Generic placeholder image

Current Neuropharmacology

Editor-in-Chief

ISSN (Print): 1570-159X
ISSN (Online): 1875-6190

Review Article

Current Progress on Peroxisome Proliferator-activated Receptor Gamma Agonist as an Emerging Therapeutic Approach for the Treatment of Alzheimer's Disease: An Update

Author(s): Mahmood Ahmad Khan*, Qamre Alam, Absarul Haque, Mohammad Ashafaq, Mohd Jahir Khan, Ghulam Md Ashraf* and Mahboob Ahmad

Volume 17, Issue 3, 2019

Page: [232 - 246] Pages: 15

DOI: 10.2174/1570159X16666180828100002

Price: $65

Abstract

Alzheimer’s disease (AD) is an age-related progressive neurodegenerative disorder, characterized by the deposition of amyloid-β within the brain parenchyma resulting in a significant decline in cognitive functions. The pathophysiological conditions of the disease are recognized by the perturbation of synaptic function, energy and lipid metabolism. In Addition deposition of amyloid plaques also triggers inflammation upon the induction of microglia. Peroxisome proliferatoractivated receptors (PPARs) are ligand-activated transcription factors known to play important role in the regulation of glucose absorption, homeostasis of lipid metabolism and are further known to involved in repressing the expression of genes related to inflammation. Therefore, agonists of this receptor represent an attractive therapeutic target for AD. Recently, both clinical and preclinical studies showed that use of Peroxisome proliferator-activated receptor gamma (PPARγ) agonist improves both learning and memory along with other AD related pathology. Thus, PPARγ signifies a significant new therapeutic target in treating AD. In this review, we have shed some light on the recent progress of how, PPARγ agonist selectively modulated different cellular targets in AD and its amazing potential in the treatment of AD.

Keywords: Alzheimer's disease, Peroxisome proliferator-activated receptors, Transactivation, β-amyloid, Thiazolidinedione, Insulin sensitivity, Rosiglitazone, Blood-brain-barrier.

Graphical Abstract
[1]
Hebert LE, Weuve J, Scherr PA, Evans DA. Alzheimer disease in the United States (2010-2050) estimated using the 2010 census. Neurology 2013; 80(19): 1778-83.
[http://dx.doi.org/ 10.1212/WNL.0b013e31828726f5] [PMID: 23390181]
[2]
Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med 2010; 362(4): 329-44.
[http://dx.doi.org/10.1056/NEJMra 0909142] [PMID: 20107219]
[3]
Manczak M, Anekonda TS, Henson E, Park BS, Quinn J, Reddy PH. Mitochondria are a direct site of A beta accumulation in Alzheimer’s disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum Mol Genet 2006; 15(9): 1437-49.
[http://dx.doi.org/10.1093/hmg/ddl066] [PMID: 16551656]
[4]
Reddy PH, Beal MF. Amyloid beta, mitochondrial dysfunction and synaptic damage: implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med 2008; 14(2): 45-53.
[http://dx.doi.org/ dx.doi.org/10.1016/j.molmed.2007.12.002] [PMID: 18218341]
[5]
Fratiglioni L, Qiu C. Prevention of common neurodegenerative disorders in the elderly. Exp Gerontol 2009; 44(1-2): 46-50.
[http://dx.doi.org/10.1016/j.exger.2008.06.006] [PMID: 18620039]
[6]
Cameron B, Landreth GE. Inflammation, microglia, and Alzheimer’s disease. Neurobiol Dis 2010; 37(3): 503-9.
[http://dx.doi.org/ dx.doi.org/10.1016/j.nbd.2009.10.006] [PMID: 19833208]
[7]
Mandrekar-Colucci S, Landreth GE. Nuclear receptors as therapeutic targets for Alzheimer’s disease. Expert Opin Ther Targets 2011; 15(9): 1085-97.
[http://dx.doi.org/10.1517/14728222.2011. 594043] [PMID: 21718217]
[8]
Zolezzi JM, Inestrosa NC. Peroxisome proliferator-activated receptors and Alzheimer’s disease: hitting the blood-brain barrier. Mol Neurobiol 2013; 48(3): 438-51.
[http://dx.doi.org/10. 1007/s12035-013-8435-5] [PMID: 23494748]
[9]
Wang L, Waltenberger B, Pferschy-Wenzig EM, et al. Natural product agonists of peroxisome proliferator-activated receptor gamma (PPARγ): a review. Biochem Pharmacol 2014; 92(1): 73-89.
[http://dx.doi.org/ 10.1016/j.bcp.2014.07.018] [PMID: 25083916]
[10]
Bookout AL, Jeong Y, Downes M, Yu RT, Evans RM, Mangelsdorf DJ. Anatomical profiling of nuclear receptor expression reveals a hierarchical transcriptional network. Cell 2006; 126(4): 789-99.
[http://dx.doi.org/10.1016/j.cell.2006.06.049] [PMID: 16923397]
[11]
Shie FS, Nivison M, Hsu PC, Montine TJ. Modulation of microglial innate immunity in Alzheimer’s disease by activation of peroxisome proliferator-activated receptor gamma. Curr Med Chem 2009; 16(6): 643-51.
[http://dx.doi.org/10.2174/092986709787458399] [PMID: 19199928]
[12]
Feinstein DL, Galea E, Gavrilyuk V, et al. Peroxisome proliferator-activated receptor-gamma agonists prevent experimental autoimmune encephalomyelitis. Ann Neurol 2002; 51(6): 694-702.
[http://dx.doi.org/ 10.1002/ana.10206] [PMID: 12112074]
[13]
Choi JM, Bothwell AL. The nuclear receptor PPARs as important regulators of T-cell functions and autoimmune diseases. Mol Cells 2012; 33(3): 217-22.
[http://dx.doi.org/10.1007/s10059-012-2297-y] [PMID: 22382683]
[14]
Nagy L, Tontonoz P, Alvarez JG, Chen H, Evans RM. Oxidized LDL regulates macrophage gene expression through ligand activation of PPARgamma. Cell 1998; 93(2): 229-40.
[http://dx.doi.org/dx. doi.org/10.1016/S0092-8674(00)81574-3] [PMID: 9568715]
[15]
Azhar S. Peroxisome proliferator-activated receptors, metabolic syndrome and cardiovascular disease. Future Cardiol 2010; 6(5): 657-91.
[http://dx.doi.org/10.2217/fca.10.86] [PMID: 20932114]
[16]
Scheen AJ. Thiazolidinediones and liver toxicity. Diabetes Metab 2001; 27(3): 305-13.
[PMID: 11431595]
[17]
Kummer MP, Schwarzenberger R, Sayah-Jeanne S, et al. Pan-PPAR modulation effectively protects APP/PS1 mice from amyloid deposition and cognitive deficits. Mol Neurobiol 2015; 51(2): 661-71.
[http://dx.doi.org/10.1007/s12035-014-8743-4] [PMID: 24838579]
[18]
Marketou ME, Kontaraki JE, Tsakountakis NA, et al. Differential effect of telmisartan and amlodipine on monocyte chemoattractant protein-1 and peroxisome proliferator-activated receptor-gamma gene expression in peripheral monocytes in patients with essential hypertension. Am J Cardiol 2011; 107(1): 59-63.
[http://dx.doi.org/10.1016/j.amjcard. 2010.08.048] [PMID: 21146687]
[19]
Sukumaran V, Watanabe K, Veeraveedu PT, et al. Telmisartan ameliorates experimental autoimmune myocarditis associated with inhibition of inflammation and oxidative stress. Eur J Pharmacol 2011; 652(1-3): 126-35.
[http://dx.doi.org/dx. doi.org/10.1016/j.ejphar.2010.10.081] [PMID: 21115000]
[20]
Shindo T, Takasaki K, Uchida K, et al. Ameliorative effects of telmisartan on the inflammatory response and impaired spatial memory in a rat model of Alzheimer’s disease incorporating additional cerebrovascular disease factors. Biol Pharm Bull 2012; 35(12): 2141-7.
[http://dx.doi.org/10.1248/bpb.b12-00387] [PMID: 23207766]
[21]
Singh B, Sharma B, Jaggi AS, Singh N. Attenuating effect of lisinopril and telmisartan in intracerebroventricular streptozotocin induced experimental dementia of Alzheimer’s disease type: possible involvement of PPAR-γ agonistic property. J Renin Angiotensin Aldosterone Syst 2013; 14(2): 124-36.
[http://dx.doi.org/dx.doi. org/10.1177/1470320312459977] [PMID: 23060470]
[22]
Tsukuda K, Mogi M, Iwanami J, et al. Cognitive deficit in amyloid-beta-injected mice was improved by pretreatment with a low dose of telmisartan partly because of peroxisome proliferator-activated receptor-gamma activation. Hypertension 2009; 54(4): 782-7.
[http://dx.doi.org/ 10.1161/HYPERTENSIONAHA.109.136879] [PMID: 19635982]
[23]
Guo L, Tabrizchi R. Peroxisome proliferator-activated receptor gamma as a drug target in the pathogenesis of insulin resistance. Pharmacol Ther 2006; 111(1): 145-73.
[http://dx.doi.org/10. 1016/j.pharmthera.2005.10.009] [PMID: 16305809]
[24]
Fajas L, Fruchart JC, Auwerx J. Transcriptional control of adipogenesis. Curr Opin Cell Biol 1998; 10(2): 165-73.
[http://dx.doi.org/ dx.doi.org/10.1016/S0955-0674(98)80138-5] [PMID: 9561840]
[25]
Bensinger SJ, Tontonoz P. Integration of metabolism and inflammation by lipid-activated nuclear receptors. Nature 2008; 454(7203): 470-7.
[http://dx.doi.org/10.1038/nature07202] [PMID: 18650918]
[26]
Chu K, Lee ST, Koo JS, et al. Peroxisome proliferator-activated receptor-gamma-agonist, rosiglitazone, promotes angiogenesis after focal cerebral ischemia. Brain Res 2006; 1093(1): 208-18.
[http://dx.doi.org/10.1016/j.brainres. 2006.03.114] [PMID: 16696956]
[27]
Liu YW, Zhu X, Zhang L, et al. Cerebroprotective effects of ibuprofen on diabetic encephalopathy in rats. Pharmacol Biochem Behav 2014; 117: 128-36.
[http://dx.doi.org/10.1016/j.pbb.2013.11.027] [PMID: 24291733]
[28]
Pisanu A, Lecca D, Mulas G, et al. Dynamic changes in pro- and anti-inflammatory cytokines in microglia after PPAR-γ agonist neuroprotective treatment in the MPTPp mouse model of progressive Parkinson’s disease. Neurobiol Dis 2014; 71: 280-91.
[http://dx.doi.org/10. 1016/j.nbd.2014.08.011] [PMID: 25134730]
[29]
Croasdell A, Duffney PF, Kim N, Lacy SH, Sime PJ, Phipps RP. PPARγ and the Innate Immune System Mediate the Resolution of Inflammation. PPAR Res 2015; 2015: 549691.
[http://dx.doi.org/10.1155/2015/549691] [PMID: 26713087]
[30]
Chung SW, Kang BY, Kim SH, et al. Oxidized low density lipoprotein inhibits interleukin-12 production in lipopolysaccharide-activated mouse macrophages via direct interactions between peroxisome proliferator-activated receptor-gamma and nuclear factor-kappa B. J Biol Chem 2000; 275(42): 32681-7.
[http://dx.doi.org/10.1074/jbc.M002577200] [PMID: 10934192]
[31]
Pascual G, Fong AL, Ogawa S, et al. A SUMOylation-dependent pathway mediates transrepression of inflammatory response genes by PPAR-gamma. Nature 2005; 437(7059): 759-63.
[http://dx.doi.org/10.1038/nature03988] [PMID: 16127449]
[32]
Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature 1998; 391(6662): 82-6.
[http://dx.doi.org/10.1038/34184] [PMID: 9422509]
[33]
O’Barr S, Cooper NR. The C5a complement activation peptide increases IL-1beta and IL-6 release from amyloid-beta primed human monocytes: implications for Alzheimer’s disease. J Neuroimmunol 2000; 109(2): 87-94.
[http://dx.doi.org/10.1016/S0165-5728(00)00291-5] [PMID: 10996210]
[34]
Lue LF, Rydel R, Brigham EF, et al. Inflammatory repertoire of Alzheimer’s disease and nondemented elderly microglia in vitro. Glia 2001; 35(1): 72-9.
[http://dx.doi.org/10.1002/glia.1072] [PMID: 11424194]
[35]
Hoozemans JJ, Rozemuller JM, van Haastert ES, Veerhuis R, Eikelenboom P. Cyclooxygenase-1 and -2 in the different stages of Alzheimer’s disease pathology. Curr Pharm Des 2008; 14(14): 1419-27.
[http://dx.doi.org/10.2174/138161208784480171] [PMID: 18537664]
[36]
Mandrekar-Colucci S, Karlo JC, Landreth GE. Mechanisms underlying the rapid peroxisome proliferator-activated receptor-γ-mediated amyloid clearance and reversal of cognitive deficits in a murine model of Alzheimer’s disease. J Neurosci 2012; 32(30): 10117-28.
[http://dx.doi.org/10.1523/JNEUROSCI.5268-11. 2012] [PMID: 22836247]
[37]
Skerrett R, Pellegrino MP, Casali BT, Taraboanta L, Landreth GE. Combined liver X receptor/peroxisome proliferator-activated receptor γ agonist treatment reduces amyloid β levels and improves behavior in amyloid precursor protein/presenilin 1 mice. J Biol Chem 2015; 290(35): 21591-602.
[http://dx.doi.org/ 10.1074/jbc.M115.652008] [PMID: 26163517]
[38]
Yamanaka M, Ishikawa T, Griep A, Axt D, Kummer MP, Heneka MT. PPARγ/RXRα-induced and CD36-mediated microglial amyloid-β phagocytosis results in cognitive improvement in amyloid precursor protein/presenilin 1 mice. J Neurosci 2012; 32(48): 17321-31.
[http://dx.doi.org/10.1523/JNEUROSCI.1569-12.2012] [PMID: 23197723]
[39]
Escribano L, Simón AM, Gimeno E, Cuadrado-Tejedor M. López de, Maturana, R.; García-Osta, A. Ricobaraza, A.; Pérez-Mediavilla, A.; Del, Río, J.; Frechilla, D. Rosiglitazone rescues memory impairment in Alzheimer’s transgenic mice: mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacol 2010; 35: 1593-604.
[http://dx.doi.org/10.1038/npp. 2010.32]
[40]
O’Reilly JA, Lynch M. Rosiglitazone improves spatial memory and decreases insoluble Aβ(1-42) in APP/PS1 mice. J Neuroimmune Pharmacol 2012; 7(1): 140-4.
[http://dx.doi.org/10.1007/s11481-011-9282-7] [PMID: 21617889]
[41]
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.
[http://dx.doi.org/10.1016/j.neulet.2014.06.010] [PMID: 24933538]
[42]
Cernuda-Morollón E, Rodríguez-Pascual F, Klatt P, Lamas S, Pérez-Sala D. PPAR agonists amplify iNOS expression while inhibiting NF-kappaB: implications for mesangial cell activation by cytokines. J Am Soc Nephrol 2002; 13(9): 2223-31.
[http://dx.doi.org/10.1097/01.ASN.0000025786.87646.B1] [PMID: 12191966]
[43]
Park EJ, Park SY, Joe EH, Jou I. 15d-PGJ2 and rosiglitazone suppress Janus kinase-STAT inflammatory signaling through induction of suppressor of cytokine signaling 1 (SOCS1) and SOCS3 in glia. J Biol Chem 2003; 278(17): 14747-52.
[http://dx.doi.org/dx.doi. org/10.1074/jbc.M210819200] [PMID: 12584205]
[44]
Straus DS, Pascual G, Li M, et al. 15-deoxy-delta 12,14-prostaglandin J2 inhibits multiple steps in the NF-kappa B signaling pathway. Proc Natl Acad Sci USA 2000; 97(9): 4844-9.
[http://dx.doi.org/10.1073/pnas.97.9.4844] [PMID: 10781090]
[45]
Lim GP, Yang F, Chu T, et al. Ibuprofen suppresses plaque pathology and inflammation in a mouse model for Alzheimer’s disease. J Neurosci 2000; 20(15): 5709-14.
[http://dx.doi.org/10.1523/JNEUROSCI.20-15-05709.2000] [PMID: 10908610]
[46]
Yan Q, Zhang J, Liu H, et al. Anti-inflammatory drug therapy alters beta-amyloid processing and deposition in an animal model of Alzheimer’s disease. J Neurosci 2003; 23(20): 7504-9.
[http://dx.doi.org/dx. doi.org/10.1523/JNEUROSCI.23-20-07504.2003] [PMID: 12930788]
[47]
Heneka MT, Sastre M, Dumitrescu-Ozimek L, et al. Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain 2005; 128(Pt 6): 1442-53.
[http://dx.doi.org/10.1093/brain/awh452] [PMID: 15817521]
[48]
Liu LP, Yan TH, Jiang LY, et al. Pioglitazone ameliorates memory deficits in streptozotocin-induced diabetic mice by reducing brain β-amyloid through PPARγ activation. Acta Pharmacol Sin 2013; 34(4): 455-63.
[http://dx.doi.org/dx.doi. org/10.1038/aps.2013.11] [PMID: 23524568]
[49]
Sastre M, Dewachter I, Rossner S, et al. Nonsteroidal anti-inflammatory drugs repress beta-secretase gene promoter activity by the activation of PPARgamma. Proc Natl Acad Sci USA 2006; 103(2): 443-8.
[http://dx.doi.org/10. 1073/pnas.0503839103] [PMID: 16407166]
[50]
Toledo EM, Inestrosa NC. 2010.
[51]
Eriksen JL, Sagi SA, Smith TE, et al. NSAIDs and enantiomers of flurbiprofen target gamma-secretase and lower Abeta 42 in vivo. J Clin Invest 2003; 112(3): 440-9.
[http://dx.doi.org/10.1172/JCI18162] [PMID: 12897211]
[52]
Prade E, Bittner HJ, Sarkar R. Lopez, Del, Amo, J.M.; Althoff-Ospelt, G.; Multhaup, G.; Hildebrand, P.W.; Reif, B. Structural mechanism of the interaction of alzheimer disease Aβ fibrils with the non-steroidal anti-inflammatory drug (NSAID). Sulindac Sulfide J Biol Chem 2015; 290: 28737-45.
[http://dx.doi.org/ 10.1074/jbc.M115.675215] [PMID: 26416887]
[53]
Whitmer RA, Sidney S, Selby J, Johnston SC, Yaffe K. Midlife cardiovascular risk factors and risk of dementia in late life. Neurology 2005; 64(2): 277-81.
[http://dx.doi.org/10.1212/01. WNL.0000149519.47454.F2] [PMID: 15668425]
[54]
Dias HK, Brown CL, Polidori MC, Lip GY, Griffiths HR. LDL-lipids from patients with hypercholesterolaemia and Alzheimer’s disease are inflammatory to microvascular endothelial cells: mitigation by statin intervention. Clin Sci (Lond) 2015; 129(12): 1195-206.
[http://dx.doi.org/10.1042/CS20150351] [PMID: 26399707]
[55]
Hendrie HC, Hake A, Lane K, et al. Statin use, incident dementia and alzheimer disease in elderly african americans. Ethn Dis 2015; 25(3): 345-54.
[http://dx.doi.org/10.18865/ed. 25.3.345] [PMID: 26673814]
[56]
Liang T, Li R, Cheng O. Statins for treating alzheimer’s disease: Truly ineffective? Eur Neurol 2015; 73(5-6): 360-6.
[http://dx.doi.org/10.1159/000382128] [PMID: 26021802]
[57]
Solomon A, Kivipelto M, Wolozin B, Zhou J, Whitmer RA. Midlife serum cholesterol and increased risk of Alzheimer’s and vascular dementia three decades later. Dement Geriatr Cogn Disord 2009; 28(1): 75-80.
[http://dx.doi.org/10.1159/000231980] [PMID: 19648749]
[58]
Mielke MM, Zandi PP, Shao H, et al. The 32-year relationship between cholesterol and dementia from midlife to late life. Neurology 2010; 75(21): 1888-95.
[http://dx.doi.org/ 10.1212/WNL.0b013e3181feb2bf] [PMID: 21068429]
[59]
Strittmatter W. J. Medicine. Old drug, new hope for Alzheimer’s disease. Science 2012; 335(6075): 1447-8.
[http://dx.doi.org/ 10.1126/science.1220725] [PMID: 22442467]
[60]
Yin J, Turner GH, Coons SW, Maalouf M, Reiman EM, Shi J. Association of amyloid burden, brain atrophy and memory deficits in aged apolipoprotein ε4 mice. Curr Alzheimer Res 2014; 11(3): 283-90.
[http://dx.doi.org/10.2174/156720501103140329220007] [PMID: 24694076]
[61]
Jiang Q, Lee CY, Mandrekar S, et al. ApoE promotes the proteolytic degradation of Abeta. Neuron 2008; 58(5): 681-93.
[http://dx.doi.org/10.1016/j.neuron.2008.04.010] [PMID: 18549781]
[62]
Koldamova R, Fitz NF, Lefterov I. The role of ATP-binding cassette transporter A1 in Alzheimer’s disease and neurodegeneration. Biochim Biophys Acta 2010; 1801(8): 824-30.
[http://dx.doi.org/dx. doi.org/10.1016/j.bbalip.2010.02.010] [PMID: 20188211]
[63]
Bonet-Costa V, Herranz-Pérez V, Blanco-Gandía M, et al. Clearing amyloid-β through PPARγ/ApoE activation by genistein is a treatment of experimental alzheimer’s disease. J Alzheimers Dis 2016; 51(3): 701-11.
[http://dx.doi.org/10.3233/JAD-151020] [PMID: 26890773]
[64]
Ogata M, Tsujita M, Hossain MA, et al. On the mechanism for PPAR agonists to enhance ABCA1 gene expression. Atherosclerosis 2009; 205(2): 413-9.
[http://dx.doi.org/ 10.1016/j.atherosclerosis.2009.01.008] [PMID: 19201410]
[65]
Mandrekar-Colucci S, Landreth GE. Nuclear receptors as therapeutic targets for Alzheimer’s disease. Expert Opin Ther Targets 2011; 15(9): 1085-97.
[http://dx.doi.org/10.1517/14728222.2011. 594043] [PMID: 21718217]
[66]
Odegaard JI, Chawla A. Alternative macrophage activation and metabolism. Annu Rev Pathol 2011; 6: 275-97.
[http://dx.doi.org/dx.doi. org/10.1146/annurev-pathol-011110-130138] [PMID: 21034223]
[67]
Talbot K, Wang HY, Kazi H, 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 Invest 2012; 122(4): 1316-38.
[http://dx.doi.org/10.1172/JCI59903] [PMID: 22476197]
[68]
Barbagallo M, Dominguez LJ. Type 2 diabetes mellitus and Alzheimer’s disease. World J Diabetes 2014; 5(6): 889-93.
[http://dx.doi.org/10.4239/wjd.v5.i6.889] [PMID: 25512792]
[69]
Fehm HL, Kern W, Peters A. The selfish brain: competition for energy resources. 2006.
[70]
Abramov AY, Duchen MR. Impaired mitochondrial bioenergetics determines glutamate-induced delayed calcium deregulation in neurons. Biochim Biophys Acta 2010; 1800(3): 297-304.
[http://dx.doi.org/ dx.doi.org/10.1016/j.bbagen.2009.08.002] [PMID: 19695307]
[71]
Agrawal R, Gomez-Pinilla F. ‘Metabolic syndrome’ in the brain: deficiency in omega-3 fatty acid exacerbates dysfunctions in insulin receptor signalling and cognition. J Physiol 2012; 590(10): 2485-99.
[http://dx.doi.org/10.1113/jphysiol.2012.230078] [PMID: 22473784]
[72]
Liu J, Wang LN, Jia JP. Peroxisome proliferator-activated receptor-gamma agonists for Alzheimer’s disease and amnestic mild cognitive impairment: a systematic review and meta-analysis. Drugs Aging 2015; 32(1): 57-65.
[http://dx.doi.org/10.1007/s40266-014-0228-7] [PMID: 25504005]
[73]
Sato T, Hanyu H, Hirao K, Kanetaka H, Sakurai H, Iwamoto T. Efficacy of PPAR-γ agonist pioglitazone in mild Alzheimer disease. Neurobiol Aging 2011; 32(9): 1626-33.
[http://dx.doi.org/dx.doi. org/10.1016/j.neurobiolaging.2009.10.009] [PMID: 19923038]
[74]
Denner LA, Rodriguez-Rivera J, Haidacher SJ, et al. Cognitive enhancement with rosiglitazone links the hippocampal PPARγ and ERK MAPK signaling pathways. J Neurosci 2012; 32(47): 16725-35a.
[http://dx.doi.org/10.1523/JNEUROSCI.2153-12.2012] [PMID: 23175826]
[75]
Jahrling JB, Hernandez CM, Denner L, Dineley KT. PPARγ recruitment to active ERK during memory consolidation is required for Alzheimer’s disease-related cognitive enhancement. J Neurosci 2014; 34(11): 4054-63.
[http://dx.doi.org/10.1523/JNEUROSCI. 4024-13.2014] [PMID: 24623782]
[76]
Searcy JL, Phelps JT, Pancani T, et al. Long-term pioglitazone treatment improves learning and attenuates pathological markers in a mouse model of Alzheimer’s disease. J Alzheimers Dis 2012; 30(4): 943-61.
[http://dx.doi.org/10.3233/JAD-2012-111661] [PMID: 22495349]
[77]
Hort J, Laczó J, Vyhnálek M, Bojar M, Bures J, Vlcek K. Spatial navigation deficit in amnestic mild cognitive impairment. Proc Natl Acad Sci USA 2007; 104(10): 4042-7.
[http://dx.doi.org/ dx.doi.org/10.1073/pnas.0611314104] [PMID: 17360474]
[78]
Hoefer M, Allison SC, Schauer GF, et al. Fear conditioning in frontotemporal lobar degeneration and Alzheimer’s disease. Brain 2008; 131(Pt 6): 1646-57.
[http://dx.doi.org/10. 1093/brain/awn082] [PMID: 18492729]
[79]
Barage SH, Sonawane KD. Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer’s disease. Neuropeptides 2015; 52: 1-18.
[http://dx.doi.org/10.1016/j.npep. 2015.06.008] [PMID: 26149638]
[80]
Prakash A, Kumar A. Role of nuclear receptor on regulation of BDNF and neuroinflammation in hippocampus of β-amyloid animal model of Alzheimer’s disease. Neurotox Res 2014; 25(4): 335-47.
[http://dx.doi.org/10.1007/s12640-013-9437-9] [PMID: 24277156]
[81]
Xiang GQ, Tang SS, Jiang LY, et al. PPARγ agonist pioglitazone improves scopolamine-induced memory impairment in mice. J Pharm Pharmacol 2012; 64(4): 589-96.
[http://dx.doi.org/10. 1111/j.2042-7158.2011.01432.x] [PMID: 22420664]
[82]
Beyer AM, Baumbach GL, Halabi CM, et al. Interference with PPARgamma signaling causes cerebral vascular dysfunction, hypertrophy, and remodeling. Hypertension 2008; 51(4): 867-71.
[http://dx.doi.org/10.1161/HYPERTENSIONAHA. 107.103648] [PMID: 18285614]
[83]
Ketsawatsomkron P, Pelham CJ, Groh S, Keen HL, Faraci FM, Sigmund CD. Does peroxisome proliferator-activated receptor-γ (PPAR γ) protect from hypertension directly through effects in the vasculature? J Biol Chem 2010; 285(13): 9311-6.
[http://dx.doi.org/10.1074/jbc.R109.025031] [PMID: 20129921]
[84]
Di BB, Li HW, Li WP, Shen XH, Sun ZJ, Wu X. Pioglitazone inhibits high glucose-induced expression of receptor for advanced glycation end products in coronary artery smooth muscle cells. Mol Med Rep 2015; 11(4): 2601-7.
[http://dx.doi.org/ 10.3892/mmr.2014.3113] [PMID: 25523934]
[85]
Mukohda M, Stump M, Ketsawatsomkron P, Hu C, Quelle FW, Sigmund CD. Endothelial PPAR-γ provides vascular protection from IL-1β-induced oxidative stress. Am J Physiol Heart Circ Physiol 2016; 310(1): H39-48.
[http://dx.doi.org/10.1152/ajpheart.00490.2015] [PMID: 26566726]
[86]
Sakatani Y, Miyoshi T, Oe H, et al. Pioglitazone prevents the endothelial dysfunction induced by ischemia and reperfusion in healthy subjects. J Cardiovasc Pharmacol 2014; 64(4): 326-31.
[http://dx.doi.org/10.1097/FJC.0000000000000124] [PMID: 24887686]
[87]
Fuenzalida K, Quintanilla R, Ramos P, et al. Peroxisome proliferator-activated receptor gamma up-regulates the Bcl-2 anti-apoptotic protein in neurons and induces mitochondrial stabilization and protection against oxidative stress and apoptosis. J Biol Chem 2007; 282(51): 37006-15.
[http://dx.doi.org/10.1074/jbc.M700447200] [PMID: 17965419]
[88]
Haorah J, Ramirez SH, Schall K, Smith D, Pandya R, Persidsky Y. Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to blood-brain barrier dysfunction. J Neurochem 2007; 101(2): 566-76.
[http://dx.doi.org/ 10.1111/j.1471-4159.2006.04393.x] [PMID: 17250680]
[89]
Fujimoto M, Takagi Y, Aoki T, et al. Tissue inhibitor of metalloproteinases protect blood-brain barrier disruption in focal cerebral ischemia. J Cereb Blood Flow Metab 2008; 28(10): 1674-85.
[http://dx.doi.org/10.1038/jcbfm.2008.59] [PMID: 18560439]
[90]
Lee SR, Kim HY, Hong JS, Baek WK, Park JW. PPARgamma agonist pioglitazone reduces matrix metalloproteinase-9 activity and neuronal damage after focal cerebral ischemia. Biochem Biophys Res Commun 2009; 380(1): 17-21.
[http://dx.doi.org/10. 1016/j.bbrc.2008.12.181] [PMID: 19135426]
[91]
Huang W, Eum SY, András IE, Hennig B, Toborek M. PPARalpha and PPARgamma attenuate HIV-induced dysregulation of tight junction proteins by modulations of matrix metalloproteinase and proteasome activities. FASEB J 2009; 23(5): 1596-606.
[http://dx.doi.org/10.1096/fj.08-121624] [PMID: 19141539]
[92]
Sanz MJ, Albertos F, Otero E, Juez M, Morcillo EJ, Piqueras L. Retinoid X receptor agonists impair arterial mononuclear cell recruitment through peroxisome proliferator-activated receptor-γ activation. J Immunol 2012; 189(1): 411-24.
[http://dx.doi.org/dx. doi.org/10.4049/jimmunol.1102942] [PMID: 22661092]
[93]
Wang L, Waltenberger B, Pferschy-Wenzig EM, et al. Natural product agonists of peroxisome proliferator-activated receptor gamma (PPARγ): a review. Biochem Pharmacol 2014; 92(1): 73-89.
[http://dx.doi.org/10. 1016/j.bcp.2014.07.018] [PMID: 25083916]
[94]
Wang P, Li B, Cai G, et al. Activation of PPAR-γ by pioglitazone attenuates oxidative stress in aging rat cerebral arteries through upregulating UCP2. J Cardiovasc Pharmacol 2014; 64(6): 497-506.
[http://dx.doi.org/10.1097/FJC.0000000000000143] [PMID: 25490415]
[95]
Mosconi L. Glucose metabolism in normal aging and Alzheimer’s disease: Methodological and physiological considerations for PET studies. Clin Transl Imaging 2013; 1(4): 1.
[http://dx.doi.org/ 10.1007/s40336-013-0026-y] [PMID: 24409422]
[96]
van der Ossenkoppele R, Flier WM, Verfaillie SC, et al. Long-term effects of amyloid, hypometabolism, and atrophy on neuropsychological functions. Neurol 2014; 82: 1768-75.
[http://dx.doi.org/10.1212/WNL.0000000000000432]
[97]
De Nuccio C, Bernardo A, Cruciani C, De Simone R, Visentin S, Minghetti L. Peroxisome proliferator activated receptor-γ agonists protect oligodendrocyte progenitors against tumor necrosis factor-alpha-induced damage: Effects on mitochondrial functions and differentiation. Exp Neurol 2015; 271: 506-14.
[http://dx.doi.org/dx. doi.org/10.1016/j.expneurol.2015.07.014] [PMID: 26210873]
[98]
Roses AD, Saunders AM, Huang Y, Strum J, Weisgraber KH, Mahley RW. Complex disease-associated pharmacogenetics: drug efficacy, drug safety, and confirmation of a pathogenetic hypothesis (Alzheimer’s disease). Pharmacogenomics J 2007; 7(1): 10-28.
[http://dx.doi.org/10.1038/sj.tpj.6500397] [PMID: 16770341]
[99]
Corona JC, de Souza SC, Duchen MR. PPARγ activation rescues mitochondrial function from inhibition of complex I and loss of PINK1. Exp Neurol 2014; 253: 16-27.
[http://dx.doi.org/ 10.1016/j.expneurol.2013.12.012] [PMID: 24374061]
[100]
Chiang MC, Cheng YC, Lin KH, Yen CH. PPARγ regulates the mitochondrial dysfunction in human neural stem cells with tumor necrosis factor alpha. Neuroscience 2013; 229: 118-29.
[http://dx.doi.org/ dx.doi.org/10.1016/j.neuroscience.2012.11.003] [PMID: 23153990]
[101]
Chiang MC, Cheng YC, Chen HM, Liang YJ, Yen CH. Rosiglitazone promotes neurite outgrowth and mitochondrial function in N2A cells via PPARgamma pathway. Mitochondrion 2014; 14(1): 7-17.
[http://dx.doi.org/10.1016/j.mito.2013.12.003] [PMID: 24370585]
[102]
Quintanilla RA, Orellana DI, González-Billault C, Maccioni RB. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp Cell Res 2004; 295(1): 245-57.
[http://dx.doi.org/10.1016/j.yexcr.2004.01. 002] [PMID: 15051507]
[103]
Chen J, Li S, Sun W, Li J. Anti-diabetes drug pioglitazone ameliorates synaptic defects in AD transgenic mice by inhibiting cyclin-dependent kinase5 activity. PLoS One 2015; 10(4): e0123864.
[http://dx.doi.org/10.1371/journal.pone.0123864] [PMID: 25875370]
[104]
Nenov MN, Laezza F, Haidacher SJ, et al. Cognitive enhancing treatment with a PPARγ agonist normalizes dentate granule cell presynaptic function in Tg2576 APP mice. J Neurosci 2014; 34(3): 1028-36.
[http://dx.doi.org/ 10.1523/JNEUROSCI.3413-13.2014] [PMID: 24431460]
[105]
Son SM, Shin HJ, Byun J, et al. Metformin facilitates amyloid-β generation by β- and γ-secretases via autophagy activation. J Alzheimers Dis 2016; 51(4): 1197-208.
[http://dx.doi.org/10.3233/JAD-151200] [PMID: 26967226]
[106]
Wang X, Zheng W, Xie JW, et al. Insulin deficiency exacerbates cerebral amyloidosis and behavioral deficits in an Alzheimer transgenic mouse model. Mol Neurodegener 2010; 5: 46-58.
[http://dx.doi.org/ 10.1186/1750-1326-5-46] [PMID: 21044348]
[107]
Benedict C, Frey WH II, Schiöth HB, Schultes B, Born J, Hallschmid M. Intranasal insulin as a therapeutic option in the treatment of cognitive impairments. Exp Gerontol 2011; 46(2-3): 112-5.
[http://dx.doi.org/10.1016/j.exger.2010.08.026] [PMID: 20849944]
[108]
McNay EC, Ong CT, McCrimmon RJ, Cresswell J, Bogan JS, Sherwin RS. Hippocampal memory processes are modulated by insulin and high-fat-induced insulin resistance. Neurobiol Learn Mem 2010; 93(4): 546-53.
[http://dx.doi.org/10.1016/j.nlm.2010.02.002] [PMID: 20176121]
[109]
Watson GS, Cholerton BA, Reger MA, et al. Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. Am J Geriatr Psychiatry 2005; 13(11): 950-8.
[PMID: 16286438]
[110]
Risner ME, Saunders AM, Altman JF, et al. Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer’s disease. Pharmacogenomics J 2006; 6(4): 246-54.
[http://dx.doi.org/10.1038/sj.tpj.6500369] [PMID: 16446752]
[111]
Craft S, Asthana S, Schellenberg G, et al. Insulin metabolism in Alzheimer’s disease differs according to apolipoprotein E genotype and gender. Neuroendocrinology 1999; 70(2): 146-52.
[http://dx.doi.org/ dx.doi.org/10.1159/000054469] [PMID: 10461029]
[112]
Feinstein DL, Galea E, Gavrilyuk V, et al. Peroxisome proliferator-activated receptor-gamma agonists prevent experimental autoimmune encephalomyelitis. Ann Neurol 2002; 51(6): 694-702.
[http://dx.doi.org/ 10.1002/ana.10206] [PMID: 12112074]
[113]
Doost Mohammadpour J, Hosseinmardi N, Janahmadi M, Fathollahi Y, Motamedi F, Rohampour K. Non-selective NSAIDs improve the amyloid-β-mediated suppression of memory and synaptic plasticity. Pharmacol Biochem Behav 2015; 132: 33-41.
[http://dx.doi.org/10.1016/j.pbb.2015.02.012] [PMID: 25697476]
[114]
Yu Y, Li X, Blanchard J, et al. Insulin sensitizers improve learning and attenuate tau hyperphosphorylation and neuroinflammation in 3xTg-AD mice. J Neural Transm (Vienna) 2015; 122(4): 593-606.
[http://dx.doi.org/10. 1007/s00702-014-1294-z] [PMID: 25113171]
[115]
Gupta R, Gupta LK. Improvement in long term and visuo-spatial memory following chronic pioglitazone in mouse model of Alzheimer’s disease. Pharmacol Biochem Behav 2012; 102(2): 184-90.
[http://dx.doi.org/10.1016/j.pbb.2012.03.028] [PMID: 22503969]
[116]
Heneka MT, Fink A, Doblhammer G. Effect of pioglitazone medication on the incidence of dementia. Ann Neurol 2015; 78(2): 284-94.
[http://dx.doi.org/10.1002/ana.24439] [PMID: 25974006]
[117]
Papadopoulos P, Rosa-Neto P, Rochford J, Hamel E. Pioglitazone improves reversal learning and exerts mixed cerebrovascular effects in a mouse model of Alzheimer’s disease with combined amyloid-β and cerebrovascular pathology. PLoS One 2013; 8(7): e68612.
[http://dx.doi.org/10.1371/journal.pone.0068612] [PMID: 23874687]
[118]
Flesch D, Ness J, Lamers C, et al. SAR-studies of γ-secretase modulators with PPARγ-agonistic and 5-lipoxygenase-inhibitory activity for Alzheimer’s disease. Bioorg Med Chem Lett 2015; 25(4): 841-6.
[http://dx.doi.org/10.1016/j.bmcl.2014.12.073] [PMID: 25575659]
[119]
de la Monte SM, Tong M, Schiano I, Didsbury J. Improved brain insulin/IGF signaling and reduced neuroinflammation with T3D-959 in an experimental model of sporadic alzheimer’s disease. J Alzheimers Dis 2017; 55(2): 849-64.
[http://dx.doi.org/10. 3233/JAD-160656] [PMID: 27802237]
[120]
Tong M, Dominguez C, Didsbury J, de la Monte SM. Targeting alzheimer’s disease Neuro-Metabolic Dysfunction with a small molecule nuclear receptor agonist (T3D-959) reverses disease pathologies. J Alzheimers Dis Parkinsonism 2016; 6(3): 238.
[121]
Almad A, Lash AT, Wei P, Lovett-Racke AE, McTigue DM. The PPAR alpha agonist gemfibrozil is an ineffective treatment for spinal cord injured mice. Exp Neurol 2011; 232(2): 309-17.
[http://dx.doi.org/10.1016/j.expneurol.2011.09.023] [PMID: 21963672]
[122]
Geldmacher DS, Fritsch T, McClendon MJ, Landreth G. A randomized pilot clinical trial of the safety of pioglitazone in treatment of patients with Alzheimer disease. Arch Neurol 2011; 68(1): 45-50.
[http://dx.doi.org/10.1001/archneurol.2010.229] [PMID: 20837824]
[123]
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): 131-46.
[http://dx.doi.org/10.1159/000318845] [PMID: 20733306]
[124]
Harrington C, Sawchak S, Chiang C, et al. Rosiglitazone does not improve cognition or global function when used as adjunctive therapy to AChE inhibitors in mild-to-moderate Alzheimer’s disease: two phase 3 studies. Curr Alzheimer Res 2011; 8(5): 592-606.
[http://dx.doi.org/10. 2174/156720511796391935] [PMID: 21592048]
[125]
Van Hildreth KL, Pelt RE, Moreau KL, et al. Effects of pioglitazone or exercise in older adults with mild cognitive impairment and insulin resistance: a pilot study. Dement Geriatr Cogn Disord Extra 2015; 5: 51-63.
[http://dx.doi.org/ dx.doi.org/10.1159/000371509]
[126]
Jiang LY, Tang SS, Wang XY, et al. PPARγ agonist pioglitazone reverses memory impairment and biochemical changes in a mouse model of type 2 diabetes mellitus. CNS Neurosci Ther 2012; 18(8): 659-66.
[http://dx.doi.org/10.1111/j.1755-5949.2012.00341.x] [PMID: 22620268]
[127]
Cheng H, Shang Y, Jiang L, Shi TL, Wang L. The peroxisome proliferators activated receptor-gamma agonists as therapeutics for the treatment of Alzheimer’s disease and mild-to-moderate Alzheimer’s disease: a meta-analysis. Int J Neurosci 2016; 126(4): 299-307.
[http://dx.doi.org/10.3109/00207454.2015.1015722] [PMID: 26001206]
[128]
Nicolakakis N, Aboulkassim T, Ongali B, et al. Complete rescue of cerebrovascular function in aged Alzheimer’s disease transgenic mice by antioxidants and pioglitazone, a peroxisome proliferator-activated receptor gamma agonist. J Neurosci 2008; 28(37): 9287-96.
[http://dx.doi.org/10.1523/JNEUROSCI.3348-08.2008] [PMID: 18784309]
[129]
Inestrosa NC, Toledo EM. The role of Wnt signaling in neuronal dysfunction in Alzheimer’s Disease. Mol Neurodegener 2008; 3: 9.
[http://dx.doi.org/10.1186/1750-1326-3-9] [PMID: 18652670]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy