open access

Vol 55, No 5 (2021)
Review Article
Submitted: 2021-01-30
Accepted: 2021-05-24
Published online: 2021-08-06
Get Citation

Alzheimer’s disease and type 2 diabetes mellitus: similarities in pathomechanisms lead to therapeutic opportunities

Krzysztof Bednarz1, Joanna Siuda1
·
Pubmed: 34355790
·
Neurol Neurochir Pol 2021;55(5):418-428.
Affiliations
  1. Department of Neurology, School of Medicine in Katowice, Medical University of Silesia, Katowice, Poland

open access

Vol 55, No 5 (2021)
Review articles
Submitted: 2021-01-30
Accepted: 2021-05-24
Published online: 2021-08-06

Abstract

Introduction. Type 2 diabetes mellitus is a metabolic disease the development of which depends on both environmental and genetic factors. The rapid increase in the number of cases observed in recent decades has been associated with the lifestyle predominant in the West, characterised by a high-calorie diet rich in carbohydrates and saturated fatty acids as well as little physical activity and chronic stress. Another disease with growing morbidity is Alzheimer’s disease, a neurodegenerative disorder characterised by progressive dementia.

State of the art. The results of numerous studies indicate many similarities between these two diseases in terms of their pathomechanisms, especially changes in the activity of enzymatic pathways, accumulation of peptides with altered structure, and chronic inflammation. Amyloid β, hyperphosphorylated tau protein, amylin, and apolipoprotein J are involved in both pathologies. The reasons for their excessive accumulation are not fully understood, but cellular metabolism disorders associated with insulin resistance and diabetes mellitus may play a key role in this process.
It is highly probable that the changes observed at cellular level, which translate into the clinical state of patients, are caused by many abnormalities common to both diseases.

Clinical implications. The discovery of pathophysiological similarities has resulted in attempts to use antidiabetic drugs in Alzheimer’s disease therapy. While animal studies have revealed the potential benefits of oral antidiabetic drugs, studies on humans have not provided clear data regarding their effectiveness. Most clinical trial results are promising, but there have also been studies that have shown no significant, or even adverse, effects of these drugs on Alzheimer’s disease course.

Future directions. Undoubtedly, further research is needed to better understand the mechanisms by which the medications used in diabetes treatment affect the nervous system, and further clinical trials to compare the effectiveness of this therapy in patients presenting different clinical conditions at different stages of Alzheimer’s disease.

Abstract

Introduction. Type 2 diabetes mellitus is a metabolic disease the development of which depends on both environmental and genetic factors. The rapid increase in the number of cases observed in recent decades has been associated with the lifestyle predominant in the West, characterised by a high-calorie diet rich in carbohydrates and saturated fatty acids as well as little physical activity and chronic stress. Another disease with growing morbidity is Alzheimer’s disease, a neurodegenerative disorder characterised by progressive dementia.

State of the art. The results of numerous studies indicate many similarities between these two diseases in terms of their pathomechanisms, especially changes in the activity of enzymatic pathways, accumulation of peptides with altered structure, and chronic inflammation. Amyloid β, hyperphosphorylated tau protein, amylin, and apolipoprotein J are involved in both pathologies. The reasons for their excessive accumulation are not fully understood, but cellular metabolism disorders associated with insulin resistance and diabetes mellitus may play a key role in this process.
It is highly probable that the changes observed at cellular level, which translate into the clinical state of patients, are caused by many abnormalities common to both diseases.

Clinical implications. The discovery of pathophysiological similarities has resulted in attempts to use antidiabetic drugs in Alzheimer’s disease therapy. While animal studies have revealed the potential benefits of oral antidiabetic drugs, studies on humans have not provided clear data regarding their effectiveness. Most clinical trial results are promising, but there have also been studies that have shown no significant, or even adverse, effects of these drugs on Alzheimer’s disease course.

Future directions. Undoubtedly, further research is needed to better understand the mechanisms by which the medications used in diabetes treatment affect the nervous system, and further clinical trials to compare the effectiveness of this therapy in patients presenting different clinical conditions at different stages of Alzheimer’s disease.

Get Citation

Keywords

Alzheimer’s disease, type 2 diabetes mellitus, insulin resistance, dementia, antidiabetic drugs

About this article
Title

Alzheimer’s disease and type 2 diabetes mellitus: similarities in pathomechanisms lead to therapeutic opportunities

Journal

Neurologia i Neurochirurgia Polska

Issue

Vol 55, No 5 (2021)

Article type

Review Article

Pages

418-428

Published online

2021-08-06

Page views

7487

Article views/downloads

1129

DOI

10.5603/PJNNS.a2021.0056

Pubmed

34355790

Bibliographic record

Neurol Neurochir Pol 2021;55(5):418-428.

Keywords

Alzheimer’s disease
type 2 diabetes mellitus
insulin resistance
dementia
antidiabetic drugs

Authors

Krzysztof Bednarz
Joanna Siuda

References (100)
  1. Saeedi P, Salpea P, Karuranga S, et al. IDF Diabetes Atlas Committee. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9 edition. Diabetes Res Clin Pract. 2019; 157: 107843.
  2. Akash MS, Rehman K, Chen S, et al. Role of inflammatory mechanisms in pathogenesis of type 2 diabetes mellitus. J Cell Biochem. 2013; 114(3): 525–531.
  3. Papatheodorou K, Papanas N, Banach M, et al. Complications of Diabetes 2016. J Diabetes Res. 2016; 2016: 6989453.
  4. Biessels GJ, Despa F. Cognitive decline and dementia in diabetes mellitus: mechanisms and clinical implications. Nat Rev Endocrinol. 2018; 14(10): 591–604.
  5. Rönnemaa E, Zethelius B, Sundelöf J, et al. Impaired insulin secretion increases the risk of Alzheimer disease. Neurology. 2008; 71(14): 1065–1071.
  6. S Roriz-Filho J, Sá-Roriz TM, Rosset I, et al. (Pre)diabetes, brain aging, and cognition. Biochim Biophys Acta. 2009; 1792(5): 432–443.
  7. Sun Y, Ma C, Sun H, et al. Metabolism: a novel shared link between diabetes mellitus and Alzheimer's disease. J Diabetes Res. 2020; 2020: 4981814.
  8. Nguyen TT, Ta QT, Nguyen TK, et al. Type 3 diabetes and its role implications in Alzheimer's disease. Int J Mol Sci. 2020; 21(9).
  9. Neth BJ, Craft S. Insulin resistance and Alzheimer's disease: bioenergetic linkages. Front Aging Neurosci. 2017; 9: 345.
  10. Weller J, Budson A. Current understanding of Alzheimer's disease diagnosis and treatment. F1000Res. 2018; 7.
  11. Sokołowska N, Sokołowski R, Oleksy E, et al. Usefulness of the Polish versions of the Montreal Cognitive Assessment 7.2 and the Mini-Mental State Examination as screening instruments for the detection of mild neurocognitive disorder. Neurol Neurochir Pol. 2020; 54(5): 440–448.
  12. Barc K, Kuźma-Kozakiewicz M. Positron emission tomography neuroimaging in neurodegenerative diseases: Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Neurol Neurochir Pol. 2019; 53(2): 99–112.
  13. De Sousa RA, Harmer AR, Freitas DA, et al. An update on potential links between type 2 diabetes mellitus and Alzheimer's disease. Mol Biol Rep. 2020; 47(8): 6347–6356.
  14. Knopman DS, Jack CR, Wiste HJ, et al. 18F-fluorodeoxyglucose positron emission tomography, aging, and apolipoprotein E genotype in cognitively normal persons. Neurobiol Aging. 2014; 35(9): 2096–2106.
  15. Monte Sd, Wands J. Alzheimer's Disease is type 3 diabetes—evidence reviewed. Journal of Diabetes Science and Technology. 2008; 2(6): 1101–1113.
  16. Meng L, Li XY, Shen L, et al. Type 2 diabetes mellitus drugs for alzheimer's disease: current evidence and therapeutic opportunities. Trends Mol Med. 2020; 26(6): 597–614.
  17. Van Dyken P, Lacoste B. Impact of metabolic syndrome on Neuroinflammation and the blood-brain barrier. Front Neurosci. 2018; 12: 930.
  18. Kellar D, Craft S. Brain insulin resistance in Alzheimer's disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol. 2020; 19(9): 758–766.
  19. Arnold SE, Arvanitakis Z, Macauley-Rambach SL, et al. Brain insulin resistance in type 2 diabetes and Alzheimer disease: concepts and conundrums. Nat Rev Neurol. 2018; 14(3): 168–181.
  20. Gabbouj S, Ryhänen S, Marttinen M, et al. Altered Insulin Signaling in Alzheimer's Disease Brain - Special Emphasis on PI3K-Akt Pathway. Front Neurosci. 2019; 13: 629.
  21. de la Monte SM, Wands JR. Alzheimer's disease is type 3 diabetes-evidence reviewed. J Diabetes Sci Technol. 2008; 2(6): 1101–1113.
  22. Sims-Robinson C, Kim B, Rosko A, et al. How does diabetes accelerate Alzheimer disease pathology? Nat Rev Neurol. 2010; 6(10): 551–559.
  23. Banks WA, Owen JB, Erickson MA. Insulin in the brain: there and back again. Pharmacol Ther. 2012; 136(1): 82–93.
  24. Rhea EM, Banks WA. Role of the blood-brain barrier in central nervous system insulin resistance. Front Neurosci. 2019; 13: 521.
  25. Kleinridders A, Ferris HA, Cai W, et al. Insulin action in brain regulates systemic metabolism and brain function. Diabetes. 2014; 63(7): 2232–2243.
  26. Ahn KC, Learman CR, Baker GB, et al. Regulation of diabetes: a therapeutic strategy for Alzheimer's disease? J Korean Med Sci. 2019; 34(46): e297.
  27. Frazier HN, Ghoweri AO, Anderson KL, et al. Broadening the definition of brain insulin resistance in aging and Alzheimer's disease. Exp Neurol. 2019; 313: 79–87.
  28. Zilliox LA, Chadrasekaran K, Kwan JY, et al. Diabetes and cognitive impairment. Curr Diab Rep. 2016; 16(9): 87.
  29. Rankin CA, Sun Q, Gamblin TC. Tau phosphorylation by GSK-3beta promotes tangle-like filament morphology. Mol Neurodegener. 2007; 2: 12.
  30. Haque R, Nazir A. Insulin-degrading enzyme: a link between Alzheimer's and type 2 diabetes mellitus. CNS Neurol Disord Drug Targets. 2014; 13(2): 259–264.
  31. Zhao L, Teter B, Morihara T, et al. Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: implications for Alzheimer's disease intervention. J Neurosci. 2004; 24(49): 11120–11126.
  32. Tundo GR, Sbardella D, Ciaccio C, et al. Multiple functions of insulin-degrading enzyme: a metabolic crosslight? Crit Rev Biochem Mol Biol. 2017; 52(5): 554–582.
  33. Madhusudhanan J, Suresh G, Devanathan V. Neurodegeneration in type 2 diabetes: Alzheimer's as a case study. Brain Behav. 2020; 10(5): e01577.
  34. Mietlicki-Baase EG. Amylin-mediated control of glycemia, energy balance, and cognition. Physiol Behav. 2016; 162: 130–140.
  35. Brito-Moreira J, Lourenco MV, Oliveira MM, et al. Interaction of amyloid-β (Aβ) oligomers with neurexin 2α and neuroligin 1 mediates synapse damage and memory loss in mice. J Biol Chem. 2017; 292(18): 7327–7337.
  36. Lutz TA, Meyer U. Amylin at the interface between metabolic and neurodegenerative disorders. Front Neurosci. 2015; 9: 216.
  37. Stanciu GD, Bild V, Ababei DC, et al. Link between diabetes and Alzheimer's disease due to the shared amyloid aggregation and deposition involving both neurodegenerative changes and neurovascular damages. J Clin Med. 2020; 9(6).
  38. Grizzanti J, Corrigan R, Servizi S, et al. Neuroprotective effects of amylin analogues on Alzheimer's disease pathogenesis and cognition. J Alzheimers Dis. 2018; 66(1): 11–23.
  39. Martinez-Valbuena I, Valenti-Azcarate R, Amat-Villegas I, et al. Amylin as a potential link between type 2 diabetes and alzheimer disease. Ann Neurol. 2019; 86(4): 539–551.
  40. Miklossy J, Qing H, Radenovic A, et al. Beta amyloid and hyperphosphorylated tau deposits in the pancreas in type 2 diabetes. Neurobiol Aging. 2010; 31(9): 1503–1515.
  41. Despa F, Goldstein LB, Biessels GJ. Amylin as a potential link between type 2 diabetes and Alzheimer disease. Ann Neurol. 2020; 87(3): 486.
  42. Kim N, Yoo JC, Han JY, et al. Human nuclear clusterin mediates apoptosis by interacting with Bcl-XL through C-terminal coiled coil domain. J Cell Physiol. 2012; 227(3): 1157–1167.
  43. Ha J, Moon MK, Kim H, et al. Plasma clusterin as a potential link between diabetes and Alzheimer disease. J Clin Endocrinol Metab. 2020; 105(9).
  44. Wang W, Zhao F, Ma X, et al. Mitochondria dysfunction in the pathogenesis of Alzheimer's disease: recent advances. Mol Neurodegener. 2020; 15(1): 30.
  45. Wijesekara N, Gonçalves RA, De Felice FG, et al. Impaired peripheral glucose homeostasis and Alzheimer's disease. Neuropharmacology. 2018; 136(Pt B): 172–181.
  46. Rigotto G, Basso E. Mitochondrial dysfunctions: a thread sewing together Alzheimer's disease, diabetes, and obesity. Oxid Med Cell Longev. 2019; 2019: 7210892.
  47. Wang CH, Wei YH. Role of mitochondrial dysfunction and dysregulation of Ca homeostasis in the pathophysiology of insulin resistance and type 2 diabetes. J Biomed Sci. 2017; 24(1): 70.
  48. Karmi A, Iozzo P, Viljanen A, et al. Increased brain fatty acid uptake in metabolic syndrome. Diabetes. 2010; 59(9): 2171–2177.
  49. Gupta S, Knight AG, Gupta S, et al. Saturated long-chain fatty acids activate inflammatory signaling in astrocytes. J Neurochem. 2012; 120(6): 1060–1071.
  50. Pugazhenthi S, Qin L, Reddy PH. Common neurodegenerative pathways in obesity, diabetes, and Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis. 2017; 1863(5): 1037–1045.
  51. Zhang R, Miller RG, Madison C, et al. Systemic immune system alterations in early stages of Alzheimer's disease. J Neuroimmunol. 2013; 256(1-2): 38–42.
  52. 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.
  53. Cummings JL, Morstorf T, Zhong K. Alzheimer's disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther. 2014; 6(4): 37.
  54. Egan MF, Kost J, Voss T, et al. Randomized trial of verubecestat for prodromal Alzheimer's disease. N Engl J Med. 2019; 380(15): 1408–1420.
  55. Egan MF, Kost J, Tariot PN, et al. Randomized trial of verubecestat for mild-to-moderate Alzheimer's disease. N Engl J Med. 2018; 378(18): 1691–1703.
  56. Egan MF, Mukai Y, Voss T, et al. Further analyses of the safety of verubecestat in the phase 3 EPOCH trial of mild-to-moderate Alzheimer's disease. Alzheimers Res Ther. 2019; 11(1): 68.
  57. Linnemann C, Lang UE. Pathways connecting late-life depression and dementia. Front Pharmacol. 2020; 11: 279.
  58. Reger MA, Watson GS, Green PS, et al. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology. 2008; 70(6): 440–448.
  59. Craft S, Claxton A, Baker LD, et al. Long-acting intranasal insulin detemir improves cognition for adults with mild cognitive impairment or early-stage Alzheimer's disease dementia. J Alzheimers Dis. 2015; 44(3): 897–906.
  60. Craft S, Claxton A, Baker LD, et al. Effects of regular and long-acting insulin on cognition and alzheimer's disease biomarkers: a pilot clinical trial. J Alzheimers Dis. 2017; 57(4): 1325–1334.
  61. Craft S, Raman R, Chow TW, et al. Safety, efficacy, and feasibility of intranasal insulin for the treatment of mild cognitive impairment and alzheimer disease dementia: a randomized clinical trial. JAMA Neurol. 2020; 77(9): 1099–1109.
  62. Chapman CD, Schiöth HB, Grillo CA, et al. Intranasal insulin in Alzheimer's disease: Food for thought. Neuropharmacology. 2018; 136(Pt B): 196–201.
  63. Schmid V, Kullmann S, Gfrörer W, et al. Safety of intranasal human insulin: A review. Diabetes Obes Metab. 2018; 20(7): 1563–1577.
  64. Tumminia A, Vinciguerra F, Parisi M, et al. Type 2 diabetes mellitus and Alzheimer's disease: role of insulin signalling and therapeutic implications. Int J Mol Sci. 2018; 19(11).
  65. Claxton A, Baker LD, Wilkinson CW, et al. Sex and ApoE genotype differences in treatment response to two doses of intranasal insulin in adults with mild cognitive impairment or Alzheimer's disease. J Alzheimers Dis. 2013; 35(4): 789–797.
  66. Zhao Na, Liu CC, Van Ingelgom AJ, et al. Apolipoprotein E4 Impairs Neuronal Insulin Signaling by Trapping Insulin Receptor in the Endosomes. Neuron. 2017; 96(1): 115–129.e5.
  67. Koenig AM, Mechanic-Hamilton D, Xie SX, et al. Effects of the insulin sensitizer metformin in Alzheimer disease: pilot data from a randomized placebo-controlled crossover study. Alzheimer Dis Assoc Disord. 2017; 31(2): 107–113.
  68. Chiang MC, Cheng YC, Chen SJ, et al. Metformin activation of AMPK-dependent pathways is neuroprotective in human neural stem cells against Amyloid-beta-induced mitochondrial dysfunction. Exp Cell Res. 2016; 347(2): 322–331.
  69. Luchsinger JA, Perez T, Chang H, et al. Metformin in amnestic mild cognitive impairment: results of a pilot randomized placebo controlled clinical trial. J Alzheimers Dis. 2016; 51(2): 501–514.
  70. Herath PM, Cherbuin N, Eramudugolla R, et al. The effect of diabetes medication on cognitive function: evidence from the PATH through life study. Biomed Res Int. 2016; 2016: 7208429.
  71. Moore EM, Mander AG, Ames D, et al. AIBL Investigators. Increased risk of cognitive impairment in patients with diabetes is associated with metformin. Diabetes Care. 2013; 36(10): 2981–2987.
  72. Imfeld P, Bodmer M, Jick SS, et al. Metformin, other antidiabetic drugs, and risk of Alzheimer's disease: a population-based case-control study. J Am Geriatr Soc. 2012; 60(5): 916–921.
  73. Hamilton A, Hölscher C. Receptors for the incretin glucagon-like peptide-1 are expressed on neurons in the central nervous system. Neuroreport. 2009; 20(13): 1161–1166.
  74. 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.
  75. 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.
  76. Batista AF, Forny-Germano L, Clarke JR, 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.
  77. Zhang Y, Xie JZ, Xu XY, et al. Liraglutide ameliorates hyperhomocysteinemia-induced Alzheimer-like pathology and memory deficits in rats via multi-molecular targeting. Neurosci Bull. 2019; 35(4): 724–734.
  78. 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.
  79. McClean PL, Parthsarathy V, Faivre E, et al. The novel GLP-1 analogue liraglutide has neuroprotective properties in a mouse model of Alzheimer's disease. Regulatory Peptides. 2010; 164(1): 40.
  80. Hansen HH, Fabricius K, Barkholt P, 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): 877–888.
  81. Watson KT, Wroolie TE, Tong G, et al. Neural correlates of liraglutide effects in persons at risk for Alzheimer's disease. Behav Brain Res. 2019; 356: 271–278.
  82. Femminella GD, Frangou E, Love SB, et al. Evaluating the effects of the novel GLP-1 analogue liraglutide in Alzheimer's disease: study protocol for a randomised controlled trial (ELAD study). Trials. 2019; 20(1): 191.
  83. Kosaraju J, Holsinger RM, Guo L, et al. Linagliptin, a dipeptidyl peptidase-4 inhibitor, mitigates cognitive deficits and pathology in the 3xTg-AD mouse model of Alzheimer's disease. Mol Neurobiol. 2017; 54(8): 6074–6084.
  84. Kornelius E, Lin CL, Chang HH, et al. DPP-4 inhibitor linagliptin attenuates Aβ-induced cytotoxicity through activation of AMPK in neuronal cells. CNS Neurosci Ther. 2015; 21(7): 549–557.
  85. Isik AT, Soysal P, Yay A, et al. The effects of sitagliptin, a DPP-4 inhibitor, on cognitive functions in elderly diabetic patients with or without Alzheimer's disease. Diabetes Res Clin Pract. 2017; 123: 192–198.
  86. Landreth G, Jiang Q, Mandrekar S, et al. PPARgamma agonists as therapeutics for the treatment of Alzheimer's disease. Neurotherapeutics. 2008; 5(3): 481–489.
  87. Muñoz-Jiménez M, Zaarkti A, García-Arnés JA, et al. Antidiabetic drugs in Alzheimer's disease and mild cognitive impairment: A systematic review. Dement Geriatr Cogn Disord. 2020; 49(5): 423–434.
  88. Quan Q, Qian Y, Li Xi, et al. Pioglitazone reduces β amyloid levels inhibition of PPARγ phosphorylation in a neuronal model of Alzheimer's disease. Front Aging Neurosci. 2019; 11: 178.
  89. Hanyu H, Sato T, Sakurai H, et al. The role of tumor necrosis factor-alpha in cognitive improvement after peroxisome proliferator-activator receptor gamma agonist pioglitazone treatment in Alzheimer's disease. J Am Geriatr Soc. 2010; 58(5): 1000–1001.
  90. Fernandez-Martos CM, Atkinson RAK, Chuah MI, et al. Combination treatment with leptin and pioglitazone in a mouse model of Alzheimer's disease. Alzheimers Dement (N Y). 2017; 3(1): 92–106.
  91. Knodt AR, Burke JR, Welsh-Bohmer KA, et al. Effects of pioglitazone on mnemonic hippocampal function: A blood oxygen level-dependent functional magnetic resonance imaging study in elderly adults. Alzheimers Dement (N Y). 2019; 5: 254–263.
  92. Hanyu H, Sato T, Kiuchi A, et al. Pioglitazone improved cognition in a pilot study on patients with Alzheimer's disease and mild cognitive impairment with diabetes mellitus. J Am Geriatr Soc. 2009; 57(1): 177–179.
  93. Sato T, Hanyu H, Hirao K, et al. Efficacy of PPAR-γ agonist pioglitazone in mild Alzheimer disease. Neurobiol Aging. 2011; 32(9): 1626–1633.
  94. Li H, Wu J, Zhu L, et al. Insulin degrading enzyme contributes to the pathology in a mixed model of Type 2 diabetes and Alzheimer's disease: possible mechanisms of IDE in T2D and AD. Biosci Rep. 2018; 38(1).
  95. Xu S, Guan Q, Wang C, et al. Rosiglitazone prevents the memory deficits induced by amyloid-beta oligomers via inhibition of inflammatory responses. Neurosci Lett. 2014; 578: 7–11.
  96. Risner ME, Saunders AM, Altman JFB, et al. Rosiglitazone in Alzheimer's Disease Study Group. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer's disease. Pharmacogenomics J. 2006; 6(4): 246–254.
  97. Gold M, Alderton C, Zvartau-Hind M, et al. Rosiglitazone monotherapy in mild-to-moderate Alzheimer's disease: results from a randomized, double-blind, placebo-controlled phase III study. Dement Geriatr Cogn Disord. 2010; 30(2): 131–146.
  98. Grizzanti J, Corrigan R, Casadesus G. Neuroprotective effects of amylin analogues on Alzheimer's disease pathogenesis and cognition. J Alzheimers Dis. 2018; 66(1): 11–23.
  99. Zhu H, Wang X, Wallack M, et al. Intraperitoneal injection of the pancreatic peptide amylin potently reduces behavioral impairment and brain amyloid pathology in murine models of Alzheimer's disease. Mol Psychiatry. 2015; 20(2): 252–262.
  100. Soudy R, Patel A, Fu W, et al. Cyclic AC253, a novel amylin receptor antagonist, improves cognitive deficits in a mouse model of Alzheimer's disease. Alzheimers Dement (N Y). 2017; 3(1): 44–56.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk, Poland
tel.:+48 58 320 94 94, fax:+48 58 320 94 60, e-mail: viamedica@viamedica.pl