Journal List > Anat Cell Biol > v.49(1) > 1071842

This article has been corrected. See "Erratum: The association between PGC-1α and Alzheimer's disease" in Volume 49 on page 163.
Sweeney and Song: The association between PGC-1α and Alzheimer's disease

Abstract

Alzheimer's disease (AD) is a neurodegenerative disorder and its reported pathophysiological features in the brain include the deposition of amyloid beta peptide, chronic inflammation, and cognitive impairment. The incidence of AD is increasing worldwide and researchers have studied various aspects of AD pathophysiology in order to improve our understanding of the disease. Thus far, the onset mechanisms and means of preventing AD are completely unknown. Peroxisome proliferator-activated receptor-γ coactivator (PGC-1α) is a protein related to various cellular mechanisms that lead to the alteration of downstream gene regulation. It has been reported that PGC-1α could protect cells against oxidative stress and reduce mitochondrial dysfunction. Moreover, it has been demonstrated to have a regulatory role in inflammatory signaling and insulin sensitivity related to cognitive function. Here, we present further evidence of the involvement of PGC-1α in AD pathogenesis. Clarifying the relationship between PGC-1α and AD pathology might highlight PGC-1α as a possible target for therapeutic intervention in AD.

Introduction

The incidence of Alzheimer's disease (AD) is expected to increase dramatically as the world population [123]. The peroxisome proliferator-activated receptor (PPAR)-γ coactivator-1 (PGC-1) family of coactivators comprises proteins that mediate responses to environmental stress [45]. Several studies have reported that the level of PGC-1α evidently decreases in the brains of AD patients [67]. In the AD brain, oxidative stress has been regarded as the core problem, leading to other AD pathologies [89]. PGC-1α controls the expression of genes related to the generation of reactive oxygen species (ROS) and prevents oxidative stress by reducing the production of ROS [10]. In AD, impaired mitochondrial biogenesis in neuronal cells causes synapse dysfunction [11] and cellular damage [12] and contributes to cognitive decline [1314]. Additionally, insulin resistance of the AD brain aggravates the rapid progress of AD pathophysiology [1516]. Much research has demonstrated that PGC-1α improves mitochondrial function [17] and insulin sensitivity [1819]. In this review, we summarize recent research on the association between PGC-1α and AD and provide new insights about PGC-1α's role in the mechanism of AD pathology.

PGC-1α

PGC-1α is highly responsive to numerous forms of environmental stress, including temperature and nutritional status [45]. PGC-1α also regulates mitochondrial biogenesis in response to diverse environmental stimuli [20]. PGC-1α forms heteromeric complexes with a variety of transcription factors including nuclear respiratory factor (NRF)-1, NRF-2, PPARα, PPARδ, PPARγ, and estrogen-related receptor α [21]. These PGC-1α transcriptional activator complexes could displace repressor proteins, such as histone deacetylase and its small heterodimer partner and thereby induce gene activation [22]. PGC-1α is commonly expressed in tissues with a high energy demand, including brown adipose tissue, skeletal muscle, and the brain [2324]. In the brain, impairment of the activity of PGC-1α triggers the degeneration of neurons by inducing mitochondrial dysfunction [1725]. PGC-1α-knockout mice display behavioral abnormalities such as frequent limb clasping [26]. Moreover, interaction between PGC-1α and signal pathways, such as that involving Cre-binding protein [27], cGMP-dependent pathways [28], and p38–mitogen activated protein kinase pathways [29] plays a critical role in the response to oxidative stress in AD [30]. PGC-1α plays a central role by influencing the genes that regulate detoxification of ROS [31]. According to clinical research, PGC-1α may be key to maintaining brain function in AD, since its levels decreased in AD patients in comparison to that in normal subjects [67]. Based on this evidence, focusing on PGC-1α's role may lead to a better understanding of the mechanisms of AD pathology.

PGC-1α and Oxidative Stress in AD

Salient features of AD include molecular aberrations such as oxidative stress [8], inflammation [32]. Of these features, oxidative stress appears to be the trigger of free radical-induced cellular damage, DNA oxidation, and aberration in DNA repair [33]. In AD, the primary brain areas where neuronal damage due to oxidative stress occurs are the hippocampus and the cortex [34]. Compared to control cases, AD patients exhibit some aspects of elevated oxidative stress including the production of proteins such as cytochrome c oxidase [34], increased lipid peroxidation [35]. Therefore, the markers of oxidative damage founded in neurons in AD are the hallmark of its pathologies and an indication of degeneration in the AD brain [36]. In AD, increased levels of ROS, including hydrogen peroxide and hydroxyl radicals, impede various cellular functions by degrading proteins [37]. PGC-1α plays a central role in the regulation of ROS detoxifying enzymes, such as superoxide dismutase 1 and 2, catalase and glutathione peroxidase-1 [17]. It has been reported that PGC-1α modulates the expression of uncoupling protein 2 [38] and uncoupling protein 3, which are both direct regulators of ROS formation [39]. Additionally, PGC-1α controls the level of sirtuin1 [40] and sirtuin3 [41] which reduce the generation of ROS [10]. Some research demonstrates that elevated PGC-1α levels protect neural cells from apoptosis due to oxidative stress through the induction of antioxidant genes [17]. One study showed that increased PGC-1α activity could ameliorate neuronal loss and improve neurological symptoms [42]. Taken together, the elevated activity of PGC-1α could protect neuronal cells from damage by reducing the oxidative stress in AD and subsequently alleviate several pathophysiological features of this disorder.

PGC-1α, Mitochondrial Dysfunction, and Cognitive Dysfunction in AD

In AD, neurodegeneration and synaptic degradation are caused by impaired mitochondrial biogenesis [12]. Mitochondria play a crucial role in the process of neuronal apoptosis in the AD brain [43] and are the pivotal organelle for the generation of ROS [44]. Mitochondrial dysfunction has been considered as one of the central cytopathologies of AD [45] and is known to contribute to cognitive decline through various pathways. In AD neurons, mitochondria are sites of amyloid beta accumulation, and these amyloid beta accumulations in mitochondria finally result in the death of the cell [46]. Impaired mitochondrial function leads to a severe loss in energy metabolism and ATP generation [47], and also to a deficiency in the scavenging of free radicals which triggers excessive oxidative damage in the AD brain [4849]. An association between mitochondrial dysfunction and memory dysfunction has been demonstrated in several human and animal studies [5051]. In AD, mitochondrial dysfunction, including an increase in oxidative stress [52] and defective mitochondrial biogenesis [53] occurs in neurodegeneration [54]. In the aged brain, PGC-1α regulates the expression of sirtuin 3, which is a factor related to the aging process [53]. It has been observed that in the brains of patients with neurodegenerative diseases, low levels of PGC-1α lead to mitochondrial dysfunction and oxidative stress [5556]. PGC-1α regulates mitochondrial density in neurons [57] and PGC-1α–knockout mice showed an increased sensitivity to the degeneration of dopaminergic and glutamatergic neurons in the brain [17]. Moreover, another study demonstrated that the reduction of mitochondrial gene expression in PGC-1α–knockout mice finally leads to neuronal dysfunction [26]. PGC-1α stimulates expression of GA-binding protein α, a known regulator of cognitive function, in a cell culture study [58]. Given that PGC-1α plays a crucial role in neuronal function [59] and regulates mitochondrial function, PGC-1α could ameliorate mitochondrial dysfunction and improve cognitive function in AD.

PGC-1α, Insulin Resistance, and Cognitive Dysfunction in AD

Insulin modulates neurotransmitter release [60], neuronal cell survival [61], and synaptic plasticity [62] and it improves cognition and memory function in the brain[6364]. Insulin resistance in the brain is defined as decreased uptake of insulin into the brain, leading to the dysregulation of amyloid beta level and inflammation [65]. In AD, insulin resistance in the brain is an important issue since it contributes to the progress of the disease [66]. Recent research demonstrated that patients with AD have defective insulin signaling [67] in the brain, as well as reduced insulin receptor sensitivity [68]. The PGC-1a gene is expressed at high levels in obese animals [69] and diabetic mice [70] compared to that in normal animals. PGC-1α is a transcriptional coactivator involved in the mitochondrial biogenic response that counteracts insulin resistance [71]. In a study conducted with PGC-1α–knockout mice, the mice exhibited insulin sensitivity by comparison with normal controls in spite of a high fat diet [26]. Moreover, PGC-1α improves glucose tolerance, insulin sensitivity and gluconeogenesis [18]. Considering that PGC-1α alleviates insulin resistance [72], it could reduce cognitive impairment related to insulin resistance in the AD brain.

Conclusion

In this review, we summarized recent evidence indicating that PGC-1α can contribute to the improvement of AD pathophysiology. Here, we highlight four points: (1) PGC-1α could protect against oxidative stress in AD and thereby prevent neuronal cell damage, (2) PGC-1α could improve mitochondrial dysfunction in AD, (3) PGC-1α could reduce insulin resistance in AD, and (4) finally, PGC-1α could ameliorate cognitive impairment caused by AD. Thus, this review raises the possibility that PGC-1α could be used as a therapeutic agent in the treatment of AD.

References

1. Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement. 2013; 9:63–75.e2.
2. Mattson MP. Energy intake and exercise as determinants of brain health and vulnerability to injury and disease. Cell Metab. 2012; 16:706–722.
3. De Felice FG. Alzheimer's disease and insulin resistance: translating basic science into clinical applications. J Clin Invest. 2013; 123:531–539.
4. Puigserver P, Wu Z, Park CW, Graves R, Wright M, Spiegelman BM. A cold-inducible coactivator of nuclear receptors linked to adaptive thermogenesis. Cell. 1998; 92:829–839.
5. Scarpulla RC. Transcriptional activators and coactivators in the nuclear control of mitochondrial function in mammalian cells. Gene. 2002; 286:81–89.
6. Qin W, Haroutunian V, Katsel P, Cardozo CP, Ho L, Buxbaum JD, Pasinetti GM. PGC-1alpha expression decreases in the Alzheimer disease brain as a function of dementia. Arch Neurol. 2009; 66:352–361.
7. Zheng B, Liao Z, Locascio JJ, Lesniak KA, Roderick SS, Watt ML, Eklund AC, Zhang-James Y, Kim PD, Hauser MA, Grunblatt E, Moran LB, Mandel SA, Riederer P, Miller RM, Federoff HJ, Wüllner U, Papapetropoulos S, Youdim MB, Cantuti-Castelvetri I, Young AB, Vance JM, Davis RL, Hedreen JC, Adler CH, Beach TG, Graeber MB, Middleton FA, Rochet JC, Scherzer CR. Global PD Gene Expression (GPEX) Consortium. PGC-1alpha, a potential therapeutic target for early intervention in Parkinson's disease. Sci Transl Med. 2010; 2:52ra73.
8. Yan MH, Wang X, Zhu X. Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic Biol Med. 2013; 62:90–101.
9. Luca M, Luca A, Calandra C. The role of oxidative damage in the pathogenesis and progression of Alzheimer's disease and vascular dementia. Oxid Med Cell Longev. 2015; 2015:504678.
10. Sahin E, Colla S, Liesa M, Moslehi J, Müller FL, Guo M, Cooper M, Kotton D, Fabian AJ, Walkey C, Maser RS, Tonon G, Foerster F, Xiong R, Wang YA, Shukla SA, Jaskelioff M, Martin ES, Heffernan TP, Protopopov A, Ivanova E, Mahoney JE, Kost-Alimova M, Perry SR, Bronson R, Liao R, Mulligan R, Shirihai OS, Chin L, DePinho RA. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature. 2011; 470:359–365.
11. Mendoza E, Miranda-Barrientos JA, Vázquez-Roque RA, Morales-Herrera E, Ruelas A, De la, Flores G, Hernández-Echeagaray E. In vivo mitochondrial inhibition alters corticostriatal synaptic function and the modulatory effects of neurotrophins. Neuroscience. 2014; 280:156–170.
12. Reddy PH, Tripathi R, Troung Q, Tirumala K, Reddy TP, Anekonda V, Shirendeb UP, Calkins MJ, Reddy AP, Mao P, Manczak M. Abnormal mitochondrial dynamics and synaptic degeneration as early events in Alzheimer's disease: implications to mitochondria-targeted antioxidant therapeutics. Biochim Biophys Acta. 2012; 1822:639–649.
13. Kimata T, Sasakura H, Ohnishi N, Nishio N, Mori I. Thermotaxis of C. elegans as a model for temperature perception, neural information processing and neural plasticity. Worm. 2012; 1:31–41.
14. Stein GM, Murphy CT. The intersection of aging, longevity pathways, and learning and memory in C. elegans. Front Genet. 2012; 3:259.
15. Willette AA, Bendlin BB, Starks EJ, Birdsill AC, Johnson SC, Christian BT, Okonkwo OC, La Rue A, Hermann BP, Koscik RL, Jonaitis EM, Sager MA, Asthana S. Association of insulin resistance with cerebral glucose uptake in late middle-aged adults at risk for Alzheimer disease. JAMA Neurol. 2015; 72:1013–1020.
16. Cai Z, Xiao M, Chang L, Yan LJ. Role of insulin resistance in Alzheimer's disease. Metab Brain Dis. 2015; 30:839–851.
17. St-Pierre J, Drori S, Uldry M, Silvaggi JM, Rhee J, Jäger S, Handschin C, Zheng K, Lin J, Yang W, Simon DK, Bachoo R, Spiegelman BM. Suppression of reactive oxygen species and neurodegeneration by the PGC-1 transcriptional coactivators. Cell. 2006; 127:397–408.
18. Baur JA, Pearson KJ, Price NL, Jamieson HA, Lerin C, Kalra A, Prabhu VV, Allard JS, Lopez-Lluch G, Lewis K, Pistell PJ, Poosala S, Becker KG, Boss O, Gwinn D, Wang M, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG, Le Couteur D, Shaw RJ, Navas P, Puigserver P, Ingram DK, de Cabo R, Sinclair DA. Resveratrol improves health and survival of mice on a high-calorie diet. Nature. 2006; 444:337–342.
19. Lagouge M, Argmann C, Gerhart-Hines Z, Meziane H, Lerin C, Daussin F, Messadeq N, Milne J, Lambert P, Elliott P, Geny B, Laakso M, Puigserver P, Auwerx J. Resveratrol improves mitochondrial function and protects against metabolic disease by activating SIRT1 and PGC-1alpha. Cell. 2006; 127:1109–1122.
20. Lin J, Wu H, Tarr PT, Zhang CY, Wu Z, Boss O, Michael LF, Puigserver P, Isotani E, Olson EN, Lowell BB, Bassel-Duby R, Spiegelman BM. Transcriptional co-activator PGC-1 alpha drives the formation of slow-twitch muscle fibres. Nature. 2002; 418:797–801.
21. Lin J, Handschin C, Spiegelman BM. Metabolic control through the PGC-1 family of transcription coactivators. Cell Metab. 2005; 1:361–370.
22. Lin J, Tarr PT, Yang R, Rhee J, Puigserver P, Newgard CB, Spiegelman BM. PGC-1beta in the regulation of hepatic glucose and energy metabolism. J Biol Chem. 2003; 278:30843–30848.
23. Lai L, Wang M, Martin OJ, Leone TC, Vega RB, Han X, Kelly DP. A role for peroxisome proliferator-activated receptor gamma coactivator 1 (PGC-1) in the regulation of cardiac mitochondrial phospholipid biosynthesis. J Biol Chem. 2014; 289:2250–2259.
24. Pinho RA, Pinho CA, Tromm CB, Pozzi BG, Souza DR, Silva LA, Tuon T, Souza CT. Changes in the cardiac oxidative metabolism induced by PGC-1{alpha}: response of different physical training protocols in infarction-induced rats. Int J Cardiol. 2013; 168:4560–4562.
25. Weydt P, Pineda VV, Torrence AE, Libby RT, Satterfield TF, Lazarowski ER, Gilbert ML, Morton GJ, Bammler TK, Strand AD, Cui L, Beyer RP, Easley CN, Smith AC, Krainc D, Luquet S, Sweet IR, Schwartz MW, La Spada AR. Thermoregulatory and metabolic defects in Huntington's disease transgenic mice implicate PGC-1alpha in Huntington's disease neurodegeneration. Cell Metab. 2006; 4:349–362.
26. Lin J, Wu PH, Tarr PT, Lindenberg KS, St-Pierre J, Zhang CY, Mootha VK, Jäger S, Vianna CR, Reznick RM, Cui L, Manieri M, Donovan MX, Wu Z, Cooper MP, Fan MC, Rohas LM, Zavacki AM, Cinti S, Shulman GI, Lowell BB, Krainc D, Spiegelman BM. Defects in adaptive energy metabolism with CNS-linked hyperactivity in PGC-1alpha null mice. Cell. 2004; 119:121–135.
27. Herzig S, Long F, Jhala US, Hedrick S, Quinn R, Bauer A, Rudolph D, Schutz G, Yoon C, Puigserver P, Spiegelman B, Montminy M. CREB regulates hepatic gluconeogenesis through the coactivator PGC-1. Nature. 2001; 413:179–183.
28. Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C, Sciorati C, Bracale R, Valerio A, Francolini M, Moncada S, Carruba MO. Mitochondrial biogenesis in mammals: the role of endogenous nitric oxide. Science. 2003; 299:896–899.
29. Puigserver P, Rhee J, Lin J, Wu Z, Yoon JC, Zhang CY, Krauss S, Mootha VK, Lowell BB, Spiegelman BM. Cytokine stimulation of energy expenditure through p38 MAP kinase activation of PPARgamma coactivator-1. Mol Cell. 2001; 8:971–982.
30. Procaccio V, Bris C, Chao de la Barca JM, Oca F, Chevrollier A, Amati-Bonneau P, Bonneau D, Reynier P. Perspectives of drug-based neuroprotection targeting mitochondria. Rev Neurol (Paris). 2014; 170:390–400.
31. Lu Z, Xu X, Hu X, Fassett J, Zhu G, Tao Y, Li J, Huang Y, Zhang P, Zhao B, Chen Y. PGC-1 alpha regulates expression of myocardial mitochondrial antioxidants and myocardial oxidative stress after chronic systolic overload. Antioxid Redox Signal. 2010; 13:1011–1022.
32. Krstic D, Knuesel I. Deciphering the mechanism underlying late-onset Alzheimer disease. Nat Rev Neurol. 2013; 9:25–34.
33. Nunomura A, Moreira PI, Castellani RJ, Lee HG, Zhu X, Smith MA, Perry G. Oxidative damage to RNA in aging and neurodegenerative disorders. Neurotox Res. 2012; 22:231–248.
34. Sultana R, Butterfield DA. Oxidative modification of brain proteins in Alzheimer's disease: perspective on future studies based on results of redox proteomics studies. J Alzheimers Dis. 2013; 33:Suppl 1. S243–S251.
35. Bradley-Whitman MA, Lovell MA. Biomarkers of lipid peroxidation in Alzheimer disease (AD): an update. Arch Toxicol. 2015; 89:1035–1044.
36. Nunomura A, Tamaoki T, Motohashi N, Nakamura M, McKeel DW Jr, Tabaton M, Lee HG, Smith MA, Perry G, Zhu X. The earliest stage of cognitive impairment in transition from normal aging to Alzheimer disease is marked by prominent RNA oxidation in vulnerable neurons. J Neuropathol Exp Neurol. 2012; 71:233–241.
37. Jahani-Asl A, Cheung EC, Neuspiel M, MacLaurin JG, Fortin A, Park DS, McBride HM, Slack RS. Mitofusin 2 protects cerebellar granule neurons against injury-induced cell death. J Biol Chem. 2007; 282:23788–23798.
38. Wu Z, Puigserver P, Andersson U, Zhang C, Adelmant G, Mootha V, Troy A, Cinti S, Lowell B, Scarpulla RC, Spiegelman BM. Mechanisms controlling mitochondrial biogenesis and respiration through the thermogenic coactivator PGC-1. Cell. 1999; 98:115–124.
39. Brand MD. Uncoupling to survive? The role of mitochondrial inefficiency in ageing. Exp Gerontol. 2000; 35:811–820.
40. Rodgers JT, Lerin C, Gerhart-Hines Z, Puigserver P. Metabolic adaptations through the PGC-1 alpha and SIRT1 pathways. FEBS Lett. 2008; 582:46–53.
41. Ozden O, Park SH, Kim HS, Jiang H, Coleman MC, Spitz DR, Gius D. Acetylation of MnSOD directs enzymatic activity responding to cellular nutrient status or oxidative stress. Aging (Albany NY). 2011; 3:102–107.
42. Tsunemi T, Ashe TD, Morrison BE, Soriano KR, Au J, Roque RA, Lazarowski ER, Damian VA, Masliah E, La Spada AR. PGC-1alpha rescues Huntington's disease proteotoxicity by preventing oxidative stress and promoting TFEB function. Sci Transl Med. 2012; 4:142ra97.
43. Obulesu M, Lakshmi MJ. Apoptosis in Alzheimer's disease: an understanding of the physiology, pathology and therapeutic avenues. Neurochem Res. 2014; 39:2301–2312.
44. Bonda DJ, Lee HP, Lee HG, Friedlich AL, Perry G, Zhu X, Smith MA. Novel therapeutics for Alzheimer's disease: an update. Curr Opin Drug Discov Devel. 2010; 13:235–246.
45. Maruszak A, Żekanowski C. Mitochondrial dysfunction and Alzheimer's disease. Prog Neuropsychopharmacol Biol Psychiatry. 2011; 35:320–330.
46. Picone P, Nuzzo D, Caruana L, Scafidi V, Di Carlo M. Mitochondrial dysfunction: different routes to Alzheimer's disease therapy. Oxid Med Cell Longev. 2014; 2014:780179.
47. Atamna H, Kumar R. Protective role of methylene blue in Alzheimer's disease via mitochondria and cytochrome c oxidase. J Alzheimers Dis. 2010; 20:S439–S452.
48. Zubenko GS, Sauer P. SOD-1 activity and platelet membrane fluidity in Alzheimer's disease. Biol Psychiatry. 1989; 25:671–678.
49. Moreira PI, Santos MS, Sena C, Nunes E, Seiça R, Oliveira CR. CoQ10 therapy attenuates amyloid beta-peptide toxicity in brain mitochondria isolated from aged diabetic rats. Exp Neurol. 2005; 196:112–119.
50. Bishop NA, Lu T, Yankner BA. Neural mechanisms of ageing and cognitive decline. Nature. 2010; 464:529–535.
51. Aksenov V, Long J, Liu J, Szechtman H, Khanna P, Matravadia S, Rollo CD. A complex dietary supplement augments spatial learning, brain mass, and mitochondrial electron transport chain activity in aging mice. Age (Dordr). 2013; 35:23–33.
52. Choksi KB, Nuss JE, DeFord JH, Papaconstantinou J. Mitochondrial electron transport chain functions in long-lived Ames dwarf mice. Aging (Albany NY). 2011; 3:754–767.
53. Hirayama M, Nakamura T, Watanabe H, Uchida K, Hama T, Hara T, Niimi Y, Ito M, Ohno K, Sobue G. Urinary 8-hydroxydeoxyguanosine correlate with hallucinations rather than motor symptoms in Parkinson's disease. Parkinsonism Relat Disord. 2011; 17:46–49.
54. Shacka JJ, Roth KA, Zhang J. The autophagy-lysosomal degradation pathway: role in neurodegenerative disease and therapy. Front Biosci. 2008; 13:718–736.
55. Handschin C, Kobayashi YM, Chin S, Seale P, Campbell KP, Spiegelman BM. PGC-1alpha regulates the neuromuscular junction program and ameliorates Duchenne muscular dystrophy. Genes Dev. 2007; 21:770–783.
56. Cui L, Jeong H, Borovecki F, Parkhurst CN, Tanese N, Krainc D. Transcriptional repression of PGC-1alpha by mutant huntingtin leads to mitochondrial dysfunction and neurodegeneration. Cell. 2006; 127:59–69.
57. Wareski P, Vaarmann A, Choubey V, Safiulina D, Liiv J, Kuum M, Kaasik A. PGC-1{alpha} and PGC-1{beta} regulate mitochondrial density in neurons. J Biol Chem. 2009; 284:21379–21385.
58. Mootha VK, Handschin C, Arlow D, Xie X, St Pierre J, Sihag S, Yang W, Altshuler D, Puigserver P, Patterson N, Willy PJ, Schulman IG, Heyman RA, Lander ES, Spiegelman BM. Erralpha and Gabpa/b specify PGC-1alpha-dependent oxidative phosphorylation gene expression that is altered in diabetic muscle. Proc Natl Acad Sci U S A. 2004; 101:6570–6575.
59. Ma D, Li S, Lucas EK, Cowell RM, Lin JD. Neuronal inactivation of peroxisome proliferator-activated receptor gamma coactivator 1alpha (PGC-1alpha) protects mice from diet-induced obesity and leads to degenerative lesions. J Biol Chem. 2010; 285:39087–39095.
60. Pandey G, Makhija E, George N, Chakravarti B, Godbole MM, Ecelbarger CM, Tiwari S. Insulin regulates nitric oxide production in the kidney collecting duct cells. J Biol Chem. 2015; 290:5582–5591.
61. Yu LY, Pei Y. Insulin neuroprotection and the mechanism. Chin Med J (Engl). 2015; 128:976–981.
62. Banks WA. The source of cerebral insulin. Eur J Pharmacol. 2004; 490:5–12.
63. Kern W, Born J, Schreiber H, Fehm HL. Central nervous system effects of intranasally administered insulin during euglycemia in men. Diabetes. 1999; 48:557–563.
64. Schäfer M, Erdö SL. Development of glutamate neurotoxicity in cortical cultures: induction of vulnerability by insulin. Brain Res Dev Brain Res. 1991; 62:293–296.
65. Baker LD, Cross DJ, Minoshima S, Belongia D, Watson GS, Craft S. Insulin resistance and Alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with prediabetes or early type 2 diabetes. Arch Neurol. 2011; 68:51–57.
66. Nuzzo D, Picone P, Baldassano S, Caruana L, Messina E, Marino Gammazza A, Cappello F, Mulè F, Carlo MD. Insulin resistance as common molecular denominator linking obesity to Alzheimer's disease. Curr Alzheimer Res. 2015; 12:723–735.
67. Lourenco MV, Clarke JR, Frozza RL, Bomfim TR, Forny-Germano L, Batista AF, Sathler LB, Brito-Moreira J, Amaral OB, Silva CA, Freitas-Correa L, Espirito-Santo S, Campello-Costa P, Houzel JC, Klein WL, Holscher C, Carvalheira JB, Silva AM, Velloso LA, Munoz DP, Ferreira ST, De Felice FG. 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; 18:831–843.
68. Talbot K, Wang HY, Kazi H, Han LY, Bakshi KP, Stucky A, Fuino RL, Kawaguchi KR, Samoyedny AJ, Wilson RS, Arvanitakis Z, Schneider JA, Wolf BA, Bennett DA, Trojanowski JQ, Arnold SE. 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; 122:1316–1338.
69. Duncan JG, Fong JL, Medeiros DM, Finck BN, Kelly DP. Insulin-resistant heart exhibits a mitochondrial biogenic response driven by the peroxisome proliferator-activated receptor-alpha/PGC-1alpha gene regulatory pathway. Circulation. 2007; 115:909–917.
70. Biddinger SB, Hernandez-Ono A, Rask-Madsen C, Haas JT, Alemán JO, Suzuki R, Scapa EF, Agarwal C, Carey MC, Stephanopoulos G, Cohen DE, King GL, Ginsberg HN, Kahn CR. Hepatic insulin resistance is sufficient to produce dyslipidemia and susceptibility to atherosclerosis. Cell Metab. 2008; 7:125–134.
71. Mitra R, Nogee DP, Zechner JF, Yea K, Gierasch CM, Kovacs A, Medeiros DM, Kelly DP, Duncan JG. The transcriptional coactivators, PGC-1alpha and beta, cooperate to maintain cardiac mitochondrial function during the early stages of insulin resistance. J Mol Cell Cardiol. 2012; 52:701–710.
72. Ha CD, Cho JK, Han T, Lee SH, Kang HS. Relationship of PGC-1alpha gene polymorphism with insulin resistance syndrome in Korean children. Asia Pac J Public Health. 2015; 27:NP544–NP551.
TOOLS
Similar articles