Yangtze Medicine
Vol.02 No.01(2018), Article ID:81036,17 pages
10.4236/ym.2018.21001

Therapeutic Potential of FGF21 in Alzheimer’s Disease

Anbang Sun1,2*, Benke Xu1, Xianwang Wang3, Lian Liu4, Yun He1, Zongwen Wang1, Yuncai Chen1,5*

1Department of Anatomy, Medical School of Yangtze University, Jingzhou, China

2The First Affiliated Hospital & The First School of Clinical Medicine, Yangtze University, Jingzhou, China

3Department of Medical Laboratory Science, Medical School of Yangtze University, Jingzhou, China

4Department of Medical Function, School of Medicine, Yangtze University, Jingzhou, China

5Department of Pediatrics, University of California, Irvine, CA, USA

Copyright © 2018 by authors and Scientific Research Publishing Inc.

This work is licensed under the Creative Commons Attribution International License (CC BY 4.0).

http://creativecommons.org/licenses/by/4.0/

Received: September 19, 2017; Accepted: December 10, 2017; Published: December 13, 2017

ABSTRACT

Alzheimer’s disease (AD) is the most common form of dementia which mostly affects persons younger than 65 years old. Mounting findings showed that amyloid-β (Aβ) peptides, oxidative stress, neuroinflammation and insulin resistance may play central role in the pathogenesis of AD. There are very many methods to slow it through affecting these aforementioned factors. However, more efficient prevention of the progression of AD is still ambiguous. Fibroblast growth factor 21 (FGF21) is an endocrine hormone that is expressed by several organs. It increases insulin sensitivity and regulates lipid metabolism and energy homeostasis. Emerging evidence demonstrates that FGF21 has potential effects in the brain involving metabolic regulation, neuroprotection and cognition. Hence, we hypothesize that FGF21 may be a protective factor in AD by attenuating Aβ generation, inflammation, oxidative stress, and insulin resistance. Our hypothesis will shed new light on the understanding of pathogenesis of AD and help to find a new way to prevent the genesis and progress of AD.

Keywords:

FGF21, Alzheimer’ Disease, Amyloid-β (Aβ), Oxidative Stress, Inflammation, Insulin Resistance

1. Introduction

Alzheimer’s disease (AD) is an insidious, progressive and fatal neurodegenerative disorder manifested by cognitive and memory deterioration, progressive impairment of daily life activities, and as well as a variety of neuropsychiatric symptoms and behavioral disturbances [1] . It is the most common form of dementia, with an estimated 5.2 million people diagnosed in the United States in 2013, of which approximately 200,000 individuals are younger than 65 [1] and 50% of people over 85 years old are affected by various degrees of AD [2] . There are more than 47 million people living with dementia worldwide in 2016, and the total estimated worldwide cost of dementia is more than 818 billion USD. It will become a trillion dollar disease by 2018 [3] . By 2050, the distribution of dementia cases is 67.2 million in Asia (51.5% of total), 15.8 million in Africa (12%), 29.9 in Americas (22.7%) and 18.6 in Europe (14.2%), (Table 1). Nearly 68% of all people living with dementia will live in low and middle income countries [4] . The etiology and pathogenesis of AD is complicated, but currently known pathological hallmarks of AD include the presence of neuritic plaques, neurofibrillary tangles, synaptic loss, ultimately neuronal death [5] [6] [7] [8] [9] . Methods to block the progression of the disease and prevent neuronal loss or even cure it are still ambiguous.

2. Pathogenesis of Alzheimer’s Disease

2.1. Amyloid-β (Aβ) Peptides

Mounting evidence showed that amyloid-β (Aβ) peptides play a central role in the pathogenesis of AD. The initial “amyloid cascade” hypothesis suggested that Aβ peptides drive the neuropathological cascade responses leading to dementia [10] . The revised amyloid cascade hypothesis suggested that soluble Aβ oligomers initiate the pathological cascade of AD leading to synaptic dysfunction, neuronal cell death, and dementia [11] . Moreover, transgenic mice with overexpression of the human amyloid precursor protein (APP-tg mice) have learning and memory deficits, as well as neuritic plaques similar to those seen in humans with AD [12] . In view of this, several immunization programs have successfully cleared the amyloid in APP-tg mice [13] [14] . However, the immunization programs attempted to clear AD patients brain amyloid through monoclonal antibodies, AN1792 [15] , Solanezumab (LY2062430) and Bapineuzumab [16] [17] , were terminated because of adverse effects 14 and lack of clinical treatment effect [16] [17] . The results of concurrent neuropsychological tests and magnetic resonance imaging (MRI) tests were surprising and disappointing, the brain

Table 1. People living with dementia around the world (million).

Figure 1. The potential mechanisms of Aβ in AD and the potential targets of FGF21. 1) Aβ derives from the amyloid precursor protein (APP), which is cleaved by β secretase (BACE1) and γ secretase to yield Aβ. The Aβ40 form is the more common isoform, but Aβ42 is the more fibrillogenic and is thus associated with disease states. It is generally believed that Aβ oligomers are the most toxic. It leads to synaptic dysfunction, neuroinflammation, neurofibrillary tangles, then causes neuronal dysfunction and death, ultimately result in learning and memory deficits which displayed by AD. 2) Most of autosomal dominant familial AD can be attributed to mutations in one of three genes: those encoding APP and presenilins 1 and 2 (PS1/2), while the apolipoprotein E4 (APOE4) is the major genetic risk factor for sporadic AD. People with trisomy 21 (Down Syndrome) almost universally exhibit at least the earliest symptoms of AD by 40 years of age. All these factors can lead to Aβ buildup, Aβ aggregation, and the sequential changes and symptoms we mentioned above. 3) There are some immunization programs attempted to clear AD patients brain amyloid through monoclonal antibodies, AN1792, Solanezumab (LY2062430) and Bapineuzumab, but they were terminated because of adverse effects and lack of clinical treatment effect. 4) FGF21 can activate peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), then decrease Aβ generation by reducing BACE1 transcription.

atrophy in immunized patients was worse than that of placebo-controlled patients [18] . These findings suggested that amyloid deposition is not the only one pathogenic cause of AD (Figure 1).

2.2. Oxidative Stress

Other research indicated that oxidative stress may be involved in the pathogenesis of AD. Further study showed that oxidative stress markedly elevated in preclinical AD patients and amnestic mild cognitive impairment (aMCI) patients [19] [20] . The mechanism may be that the oxidative stress leads to membrane damage, cytoskeleton alterations [21] and destroy the synaptic homeostasis in the hippocampus early in the AD process and ultimately speeding up the progression of AD [19] . Growing evidence indicates that many oxidative markers are increased in the AD brain, including protein oxidation [22] , lipid peroxidation [23] , and nucleic acid oxidation [21] . Reactive oxygen species (ROS), which are cytotoxic byproducts of oxygen metabolism, are accumulated during aging, hyperglycemia, hypoxic insults and inflammation [24] . In the AD brain, these may induce synaptic loss and eventually promote neurofibrillary tangles and neuritic plaques formation [25] . Moreover, there is a positive feedback loop between oxidative stress and Aβ, which contributes to the pathologic progress of AD [26] [27] [28] . This is because ROS alter the expression of β-APP cleaving enzyme (BACE1), the rate-limiting enzyme of AD [29] [30] , and Presenilins1 (PS1), the catalytic submit of the γ-secretase [26] , thereby promoting Aβ production in AD. Meanwhile, Aβ induces oxidative stress and the generation of HNE, which is a neurotoxic lipid peroxidation product and associated with Aβ pathology in AD [26] . Several studies suggested that antioxidants vitamin E and vitamin C play a role in delaying the onset of AD [31] [32] [33] [34] . These studies further confirmed that oxidative stress is involved in the occurrence and progression of AD (Figure 2).

Figure 2. The potential mechanisms of oxidative stress in AD and the potential targets of FGF21. 1) Oxidative stress leads to membrane damage, cytoskeleton alterations and destroy the synaptic homeostasis in the hippocampus early in the AD process and ultimately speeding up the progression of AD. During these process, there are many oxidative markers are increased in the AD brain, including protein oxidation, lipid peroxidation, and nucleic acid oxidation. 2) Oxidative stress can also increase the production of ROS which alter the expression of BACE1 and PS1, then promote the production of Aβ in AD. 3) There is a positive feedback loop between oxidative stress and Aβ. All the factors mentioned above will finally lead to AD which illustrated in Figure 1. 4) FGF21 can upregulate the expression of genes encoding proteins involved in antioxidative pathways, including mitochondrial uncoupling proteins (Ucp2 and Ucp3), superoxide dismutase-2 (Sod2), reduced ROS production. It also could inhibit D-galactose-induced ROS production in a dose dependent manner, through preventing NF-κB nuclear translation and IκBα degradation.

2.3. Neuroinflammation

Besides the amyloid plaques and oxidative stress, neuroinflammation in the central nervous system (CNS) also causes neuronal injury, and pro-inflammatory changes exist in early stages of the disease in AD patients, when microglial cells are activated and produce pro-inflammatory mediators [35] . Acute systemic inflammatory events with increased levels of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), were associated with an increasing rate of cognitive decline in mild to severe dementia [36] . Animal models of lipopolysaccharide (LPS) induced sepsis showed CNS inflammation, neuronal death and cognitive decline [37] [38] [39] . Correspondingly, non-steroidal anti-inflammatory drugs (NSAIDs) can ameliorate behavioral and pathology deficits in AD transgenic mouse models [40] [41] . Although clinical trials have failed to reproduce the beneficial effects of NSAIDs in AD patients, they may be beneficial when administered in the early stage of the disease [42] . The anti-inflammatory effects of NSAIDs may be that NSAIDs directly bind to peroxisome proliferator-activated receptor γ (PPAR γ) [42] , which expressed in human brain [43] and inhibited microglial activation and the expression of a wide range of proinflammatory genes [42] [44] [45] , and activate its transcriptional regulatory activities [42] . Other anti-inflammatory drugs such as trifusal showed a significant low rate of conversion to dementia in clinical trials with mild cognitive impairment patients [37] [46] . The diversity of effects may depend on the drug, because different anti- inflammatory drugs may have different molecular targets. And anti-TNFα treatment with the antibody against TNFα, infliximab, reduced Aβ and tau phosphorylation in transgenic mice [41] . In addition, the TNFα inhibitor thalidomide can decrease the activation of both astrocytes and microglia, Aβ load, plaque formation and tau phosphorylation [41] (Figure 3). Further studies assessing the potential for targeting these specific inflammatory processes are needed to elucidate more effective treatments and provide a clearer understanding of the complexities of inflammatory signaling in AD [41] .

2.4. Insulin Resistance

Accumulating evidence also suggests that insulin resistance acts as a known risk factor for AD [2] [47] . Brain insulin signaling plays a critical role in the regulation of food intake, body weight, reproduction, and learning and memory [48] [49] . Defective insulin signaling is associated with decreased cognitive ability and the development of dementia and AD [2] [50] . Insulin resistance, induced by peripheral metabolic syndrome, impairs the insulin signaling in the brain, which mainly impacts the PIK3/Akt pathway, then reduces Aβ and tau phosphorylation by inhibiting the activation of glycogen synthase kinase 3-α and -β separately, which are the key signaling molecules downstream of Akt [51] . There is a feed-forward interaction between impaired insulin signaling and Aβ production, which contributes to the pathologic progress of the AD and cognitive decline [2] [52] . Insulin resistance also can promote Aβ generation through altering

Figure 3. The potential mechanisms of neuroinflammation in AD and the potential targets of FGF21. 1) Neuroinflammation and acute systemic inflammatory events in the CNS causes neuronal injury, then activate the microglia cells and produce pro-inflammatory factors (TNF-α, IL-6, MCP1) exist in early stages of the disease in AD patients. The pro-inflammatory factors increase Aβ formation and tau phosphorylation, then lead to dementia with Aβ plaques and tau tangles. 2) The anti-inflammatory drugs (NSAIDs, trifusal), directly bind to PPARγ, then inhibit microglial activation and the expression of a wide range of proinflammatory genes. Infliximab, the antibody against TNFα, can reduce Aβ and tau phosphorylation. Thalidomide, the TNFα inhibitor, can decrease the activation of both astrocytes and microglia, Aβ load, plaque formation and tau phosphorylation. All the factors mentioned above can improve AD through different pathways. 3) FGF21 could significantly reduce the mRNA expression level of TNFα. It also can inhibit macrophage-mediated inflammation, by activating the nuclear transcription factor-E2-related factor 2 (Nrf2) and suppressing the NF-κB signaling pathway.

insulin signal transduction, increasing BACE1 and γ-secretase activities, and accumulation of autophagosomes [52] [53] . Moreover, insulin resistance causes both cerebral glucose hypometabolism and a systemic hyperinsulinemic state [49] . Brain glucose hypometabolism was found even at the preclinical stage of AD [49] . Peripheral insulin resistance not only reduced cerebral glucose metabolism, but also decreased Aβ clearance in the CNS [47] [48] . Rosiglitazone, a ligand for peroxisome proliferator-activated receptors (PPARs), improve cognitive function by facilitating Aβ clearance, reducing amyloid plaques and tau phosphorylation in AD mouse models [2] [54] [55] . Rosiglitazone can also protect cognitive decline in MCI patients [56] [57] [58] , while pioglitazone can improve cognitive deficiency and stabilize the disease in the individuals with mild AD [59] [60] . Treatments to enhance cerebral glucose metabolism showed improvement in cognition and AD symptomatology [61] . Collectively, insulin resistance is closely related with the pathologic process of AD (Figure 4).

Figure 4. The potential mechanisms of insulin resistance in AD and the potential targets of FGF21. 1) Insulin resistance impairs the insulin signaling in the brain, which mainly impacts the PIK3/Akt pathway, then reduces Aβ and tau phosphorylation by inhibiting the activation of glycogen synthase kinase 3-α and -β separately. It also can promote Aβ generation through altering insulin signal transduction, increasing BACE1 and γ-secretase activities, and accumulation of autophagosomes. 2) There is a positive feedback loop between insulin resistance and Aβ. All the factors mentioned above will finally cause AD. 3) Rosiglitazone, a ligand for peroxisome proliferator-activated receptors (PPARs), improve cognitive function by facilitating Aβ clearance, reducing amyloid plaques and tau phosphorylation in AD. 4) FGF21 can potentially ameliorate cognition through improving insulin resistance and cerebral glucose hypometabolism.

In summary, Aβ peptides play a central role in the pathogenesis of AD, while oxidative stress, neuroinflammation and insulin resistance participate in the pathogenic process of AD.

3. Fibroblast Growth Factor 21(FGF21) in Alzheimer’s Disease

FGF21 is one member of the FGF family. It acts as either a paracrine or an endocrine hormone, and is expressed by adipose tissue, muscles, liver, pancreas, heart, and brain [62] [63] . FGF21 can lower glucose and lipid levels, increase insulin sensitivity and regulate energy homeostasis in rodents [64] . Its activity occurs when FGF21 binds to the fibroblast growth factor receptor (FGFR) and β-klotho (KLB), a single-pass transmembrane protein [64] and an essential co-receptor for FGF21 [65] [66] . There are seven major isoforms of FGFR, including 1b, 1c, 2b, 2c, 3b, 3c and 4. In vivo study, FGFR1c is the primary receptor to mediate its activity [67] [68] [69] . In fact, FGF21 is expressed in several areas of the brain, including the substantia nigra, striatum, hippocampus and cortex [70] . It can enter the brain from blood and can also be detected in human cerebrospinal fluid [71] [72] [73] . Moreover, FGFs and KLB have been found in several brain areas [74] [75] [76] . These findings indicate that FGF21 may have potential regulating roles in the CNS.

3.1. FGF21 and Amyloid-β (Aβ) Peptides

Growing evidence demonstrates that FGF21 activated peroxisome proliferator- activated receptor γ coactivator-1α (PGC-1α) [70] [77] [78] [79] [80] , which is abundantly expressed in the brain [81] [82] . The expression of PGC-1α is reduced in the brain of AD patients. Exogenous human PGC-1α (hPGC-1α) expressed in primary neurons from the Tg2576 mouse of AD decreased Aβ generation by reducing BACE1 transcription which was dependent on PPAR γ [83] [84] . In accordance with this result, gene delivery of hPGC-1α in the brain of transgenic APP23 mice reduced amyloid deposition, which correlated with a decrease in BACE1 expression [85] . These findings suggest that FGF21 may reduce Aβ generation by decreasing BACE1 expression, as illustrated in Figure 1.

3.2. FGF21 and Oxidative Stress

As widely known, ROS, the cytotoxic byproducts of oxygen metabolism, could induce synaptic loss, promote neurofibrillary tangles, form neuritic plaques and finally cause neuron death [24] . This plays an essential role in the pathologic progress of AD. Recent work has shown that FGF21 participates in regulating oxidative stress [86] [87] [88] . It upregulated the expression of genes encoding proteins involved in antioxidative pathways, including mitochondrial uncoupling proteins (Ucp2 and Ucp3), superoxide dismutase-2 (Sod2), reduced ROS production in cardiomyocytes, and ameliorated cardiac tissue injury [86] [87] . In the brain of aging mice, FGF21could inhibit D-galactose-induced ROS production in a dose dependent manner [87] , through preventing NF-κB nuclear translation and IκBα degradation [88] . Hence, as showed in Figure 2, FGF21 may ameliorate the oxidative stress of AD by enhancing the activities of SOD and reducing the production of ROS.

3.3. FGF21 and Inflammation

Increasing evidences demonstrated that inflammatory process play a critical role in AD progression [89] [90] [91] . However, FGF21 was demonstrated a modulatory role in the inflammatory processes [61] [73] . As demonstrated in Figure 3, it acted as a positive acute phase response (APR) protein and protected mice from the challenge of LPS and sepsis [61] . The inflammatory genes such as interleukin-6 (IL-6), and monocyte chemoattractant protein-1 (MCP-1) were significantly higher in FGF21 knockout mice [86] . While in db/db mice models, FGF21 treatment significantly reduced the mRNA expression level of TNFα [92] . FGF21 inhibited macrophage-mediated inflammation, by activating the nuclear transcription factor-E2-related factor 2 (Nrf2) and suppressing the NF-κB signaling pathway [93] .

3.4. FGF21 and Insulin Resistance

A growing body of findings suggests that insulin resistance is closely related with the pathologic process of AD [2] [50] [51] . FGF21 acts as one of the metabolic regulators. It has been demonstrated as a potent regulator of glycemia, lipid metabolism and energy homeostasis [94] [95] . Recombinant FGF21 treatment can improve whole-body insulin sensitivity and reduce plasma levels of glucose and triglycerides in diabetic mice [94] , while loss of endogenous FGF21 in vivo led to increased insulin resistance and pancreatic islet hyperplasia and dysfunction [95] . As presented in Figure 4, FGF21 can potentially ameliorate cognition through improving insulin resistance and cerebral glucose hypometabolism.

4. Conclusions

Here, we hypothesize that the FGF21 may be a protective factor in AD by attenuating Aβ generation, inflammation, oxidative stress, and insulin resistance.

FGF21 increased energy expenditure and insulin sensitivity in obese rats, and intracerebroventricular injection of FGF21 into rats increased metabolic rate and insulin sensitivity [96] . FGF21 also acts as a robust neuroprotective factor and a potentially new therapeutic target for CNS disorders. For example, exogenous FGF21 protein completely protected aging neurons from glutamate challenge [97] . The serumal FGF21 levels increased significantly in patients with schizophrenia [96] . It may be involved in regulating glucose metabolism in schizophrenia, with positive correlations with pyruvate, lactate, 2-oxoglutarate, and malate in the schizophrenia group [96] . On the other hand, FGF21 activated PGC-1α and increased mitochondrial efficacy in human dopaminergic neurons which suggest that FGF21 could potentially play a role in dopaminergic neuron viability and in Parkinson’s disease [70] .

Moreover, increasing evidence indicates that FGF21 has beneficial roles on behavior and cognition. The activity of transgenic overexpression of FGF21 mice (TgFgf21) increased during light phase, but decreased during dark phase. This circadian behavior in mice can also be altered by genetically deleting KLB in the brain [75] . The locomotor activity of FGF21 transgenic mice was reduced after a 24 hours fast [98] . D-galactose-induced aging mice, which were administrated with FGF21, had preserved cognitive function. This may be related to FGF21’s ability to reduce brain cell damage in hippocampus by attenuating oxidative stress, increasing anti-oxidant activity, decreasing the enhanced total cholinesterase activity in the brain and reducing the expression of pro-inflammation cytokines such as IL-6 and TNF-α [63] [88] .

Taken together, FGF21 acts as a neuroprotective factor, performs its decreased Aβ generation, anti-inflammatory, anti-oxidative stress, and glucose homeostatic effects. In view of the complex pathogenesis of AD, we propose that FGF21 may be a protective factor in AD by attenuating Aβ generation, inflammation, oxidative stress, and insulin resistance. It may be a potential therapeutic for AD.

Acknowledgements

The study was supported by the Yangtze Youth Fund (Grant NO.2015cqn79), the Chutian Scholars Program (2012-12) and the Yangtze Fund for Youth Teams of Science and Technology Innovation (Grant NO.2016cqt04).

Cite this paper

Sun, A.B., Xu, B.K., Wang, X.W., Liu, L., He, Y., Wang, Z.W. and Chen, Y.C. (2018) Therapeutic Potential of FGF21 in Alzheimer’s Disease. Yangtze Medicine, 1, 1-17. https://doi.org/10.4236/ym.2018.21001

References

  1. 1. Weuve, J., Hebert, L.E., Scherr, P.A. and Evans, D.A. (2014) Deaths in the United States among Persons with Alzheimer’s Disease (2010–2050). Alzheimers & Dementia: The Journal of the Alzheimers Association, 10, e40-e46. https://doi.org/10.1016/j.jalz.2014.01.004

  2. 2. Kim, B. and Feldman, E.L. (2015) Insulin Resistance as a Key Link for the Increased Risk of Cognitive Impairment in the Metabolic Syndrome. Experimental & Molecular Medicine, 47, e149. https://doi.org/10.1038/emm.2015.3

  3. 3. Herrera, A.C., Prince, M., Knapp, M., Karagiannidou, M. and Guerchet, M. (2016) World Alzheimer Report 2016. The Global Impact of Dementia. An Analysis of Prevalence, Incidence, Cost and Trends.

  4. 4. Prince, M., Wimo, A., Guerchet, M., Ali, G.C., Wu, Y.T. and Prina, M. (2015) World Alzheimer Report 2015. The Global Impact of Dementia. An Analysis of Prevalence, Incidence, Cost and Trends.

  5. 5. Mckeith, I.G., Galasko, D., Kosaka, K., et al. (1996) Consensus Guidelines for Clinical and Pathological Diagnosis of Dementia with Lewy Bodies (DLB): Report of the CDLB International Workshop. Neurology, 47, 1113-1124.https://doi.org/10.1212/WNL.47.5.1113

  6. 6. Vickers, J.C., Dickson, T.C., Adlard, P.A., Saunders, H.L., King, C.E. and Mccormack, G. (2000) The Cause of Neuronal Degeneration in Alzheimer’s Disease. Progress in Neurobiology, 60, 139-165. https://doi.org/10.1016/S0301-0082(99)00023-4

  7. 7. Jack, C.R.Jr., Dickson, D.W., Parisi, J.E., et al. (2002) Antemortem MRI Findings Correlate with Hippocampal Neuropathology in Typical Aging and Dementia. Neurology, 58, 750-757. https://doi.org/10.1212/WNL.58.5.750

  8. 8. Bennett, D.A., Wilson, R.S., Schneider, J.A., et al. (2003) Apolipoprotein E epsilon4 allele, AD Pathology, and the Clinical Expression of Alzheimer’s Disease. Neurology, 60, 246-252. https://doi.org/10.1212/01.WNL.0000042478.08543.F7

  9. 9. Hoozemans, J.J., Veerhuis, R., Van Haastert, E.S., et al. (2005) The Unfolded Protein Response Is Activated in Alzheimer’s Disease. Acta Neuropathologica, 110, 165-172. https://doi.org/10.1007/s00401-005-1038-0

  10. 10. Hardy, J. (2002) Testing Times for the “Amyloid Cascade Hypothesis”. Neurobiology Aging, 23, 1073-1074. https://doi.org/10.1016/S0197-4580(02)00042-8

  11. 11. Mor, F. and Monsonego, A. (2006) Immunization Therapy in Alzheimer’s Disease. Expert Review of Neurotherapeutics, 6, 653-659. https://doi.org/10.1586/14737175.6.5.653

  12. 12. Mufson, E.J., Ikonomovic, M.D., Counts, S.E., et al. (2016) Molecular and Cellular Pathophysiology of Preclinical Alzheimer’s Disease. Behavioural Brain Research, 311, 54-69. https://doi.org/10.1016/j.bbr.2016.05.030

  13. 13. Qu, B., Boyer, P.J., Johnston, S.A., Hynan, L.S. and Rosenberg, R.N. (2006) Abeta42 Gene Vaccination Reduces Brain Amyloid Plaque Burden in Transgenic Mice. The Journal of the Neurological Sciences, 244, 151-158. https://doi.org/10.1016/j.jns.2006.02.006

  14. 14. Gilman, S., Koller, M., Black, R.S., et al. (2005) Clinical Effects of Abeta Immunization (AN1792) in Patients with AD in an Interrupted Trial. Neurology, 64, 1553-1562. https://doi.org/10.1212/01.WNL.0000159740.16984.3C

  15. 15. Fox, N.C., Black, R.S., Gilman, S., et al. (2005) Effects of Abeta Immunization (AN1792) on MRI Measures of Cerebral Volume in Alzheimer Disease. Neurology, 64, 1563-1572. https://doi.org/10.1212/01.WNL.0000159743.08996.99

  16. 16. Salloway, S., Sperling, R., Fox, N.C., et al. (2014) Two Phase 3 Trials of Bapineuzumab in Mild-to-Moderate Alzheimer’s Disease. The New England Journal of Medicine, 370, 322-333. https://doi.org/10.1056/NEJMoa1304839

  17. 17. Doody, R.S., Thomas, R.G., Farlow, M., et al. (2014) Phase 3 Trials of Solanezumab for Mild-to-Moderate Alzheimer’s Disease. The New England Journal of Medicine, 370, 311-321. https://doi.org/10.1056/NEJMoa1312889

  18. 18. Cummings, J.L. (2004) Alzheimer’s Disease. The New England Journal of Medicine, 351, 56-67. https://doi.org/10.1056/NEJMra040223

  19. 19. Esposito, E., Rotilio, D., Di Matteo, V., Di Giulio, C., Cacchio, M. and Algeri, S. (2002) A Review of Specific Dietary Antioxidants and the Effects on Biochemical Mechanisms Related to Neurodegenerative Processes. Neurobiology of Aging, 23, 719-735. https://doi.org/10.1016/S0197-4580(02)00078-7

  20. 20. Lovell, M.A. and Markesbery, W.R. (2001) Ratio of 8-Hydroxyguanine in Intact DNA to Free 8-Hydroxyguanine Is Increased in Alzheimer Disease Ventricular Cerebrospinal Fluid. Archives of Neurology, 58, 392-396. https://doi.org/10.1001/archneur.58.3.392

  21. 21. Perry, G., Nunomura, A., Hirai, K., Takeda, A., Aliev, G. and Smith, M.A. (2000) Oxidative Damage in Alzheimer’s Disease: The Metabolic Dimension. International Journal of Developmental Neuroscience, 18, 417-421. https://doi.org/10.1016/S0736-5748(00)00006-X

  22. 22. Smith, M.A., Richey Harris, P.L., Sayre, L.M., Beckman, J.S. and Perry, G. (1997) Widespread Peroxynitrite-Mediated Damage in Alzheimer’s Disease. The Journal of Neuroscience, 17, 2653-2657.

  23. 23. Mark, R.J., Pang, Z., Geddes, J.W., Uchida, K. and Mattson, M.P. (1997) Amyloid Beta-Peptide Impairs Glucose Transport in Hippocampal and Cortical Neurons: Involvement of Membrane Lipid Peroxidation. The Journal of Neuroscience, 17, 1046-1054.

  24. 24. Guglielmotto, M., Giliberto, L., Tamagno, E. and Tabaton, M. (2010) Oxidative Stress Mediates the Pathogenic Effect of Different Alzheimer’s Disease Risk Factors. Frontiers in Aging Neuroscience, 2, 3. https://doi.org/10.3389/neuro.24.003.2010

  25. 25. Kern, A. and Behl, C. (2009) The Unsolved Relationship of Brain Aging and Late-Onset Alzheimer Disease. Biochimica et Biophysica Acta, 1790, 1124-1132. https://doi.org/10.1016/j.bbagen.2009.07.016

  26. 26. Tamagno, E., Guglielmotto, M., Aragno, M., et al. (2008) Oxidative Stress Activates a Positive Feedback between the Gamma- and Beta-Secretase Cleavages of the Beta-Amyloid Precursor Protein. Journal of Neurochemistry, 104, 683-695.

  27. 27. Butterfield, D.A., Swomley, A.M. and Sultana, R. (2013) Amyloid Beta-Peptide (1-42)-Induced Oxidative Stress in Alzheimer Disease: Importance in Disease Pathogenesis and Progression. Antioxidants & Redox Signaling, 19, 823-835.https://doi.org/10.1089/ars.2012.5027

  28. 28. Tamagno, E., Guglielmotto, M., Monteleone, D. and Tabaton, M. (2012) Amyloid-Beta Production: Major Link between Oxidative Stress and BACE1. Neurotoxicity Research, 22, 208-219. https://doi.org/10.1007/s12640-011-9283-6

  29. 29. Tan, J.L., Li, Q.X., Ciccotosto, G.D., et al. (2013) Mild Oxidative Stress Induces Redistribution of BACE1 in Non-Apoptotic Conditions and Promotes the Amyloidogenic Processing of Alzheimer’s Disease Amyloid Precursor Protein. PLoS ONE, 8, e61246. https://doi.org/10.1371/journal.pone.0061246

  30. 30. Jiang, Y., Rigoglioso, A., Peterhoff, C.M., et al. (2016) Partial BACE1 Reduction in a Down Syndrome Mouse Model Blocks Alzheimer-Related Endosomal Anomalies and Cholinergic Neurodegeneration: Role of APP-CTF. Neurobiology of Aging, 39, 90-98. https://doi.org/10.1016/j.neurobiolaging.2015.11.013

  31. 31. Calsolaro, V. and Edison, P. (2016) Neuroinflammation in Alzheimer’s Disease: Current Evidence and Future Directions. Alzheimer’s & Dementia, 12, 719-732. https://doi.org/10.1016/j.jalz.2016.02.010

  32. 32. Kontush, A., Mann, U., Arlt, S., et al. (2001) Influence of Vitamin E and C Supplementation on Lipoprotein Oxidation in Patients with Alzheimer’s Disease. Free Radical Biology and Medicine, 31, 345-354. https://doi.org/10.1016/S0891-5849(01)00595-0

  33. 33. Salerno-Kennedy, R. and Cashman, K.D. (2005) Relationship between Dementia and Nutrition-Related Factors and Disorders: An Overview. International Journal for Vitamin and Nutrition Research, 75, 83-95. https://doi.org/10.1024/0300-9831.75.2.83

  34. 34. Whalley, L.J., Starr, J.M. and Deary, I.J. (2004) Diet and Dementia. Post Reproductive Health, 10, 113-117. https://doi.org/10.1258/1362180043654575

  35. 35. Holmes, C., Cunningham, C., Zotova, E., et al. (2009) Systemic Inflammation and Disease Progression in Alzheimer Disease. Neurology, 73, 768-774.https://doi.org/10.1212/WNL.0b013e3181b6bb95

  36. 36. Cunningham, C. (2013) Microglia and Neurodegeneration: The Role of Systemic Inflammation. Glia, 61, 71-90. https://doi.org/10.1002/glia.22350

  37. 37. McGeer, P.L. and McGeer, E.G. (2007) NSAIDs and Alzheimer Disease: Epidemiological, Animal Model and Clinical Studies. Neurobiology of Aging, 28, 639-647. https://doi.org/10.1016/j.neurobiolaging.2006.03.013

  38. 38. Sun, J., Zhang, S., Zhang, X., Zhang, X., Dong, H. and Qian, Y. (2015) IL-17A Is Implicated in Lipopolysaccharide-Induced Neuroinflammation and Cognitive Impairment in Aged Rats via Microglial Activation. Journal of Neuroinflammation, 12, 165. https://doi.org/10.1186/s12974-015-0394-5

  39. 39. Zhang, X.Y., Cao, J.B., Zhang, L.M., Li, Y.F. and Mi, W.D. (2015) Deferoxamine Attenuates Lipopolysaccharide-Induced Neuroinflammation and Memory Impairment in Mice. Journal of Neuroinflammation, 12, 20. https://doi.org/10.1186/s12974-015-0238-3

  40. 40. Craft, S. (2007) Insulin Resistance and Alzheimer’s Disease Pathogenesis: Potential Mechanisms and Implications for Treatment. Current Alzheimer Research, 4, 147-152. https://doi.org/10.2174/156720507780362137

  41. 41. Birch, A.M., Katsouri, L. and Sastre, M. (2014) Modulation of Inflammation in Transgenic Models of Alzheimer’s Disease. Journal of Neuroinflammation, 11, 25. https://doi.org/10.1186/1742-2094-11-25

  42. 42. Landreth, G.E. and Heneka, M.T. (2001) Anti-Inflammatory Actions of Peroxisome Proliferator-Activated Receptor Gamma Agonists in Alzheimer’s Disease. Neurobiology of Aging, 22, 937-944. https://doi.org/10.1016/S0197-4580(01)00296-2

  43. 43. Kitamura, Y., Shimohama, S., Koike, H., et al. (1999) Increased Expression of Cyclooxygenases and Peroxisome Proliferator-Activated Receptor-Gamma in Alzheimer’s Disease Brains. Biochemical and Biophysical Research Communications, 254, 582-586. https://doi.org/10.1006/bbrc.1998.9981

  44. 44. Ricote, M., Huang, J.T., Welch, J.S. and Glass, C.K. (1999) The Peroxisome Proliferator-Activated Receptor (PPARgamma) as a Regulator of Monocyte/Macrophage Function. Journal of Leukocyte Biology, 66, 733-739.

  45. 45. Murphy, G.J. and Holder, J.C. (2000) PPAR-Gamma Agonists: Therapeutic Role in Diabetes, Inflammation and Cancer. Trends in Pharmacological Sciences, 21, 469-474. https://doi.org/10.1016/S0165-6147(00)01559-5

  46. 46. Sastre, M. and Gentleman, S.M. (2010) NSAIDs: How They Work and Their Prospects as Therapeutics in Alzheimer’s Disease. Frontiers in Aging Neuroscience, 2, 20.

  47. 47. Mosconi, L., Pupi, A. and De Leon, M.J. (2008) Brain Glucose Hypometabolism and Oxidative Stress in Preclinical Alzheimer’s Disease. Annals of the New York Academy of Sciences, 1147, 180-195. https://doi.org/10.1196/annals.1427.007

  48. 48. Kim, B. and Feldman, E.L. (2015) Insulin Resistance as a Key Link for the Increased Risk of Cognitive Impairment in the Metabolic Syndrome. Experimental & Molecular Medicine, 47, e149. https://doi.org/10.1038/emm.2015.3

  49. 49. Craft, S. (2005) Insulin Resistance Syndrome and Alzheimer’s Disease: Age- and Obesity-Related Effects on Memory, Amyloid, and Inflammation. Neurobiology of Aging, 26, 65-69. https://doi.org/10.1016/j.neurobiolaging.2005.08.021

  50. 50. de la Monte, S.M. (2009) Insulin Resistance and Alzheimer’s Disease. BMB Reports, 42, 475-481. https://doi.org/10.5483/BMBRep.2009.42.8.475

  51. 51. Takashima, A. (2006) GSK-3 Is Essential in the Pathogenesis of Alzheimer’s Disease. Journal of Alzheimer’s Disease, 9, 309-317. https://doi.org/10.3233/JAD-2006-9S335

  52. 52. Cai, Z., Xiao, M., Chang, L. and Yan, L.J. (2015) Role of Insulin Resistance in Alzheimer’s Disease. Metabolic Brain Disease, 30, 839-851.https://doi.org/10.1007/s11011-014-9631-3

  53. 53. Son, S.M., Song, H., Byun, J., et al. (2012) Accumulation of Autophagosomes Contributes to Enhanced Amyloidogenic APP Processing under Insulin-Resistant Conditions. Autophagy, 8, 1842-1844. https://doi.org/10.4161/auto.21861

  54. 54. Yu, Y., Li, X., Blanchard, J., et al. (2015) Insulin Sensitizers Improve Learning and Attenuate Tau Hyperphosphorylation and Neuroinflammation in 3xTg-AD Mice. Journal of Neural Transmission (Vienna), 122, 593-606.https://doi.org/10.1007/s00702-014-1294-z

  55. 55. Escribano, L., Simon, A.M., Gimeno, E., et al. (2010) Rosiglitazone Rescues Memory Impairment in Alzheimer’s Transgenic Mice: Mechanisms Involving a Reduced Amyloid and Tau Pathology. Neuropsychopharmacology, 35, 1593-1604.https://doi.org/10.1038/npp.2010.32

  56. 56. Abbatecola, A.M., Lattanzio, F., Molinari, A.M., et al. (2010) Rosiglitazone and Cognitive Stability in Older Individuals with Type 2 Diabetes and Mild Cognitive Impairment. Diabetes Care, 33, 1706-1711. https://doi.org/10.2337/dc09-2030

  57. 57. Fei, L., Yong-Jun, H., Zhang-Min, M., et al. (2015) Rosiglitazone Attenuates Memory Impairment in Aged Rat with Diabetes by Inhibiting NF-Kappa B Signal Pathway Activation. Experimental and Clinical Endocrinology & Diabetes, 123, 536-542.https://doi.org/10.1055/s-0035-1559607

  58. 58. Watson, G.S., Cholerton, B.A., Reger, M.A., et al. (2005) Preserved Cognition in Patients with Early Alzheimer Disease and Amnestic Mild Cognitive Impairment during Treatment with Rosiglitazone: A Preliminary Study. The American Journal of Geriatric Psychiatry, 13, 950-958.

  59. 59. Sato, T., Hanyu, H., Hirao, K., Kanetaka, H., Sakurai, H. and Iwamoto, T. (2011) Efficacy of PPAR-Gamma Agonist Pioglitazone in Mild Alzheimer Disease. Neurobiology of Aging, 32, 1626-1633. https://doi.org/10.1016/j.neurobiolaging.2009.10.009

  60. 60. Geldmacher, D.S., Fritsch, T., McClendon, M.J. and Landreth, G. (2011) A Randomized Pilot Clinical Trial of the Safety of Pioglitazone in Treatment of Patients with Alzheimer Disease. Archives of Neurology, 68, 45-50.https://doi.org/10.1001/archneurol.2010.229

  61. 61. Feingold, K.R., Grunfeld, C., Heuer, J.G., et al. (2012) FGF21 Is Increased by Inflammatory Stimuli and Protects Leptin-Deficient ob/ob Mice from the Toxicity of Sepsis. Endocrinology, 153, 2689-2700. https://doi.org/10.1210/en.2011-1496

  62. 62. Fisher, F.M. and Maratos-Flier, E. (2016) Understanding the Physiology of FGF21. Annual Review of Physiology, 78, 223-241. https://doi.org/10.1146/annurev-physiol-021115-105339

  63. 63. Sa-Nguanmoo, P., Chattipakorn, N. and Chattipakorn, S.C. (2016) Potential Roles of Fibroblast Growth Factor 21 in the Brain. Metabolic Brain Disease, 31, 239-248.https://doi.org/10.1007/s11011-015-9789-3

  64. 64. Kharitonenkov, A. (2009) FGFs and Metabolism. Current Opinion in Pharmacology, 9, 805-810. https://doi.org/10.1016/j.coph.2009.07.001

  65. 65. Ogawa, Y., Kurosu, H., Yamamoto, M., et al. (2007) βKlotho Is Required for Metabolic Activity of Fibroblast Growth Factor 21. Proceedings of the National Academy of Sciences of the United States of America, 104, 7432-7437.https://doi.org/10.1073/pnas.0701600104

  66. 66. Adams, A.C., Cheng, C.C., Coskun, T. and Kharitonenkov, A. (2012) FGF21 Requires Betaklotho to Act in Vivo. PLoS ONE, 7, e49977. https://doi.org/10.1371/journal.pone.0049977

  67. 67. Ge, H., Baribault, H., Vonderfecht, S., et al. (2012) Characterization of a FGF19 Variant with Altered Receptor Specificity Revealed a Central Role for FGFR1c in the Regulation of Glucose Metabolism. PLoS ONE, 7, e33603.https://doi.org/10.1371/journal.pone.0033603

  68. 68. Yang, C., Jin, C., Li, X., Wang, F., McKeehan, W.L. and Luo, Y. (2012) Differential Specificity of Endocrine FGF19 and FGF21 to FGFR1 and FGFR4 in Complex with KLB. PLoS ONE, 7, e33870. https://doi.org/10.1371/journal.pone.0033870

  69. 69. Tomiyama, K., Maeda, R., Urakawa, I., et al. (2010) Relevant Use of Klotho in FGF19 Subfamily Signaling System in Vivo. Proceedings of the National Academy of Sciences of the United States of America, 107, 1666-1671.https://doi.org/10.1073/pnas.0913986107

  70. 70. Makela, J., Tselykh, T.V., Maiorana, F., et al. (2014) Fibroblast Growth Factor-21 Enhances Mitochondrial Functions and Increases the Activity of PGC-1Alpha in Human Dopaminergic Neurons via Sirtuin-1. SpringerPlus, 3, 2. https://doi.org/10.1186/2193-1801-3-2

  71. 71. Leng, Y., Wang, Z., Tsai, L.K., et al. (2015) FGF-21, a Novel Metabolic Regulator, Has a Robust Neuroprotective Role and Is Markedly Elevated in Neurons by Mood Stabilizers. Molecular Psychiatry, 20, 215-223. https://doi.org/10.1038/mp.2013.192

  72. 72. Tan, B.K., Hallschmid, M., Adya, R., Kern, W., Lehnert, H. and Randeva, H.S. (2011) Fibroblast Growth Factor 21 (FGF21) in Human Cerebrospinal Fluid: Relationship with Plasma FGF21 and Body Adiposity. Diabetes, 60, 2758-2762. https://doi.org/10.2337/db11-0672

  73. 73. Hsuchou, H., Pan, W. and Kastin, A.J. (2007) The Fasting Polypeptide FGF21 Can Enter Brain from Blood. Peptides, 28, 2382-2386. https://doi.org/10.1016/j.peptides.2007.10.007

  74. 74. Liang, Q., Zhong, L., Zhang, J., et al. (2014) FGF21 Maintains Glucose Homeostasis by Mediating the Cross Talk between Liver and Brain during Prolonged Fasting. Diabetes, 63, 4064-4075. https://doi.org/10.2337/db14-0541

  75. 75. Bookout, A.L., de Groot, M.H., Owen, B.M., et al. (2013) FGF21 Regulates Metabolism and Circadian Behavior by Acting on the Nervous System. Nature Medicine, 19, 1147-1152. https://doi.org/10.1038/nm.3249

  76. 76. Fon Tacer, K., Bookout, A.L., Ding, X., et al. (2010) Research Resource: Comprehensive Expression Atlas of the Fibroblast Growth Factor System in Adult Mouse. Molecular Endocrinology, 24, 2050-2064. https://doi.org/10.1210/me.2010-0142

  77. 77. Chau, M.D., Gao, J., Yang, Q., Wu, Z. and Gromada, J. (2010) Fibroblast Growth Factor 21 Regulates Energy Metabolism by Activating the AMPK-SIRT1-PGC-1Alpha Pathway. Proceedings of the National Academy of Sciences of the United States of America, 107, 12553-12558. https://doi.org/10.1073/pnas.1006962107

  78. 78. Ye, D., Wang, Y., Li, H., et al. (2014) Fibroblast Growth Factor 21 Protects against Acetaminophen-Induced Hepatotoxicity by Potentiating Peroxisome Proliferator-Activated Receptor Coactivator Protein-1Alpha-Mediated Antioxidant Capacity in Mice. Hepatology, 60, 977-989. https://doi.org/10.1002/hep.27060

  79. 79. Fisher, F.M., Kleiner, S., Douris, N., et al. (2012) FGF21 Regulates PGC-1Alpha and Browning of White Adipose Tissues in Adaptive Thermogenesis. Genes & Development, 26, 271-281. https://doi.org/10.1101/gad.177857.111

  80. 80. Potthoff, M.J., Inagaki, T., Satapati, S., et al. (2009) FGF21 Induces PGC-1Alpha and Regulates Carbohydrate and Fatty Acid Metabolism during the Adaptive Starvation Response. Proceedings of the National Academy of Sciences of the United States of America, 106, 10853-10858. https://doi.org/10.1073/pnas.0904187106

  81. 81. Katsouri, L., Blondrath, K. and Sastre, M. (2012) Peroxisome Proliferator-Activated Receptor-Gamma Cofactors in Neurodegeneration. IUBMB Life, 64, 958-964.https://doi.org/10.1002/iub.1097

  82. 82. Austin, S. and St-Pierre, J. (2012) PGC1Alpha and Mitochondrial Metabolism—Emerging Concepts and Relevance in Ageing and Neurodegenerative Disorders. Journal of Cell Science, 125, 4963-4971. https://doi.org/10.1242/jcs.113662

  83. 83. Qin, W., Haroutunian, V., Katsel, P., et al. (2009) PGC-1Alpha Expression Decreases in the Alzheimer Disease Brain as a Function of Dementia. Archives of Neurology, 66, 352-361. https://doi.org/10.1001/archneurol.2008.588

  84. 84. Katsouri, L., Parr, C., Bogdanovic, N., Willem, M. and Sastre, M. (2011) PPARγ Co-Activator-1α (PGC-1α) Reduces Amyloid-β Generation through a PPARγ-Dependent Mechanism. Journal of Alzheimer’s Disease, 25, 151-162.

  85. 85. Katsouri, L., Lim, Y.M., Blondrath, K., et al. (2016) PPARγ-Coactivator-1α Gene Transfer Reduces Neuronal Loss and Amyloid-β generation by Reducing β-Secretase in an Alzheimer’s Disease Model. Proceedings of the National Academy of Sciences of the United States of America, 113, 12292-12297. https://doi.org/10.1073/pnas.1606171113

  86. 86. Planavila, A., Redondo, I., Hondares, E., et al. (2013) Fibroblast Growth Factor 21 Protects against Cardiac Hypertrophy in Mice. Nature Communications, 4, Article No. 2019. https://doi.org/10.1038/ncomms3019

  87. 87. Planavila, A., Redondo-Angulo, I., Ribas, F., et al. (2015) Fibroblast Growth Factor 21 Protects the Heart from Oxidative Stress. Cardiovascular Research, 106, 19-31. https://doi.org/10.1093/cvr/cvu263

  88. 88. Yu, Y., Bai, F., Wang, W., et al. (2015) Fibroblast Growth Factor 21 Protects Mouse Brain against D-Galactose Induced Aging via Suppression of Oxidative Stress Response and Advanced Glycation end Products Formation. Pharmacology, Biochemistry and Behavior, 133, 122-131. https://doi.org/10.1016/j.pbb.2015.03.020

  89. 89. Breitner, J.C. (1996) The Role of Anti-Inflammatory Drugs in the Prevention and Treatment of Alzheimer’s Disease. Annual Review of Medicine, 47, 401-411. https://doi.org/10.1146/annurev.med.47.1.401

  90. 90. Akiyama, H., Barger, S., Barnum, S., et al. (2000) Inflammation and Alzheimer’s Disease. Neurobiology of Aging, 21, 383-421. https://doi.org/10.1016/S0197-4580(00)00124-X

  91. 91. Heneka, M.T. and O’Banion, M.K. (2007) Inflammatory Processes in Alzheimer’s Disease. Journal of Neuroimmunology, 184, 69-91. https://doi.org/10.1016/j.jneuroim.2006.11.017

  92. 92. Kim, H.W., Lee, J.E., Cha, J.J., et al. (2013) Fibroblast Growth Factor 21 Improves Insulin Resistance and Ameliorates Renal Injury in db/db Mice. Endocrinology, 154, 3366-3376. https://doi.org/10.1210/en.2012-2276

  93. 93. Yu, Y., He, J., Li, S., et al. (2016) Fibroblast Growth Factor 21 (FGF21) Inhibits Macrophage-Mediated Inflammation by Activating Nrf2 and Suppressing the NF-κB Signaling Pathway. International Immunopharmacology, 38, 144-152. https://doi.org/10.1016/j.intimp.2016.05.026

  94. 94. Xu, J., Lloyd, D.J., Hale, C., et al. (2009) Fibroblast Growth Factor 21 Reverses Hepatic Steatosis, Increases Energy Expenditure, and Improves Insulin Sensitivity in Diet-Induced Obese Mice. Diabetes, 58, 250-259. https://doi.org/10.2337/db08-0392

  95. 95. So, W.Y., Cheng, Q., Xu, A., Lam, K.S. and Leung, P.S. (2015) Loss of Fibroblast Growth Factor 21 Action Induces Insulin Resistance, Pancreatic Islet Hyperplasia and Dysfunction in Mice. Cell Death & Disease, 6, e1707. https://doi.org/10.1038/cddis.2015.80

  96. 96. Sarruf, D.A., Thaler, J.P., Morton, G.J., et al. (2010) Fibroblast Growth Factor 21 Action in the Brain Increases Energy Expenditure and Insulin Sensitivity in Obese Rats. Diabetes, 59, 1817-1824. https://doi.org/10.2337/db09-1878

  97. 97. Qing, Y., Yang, J. and Wan, C. (2015) Increased Serum Fibroblast Growth Factor 21 Levels in Patients with Schizophrenia. Australian and New Zealand Journal of Psychiatry, 49, 849-850. https://doi.org/10.1177/0004867415575380

  98. 98. Inagaki, T., Dutchak, P., Zhao, G., et al. (2007) Endocrine Regulation of the Fasting Response by PPARα-Mediated Induction of Fibroblast Growth Factor 21. Cell Metabolism, 5, 415-425. https://doi.org/10.1016/j.cmet.2007.05.003

Abbreviations

Aβ amyloid-β

AD Alzheimer’s disease

APOE4 apolipoprotein E4

APP amyloid precursor protein

APR acute phase response

BACE1 β-APP cleaving enzyme

CNS central nervous system

FGF21 fibroblast growth factor 21

FGFR fibroblast growth factor receptor

hPGC-1α human PGC-1α

IL-6 interleukin-6

KLB β-klotho

LPS lipopolysaccharide

MCI mild cognitive impairment

MCP-1 monocyte chemoattractant protein-1

MRI magnetic resonance imaging

Nrf2 nuclear transcription factor-E2-related factor 2

NSAIDs non-steroidal anti-inflammatory drugs

PS1/2 presenilins 1 and 2

PGC-1α peroxisome proliferator-activated receptor γ coactivator-1α

PPAR γ peroxisome proliferator-activated receptor γ

PPARs peroxisome proliferator-activated receptors

ROS reactive oxygen species

Sod2 superoxide dismutase-2

TgFgf21 transgenic overexpression of FGF21 mice

TNF-α tumor necrosis factor-α

Ucp2/Ucp3 uncoupling proteins2/3