  • Pipatpiboon N, Pintana H, Pratchayasakul W, et al. DPP4-inhibitor improves neuronal insulin receptor function, brain mitochondrial function and cognitive function in rats with insulin resistance induced by high-fat diet consumption. Eur J Neurosci. 2013;37(5):839–849.
  • Drucker DJ, Nauck MA. The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368(9548):1696–1705.
  • Nauck MA. Incretin-based therapies for type 2 diabetes mellitus: properties, functions, and clinical implications. Am J Med. 2011;124(1 Suppl):S3–S18.
  • Karagiannis T, Paschos P, Paletas K, et al. Dipeptidyl peptidase-4 inhibitors for treatment of type 2 diabetes mellitus in the clinical setting: systematic review and meta-analysis. Bmj. 2012;344:e1369.
  • Fura A, Khanna A, Vyas V, et al. Pharmacokinetics of the dipeptidyl peptidase 4 inhibitor saxagliptin in rats, dogs, and monkeys and clinical projections. Drug Metab Dispos. 2009;37(6):1164–1171.
  • Fuchs H, Binder R, Greischel A. Tissue distribution of the novel DPP-4 inhibitor BI 1356 is dominated by saturable binding to its target in rats. Biopharm Drug Dispos. 2009;30(5):229–240.
  • Deacon CF. Dipeptidyl peptidase-4 inhibitors in the treatment of type 2 diabetes: a comparative review. Diabetes Obes Metab. 2011;13(1):7–18.
  • Nielsen LL, Young AA, Parkes DG. Pharmacology of exenatide (synthetic exendin-4): a potential therapeutic for improved glycemic control of type 2 diabetes. Regul Pept. 2004;117(2):77–88.
  • Hinckelmann MV, Zala D, Saudou F. Releasing the brake: restoring fast axonal transport in neurodegenerative disorders. Trends Cell Biol. 2013;23(12):634–643.
  • Goldstein LS. Axonal transport and neurodegenerative disease: can we see the elephant? Prog Neurobiol. 2012;99(3):186–190.
  • Liu YH, Ruus P, Steinstraesser A, et al. Effect of the GLP-1 agonist lixisenatide on the pharmacokinetics of warfarin. Diabetes. 2010;59:A558–A558.
  • Hunter K, Holscher C. Drugs developed to treat diabetes, liraglutide and lixisenatide, cross the blood brain barrier and enhance neurogenesis. BMC Neuroscience. 2012;13:33.
  • Dungan KM, Povedano ST, Forst T, et al. Once-weekly dulaglutide versus once-daily liraglutide in metformin-treated patients with type 2 diabetes (AWARD-6): a randomised, open-label, phase 3, non-inferiority trial. Lancet. 2014;384(9951):1349–1357.
  • Green BD, Lavery KS, Irwin N, et al. Novel glucagon-like peptide-1 (GLP-1) analog (Val8)GLP-1 results in significant improvements of glucose tolerance and pancreatic beta-cell function after 3-week daily administration in obese diabetic (ob/ob) mice. J Pharmacol Exp Ther. 2006;318(2):914–921.
  • Tomas E, Stanojevic V, Habener JF. GLP-1-derived nonapeptide GLP-1(28-36)amide targets to mitochondria and suppresses glucose production and oxidative stress in isolated mouse hepatocytes. Regul Pept. 2011;167(2–3):177–184.
  • Janzen KM, Steuber TD, Nisly SA. GLP-1 agonists in type 1 diabetes mellitus. Ann Pharmacother. 2016;50(8):656–665.
  • Shiraki A, Oyama J, Komoda H, et al. The glucagon-like peptide 1 analog liraglutide reduces TNF-alpha-induced oxidative stress and inflammation in endothelial cells. Atherosclerosis. 2012;221(2):375–382.
  • Krasner NM, Ido Y, Ruderman NB, et al. Glucagon-like peptide-1 (GLP-1) analog liraglutide inhibits endothelial cell inflammation through a calcium and AMPK dependent mechanism. PLoS One. 2014;9(5):e97554.
  • Qin L, Chong T, Rodriguez R, et al. Glucagon-like peptide-1-mediated modulation of inflammatory pathways in the diabetic brain: relevance to Alzheimer’s disease. Curr Alzheimer Res. 2016;13:1-1.
  • Kim DH, Huh JW, Jang M, et al. Sitagliptin increases tau phosphorylation in the hippocampus of rats with type 2 diabetes and in primary neuron cultures. Neurobiol Dis. 2012;46(1):52–58.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.