Pro-inflammatory interleukin-6 signaling links cognitive impairments and peripheral metabolic alterations in Alzheimer's disease

Natalia M Lyra E Silva, Rafaella A Gonçalves, Tharick A Pascoal, Ricardo A S Lima-Filho, Elisa de Paula França Resende, Erica L M Vieira, Antonio L Teixeira, Leonardo C de Souza, Julyanna A Peny, Juliana T S Fortuna, Isadora C Furigo, Debora Hashiguchi, Vivian S Miya-Coreixas, Julia R Clarke, Jose F Abisambra, Beatriz M Longo, Jose Donato Jr, Paul E Fraser, Pedro Rosa-Neto, Paulo Caramelli, Sergio T Ferreira, Fernanda G De Felice, Natalia M Lyra E Silva, Rafaella A Gonçalves, Tharick A Pascoal, Ricardo A S Lima-Filho, Elisa de Paula França Resende, Erica L M Vieira, Antonio L Teixeira, Leonardo C de Souza, Julyanna A Peny, Juliana T S Fortuna, Isadora C Furigo, Debora Hashiguchi, Vivian S Miya-Coreixas, Julia R Clarke, Jose F Abisambra, Beatriz M Longo, Jose Donato Jr, Paul E Fraser, Pedro Rosa-Neto, Paulo Caramelli, Sergio T Ferreira, Fernanda G De Felice

Abstract

Alzheimer's disease (AD) is associated with memory impairment and altered peripheral metabolism. Mounting evidence indicates that abnormal signaling in a brain-periphery metabolic axis plays a role in AD pathophysiology. The activation of pro-inflammatory pathways in the brain, including the interleukin-6 (IL-6) pathway, comprises a potential point of convergence between memory dysfunction and metabolic alterations in AD that remains to be better explored. Using T2-weighted magnetic resonance imaging (MRI), we observed signs of probable inflammation in the hypothalamus and in the hippocampus of AD patients when compared to cognitively healthy control subjects. Pathological examination of post-mortem AD hypothalamus revealed the presence of hyperphosphorylated tau and tangle-like structures, as well as parenchymal and vascular amyloid deposits surrounded by astrocytes. T2 hyperintensities on MRI positively correlated with plasma IL-6, and both correlated inversely with cognitive performance and hypothalamic/hippocampal volumes in AD patients. Increased IL-6 and suppressor of cytokine signaling 3 (SOCS3) were observed in post-mortem AD brains. Moreover, activation of the IL-6 pathway was observed in the hypothalamus and hippocampus of AD mice. Neutralization of IL-6 and inhibition of the signal transducer and activator of transcription 3 (STAT3) signaling in the brains of AD mouse models alleviated memory impairment and peripheral glucose intolerance, and normalized plasma IL-6 levels. Collectively, these results point to IL-6 as a link between cognitive impairment and peripheral metabolic alterations in AD. Targeting pro-inflammatory IL-6 signaling may be a strategy to alleviate memory impairment and metabolic alterations in the disease.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1. T2-weighted signal hyperintensities in the…
Fig. 1. T2-weighted signal hyperintensities in the AD hypothalamus and hippocampus correlate inversely with MMSE scores and gray matter densities.
a Representative transversal (left) and coronal (right) T2-weighted images from control (CTL, top) and AD (bottom) patients. Arrows indicate the placement of right and left ROIs in the hypothalamus (yellow arrows) and hippocampus (white arrows). b Bars represent means ± SEM of T2-intensity signal ratios (hypothalamus relative to putamen) in control subjects (n = 18) and AD patients (n = 16); symbols represent individual subjects; p-value was calculated from student’s t-test and is shown in the graph. c Correlation between hypothalamus/putamen T2-intensity signal ratios and MMSE scores. Pearson correlation coefficient (r) and p-value are shown in the graph (n = 34). d Bars represent means ± SEM of T2-intensity signal ratios (hippocampus relative to putamen) in control subjects (n = 18) and AD patients (n = 16); symbols represent individual patients; p-value (Mann-Whitney test) is shown in the graph. e Correlation between hippocampus/putamen T2-intensity signal ratios and MMSE scores. Spearman correlation coefficient (r) and p-value are shown in the graph (n = 34). Bars represent means ± SEM of hypothalamic (f) and hippocampal (g) gray matter densities measured by voxel-based morphometry in control subjects (n = 17) and AD patients (n = 13); symbols represent individual patients; p-value (Student’s t-test) is shown in graph. h Correlation between hypothalamus gray matter density and hypothalamus/putamen T2-intensity signal ratios. Pearson correlation coefficient (r) and p-value are shown in the graph (n = 30). i Correlation between hippocampus gray matter density and hippocampus/putamen T2-intensity signal ratios. Spearman correlation coefficient (r) and p-value are shown in the graph (n = 30).
Fig. 2. Intraneuronal hyperphosphorylated tau aggregates, Aβ…
Fig. 2. Intraneuronal hyperphosphorylated tau aggregates, Aβ deposits and inflammatory markers in post-mortem AD hypothalamus.
a Representative photomicrographs showing immunostaining of Aβ deposits (6E10 antibody; red) in the lateral hypothalamus of AD cases (AD1: Female 77 years old; AD2: Female, 88 years old; AD3: Female, 82 years old) and age-matched control (CTL1: Female, 71 years old; CTL2: Female, 80 years old). Graph represents means ± SEM of number of plaques in AD patients or control individuals; p-value was calculated from student’s t-test, and is shown in the graph. b Representative photomicrographs of the lateral hypothalamus showing double immunofluorescence staining for Aβ (Abcam ab134022; red) and astrocytes (GFAP; green) in post-mortem AD (AD1: Female 77 years old; AD2: Female, 88 years old) and age-matched control (CTL: Female, 71 years old). Magnified images of specific fields are indicated by the dashed rectangles in the main panels. c Representative photomicrographs of the lateral hypothalamus showing positive intraneuronal immunostaining for AT8 (p-tau antibody; red) in neurons in AD cases (AD1: Female, 82 years old; AD2: Female 77 years old) and absence of AT8-positive cells in age-matched control (CTL1: Female, 71 years old; CTL2: Female, 80 years old). Graph represents means + SEM of number of plaques in AD patients or control individuals; p-value (Student’s t-test) is shown in the graph. d, e Representative micrographs of triple immunofluorescence staining for Aβ (Abcam ab134022; red), astrocytes (GFAP; green), and IL-6 (blue) in AD lateral hypothalamus (Female, 77 years old). White arrows indicate a cell with triple staining for GFAP, Aβ and IL-6, and yellow arrows show a GFAP-positive cell that does not colocalize with either Aβ or IL-6 immunoreactivities.
Fig. 3. IL-6 is upregulated in AD…
Fig. 3. IL-6 is upregulated in AD brain and plasma, and correlates inversely with MMSE.
Representative photomicrographs of triple immunofluorescence staining for Aβ (Abcam ab134022; red), astrocytes (GFAP; green), and IL-6 (blue) in post-mortem cingulate cortex from AD subjects (AD1: Female, 82 years old; AD2: Female, 88 years old) (a, b) and an age-matched control (CTL: Female, 80 years old) (c). d Means ± SEM of IL-6 protein levels measured by ELISA in post-mortem prefrontal cortex from AD (n = 8) or control subjects (n = 5); symbols represent different individuals; p-value was calculated from student’s t-test and is shown in the graph. e Representative immunoblot of SOCS3 (β-actin used as loading control) in post-mortem prefrontal cortex (PFC) from AD (n = 8) or control (n = 9) subjects. Graph shows means ± SEM of SOCS3/β-actin ratios; symbols represent different individuals; p-value (Student’s t-test) is shown in the graph. f Means ± SEM of plasma IL-6 in AD patients (n = 12) or control individuals (n = 14) from the same cohort that was studied by MRI in Fig. 1; symbols represent different individuals; p-value was calculated from unpaired t-test with Welch’s correction, and is shown in the graph. g Correlation between plasma IL-6 and hippocampus/putamen T2-intensity signal ratios for patients in the studied cohort (n = 26). Spearman correlation coefficient (r) and p-value are shown in the graph. h, i Correlations between plasma IL-6 and MMSE or FAB scores for patients in the studied cohort. Symbols represent different individuals (n = 26). Pearson correlation coefficients (r) and p-values are shown in the graphs.
Fig. 4. Brain IL-6 signaling is upregulated…
Fig. 4. Brain IL-6 signaling is upregulated and mediates memory impairment in AD mouse models.
a Means ± SEM of IL-6 protein levels in the hippocampus of 11-month-old APP/PS1 (n = 7) and WT littermates (n = 5); symbols represent individual animals; p-value (Student’s t-test) is shown in graph. b Representative immunoblot of pSTAT3, STAT3, and β-actin (used as a loading control) in the hippocampus of 11-month-old APP/PS1 (n = 6) and WT mice (n = 8). Graph shows pSTAT3/STAT3 ratio; symbols represent individual animals; p-value (Student’s t-test) is shown in graph. c, d Means ± SEM of IL-6 (n = 4 Veh; 6 AβOs) and SOCS3 expression (n = 7 Veh; 8 AβOs) (mRNA levels, expressed as fold-change relative to vehicle-infused mice) in mouse hippocampus 24 h after i.c.v. infusion of AβOs. β-actin was used as a housekeeping control. Symbols represent individual animals; p-values were calculated from student’s t-test and unpaired t-test with Welch’s correction, and are shown in graphs. e Representative immunoblot of pSTAT3, STAT3, and β-actin (loading control) in mouse hippocampus 24 h after i.c.v. infusion of AβOs (n = 8) (or vehicle, n = 7). Graph shows pSTAT3/STAT3 ratio; symbols represent individual animals; p-value (Student’s t-test) is shown in graph. f Representative immunofluorescence staining for pSTAT3 (red), MAP2 (green), and DAPI (blue) in a primary hippocampal culture exposed to AβOs (or vehicle) for 24 h. Yellow arrowhead indicates pSTAT3 staining (red) in the nucleus of a cell that is not positive for MAP2 (green). g Novel Object Recognition (NOR) task with 11-month-old male APP/PS1 mice and WT littermates (n = 7–12) after 3 i.c.v. injections of anti-IL-6 (αIL6) or anti-GFP (αGFP), as indicated. Symbols represent individual mice, n = 7–12 per experimental condition; *p < 0.05, **p < 0.01, ***p < 0.001, one-sample Student’s t-test. h NOR task with 3-month-old mice i.c.v-infused with AβOs (or vehicle) and AG490. Bars represent means ± SEM. Symbols represent individual mice, n = 7–10 per experimental condition; *p < 0.05, **p < 0.01, one-sample Student’s t-test. Data are representative of at least two independent experiments.
Fig. 5. Central IL-6 promotes peripheral metabolic…
Fig. 5. Central IL-6 promotes peripheral metabolic dysfunction in AD mouse models.
a Expression of IL-6 in the mouse hypothalamus 4 h after i.c.v. infusion of AβOs (n = 5) (or vehicle, n = 4). β-actin was used as housekeeping control. Bars represent means ± SEM; symbols correspond to individual mice; p-value (Student’s t-test) is shown in graph. b IL-6 in the mouse hypothalamus 4 h after AβOs (n = 5) or vehicle (n = 5) were i.c.v.-infused. Bars represent means ± SEM; symbols correspond to individual mice; p-value (Student’s t-test) is shown in graph. c Representative in situ hybridization images for SOCS3 in the mouse hypothalamus 7 days after AβOs (n = 4) or vehicle (n = 4) were i.c.v.-infused. Graph shows integrated densities (means ± SEM) in the regions indicated by dashed ellipses in AβO- or vehicle-infused mice. Symbols correspond to individual mice; p-value (Student’s t-test) is shown in the graph. d IL-6 in hypothalamic homogenates from 11-month-old APP/PS1 mice (n = 6) or WT litttermates (n = 8). Bars correspond to means ± SEM. Symbols correspond to individual mice; p-value (Student’s t-test) is shown in the graph. e Representative immunoblot for SOCS3 in the hypothalamus of 11-month-old male APP/PS1 mice (n = 4) or WT littermates (n = 4). Bars in graph correspond to means ± SEM. Symbols correspond to individual mice; p-value (Mann–Whitney test) is shown in graph. f Glucose tolerance test in WT mice 36 h after i.c.v. infusion of AβOs (or vehicle) and treatment with AG490. Symbols correspond to individual mice and represent means ± SEM, n = 4–6 per experimental group; *p < 0.05 from two-way ANOVA followed by Tukey’s post-hoc test comparing Veh vs. AβOs and AβOs vs. AβOs + AG490). g Glucose tolerance test in 11-month-old APP/PS1 mice after 4 i.c.v. injections of anti-IL-6 (αIL6) or anti-GFP (αGFP). Mice received 1 g/kg of glucose (i.p.) and blood glucose was measured at the indicated timepoints. Symbols correspond to individual mice and represent means ± SEM, n = 4-6 per experimental group; ***p < 0.001, ****p < 0.0001 from two-way ANOVA followed by Tukey’s post-hoc test comparing WT + αGFP vs. APP/PS1 + αGFP and APP/PS1 + αGFP vs. APP/PS1 + αIL6). h Plasma IL-6 in 11-month-old APP/PS1 mice after 4 i.c.v. injections of anti-IL-6 (αIL6) or anti-GFP (αGFP). Bars represent means ± SEM. Symbols correspond to individual mice, n = 8–12 per experimental condition; p-values were calculated from two-way ANOVA followed by Tukey’s post-hoc test and are shown in the graph. Data are representative of at least two independent experiments.

References

    1. Alzheimer’s A. Alzheimer’s disease facts and figures. Alzheimer’s Dement. J. Alzheimer’s Assoc. 2016;12:459–509.
    1. Clarke JR, et al. Alzheimer-associated Abeta oligomers impact the central nervous system to induce peripheral metabolic deregulation. EMBO Mol. Med. 2015;7:190–210. doi: 10.15252/emmm.201404183.
    1. Craft S, et al. Cerebrospinal fluid and plasma insulin levels in Alzheimer’s disease: relationship to severity of dementia and apolipoprotein E genotype. Neurology. 1998;50:164–168. doi: 10.1212/WNL.50.1.164.
    1. Gil-Bea FJ, et al. Insulin levels are decreased in the cerebrospinal fluid of women with prodomal Alzheimer’s disease. J. Alzheimer’s Dis.: JAD. 2010;22:405–413. doi: 10.3233/JAD-2010-100795.
    1. Janson J, et al. Increased risk of type 2 diabetes in Alzheimer disease. Diabetes. 2004;53:474–481. doi: 10.2337/diabetes.53.2.474.
    1. Heni M, et al. Evidence for altered transport of insulin across the blood-brain barrier in insulin-resistant humans. Acta diabetologica. 2014;51:679–681. doi: 10.1007/s00592-013-0546-y.
    1. Takechi R, et al. Blood-brain barrier dysfunction precedes cognitive decline and neurodegeneration in diabetic insulin resistant mouse model: an implication for causal link. Front. Aging Neurosci. 2017;9:399. doi: 10.3389/fnagi.2017.00399.
    1. Fakhoury M. Microglia and astrocytes in Alzheimer’s disease: implications for therapy. Curr. Neuropharmacol. 2018;16:508–518. doi: 10.2174/1570159X15666170720095240.
    1. Heneka MT, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015;14:388–405. doi: 10.1016/S1474-4422(15)70016-5.
    1. Ferreira ST, Clarke JR, Bomfim TR, De Felice FG. Inflammation, defective insulin signaling, and neuronal dysfunction in Alzheimer’s disease. Alzheimer’s Dement.: J. Alzheimer’s Assoc. 2014;10:S76–S83. doi: 10.1016/j.jalz.2013.12.010.
    1. Lourenco MV, et al. TNF-alpha mediates PKR-dependent memory impairment and brain IRS-1 inhibition induced by Alzheimer’s beta-amyloid oligomers in mice and monkeys. Cell Metab. 2013;18:831–843. doi: 10.1016/j.cmet.2013.11.002.
    1. Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann. Transl. Med. 2015;3:136.
    1. Dunn AJ. Cytokine activation of the HPA axis. Ann. N. Y. Acad. Sci. 2000;917:608–617. doi: 10.1111/j.1749-6632.2000.tb05426.x.
    1. Elwood E, Lim Z, Naveed H, Galea I. The effect of systemic inflammation on human brain barrier function. Brain, Behav., Immun. 2017;62:35–40. doi: 10.1016/j.bbi.2016.10.020.
    1. Bettcher BM, Kramer JH. Longitudinal inflammation, cognitive decline, and Alzheimer’s disease: a mini-review. Clin. Pharmacol. therapeutics. 2014;96:464–469. doi: 10.1038/clpt.2014.147.
    1. Fraga VG, et al. Inflammatory and pro-resolving mediators in frontotemporal dementia and Alzheimer’s disease. Neuroscience. 2019;421:123–135. doi: 10.1016/j.neuroscience.2019.09.008.
    1. Miwa K, Okazaki S, Sakaguchi M, Mochizuki H, Kitagawa K. Interleukin-6, interleukin-6 receptor gene variant, small-vessel disease and incident dementia. Eur. J. Neurol. 2016;23:656–663. doi: 10.1111/ene.12921.
    1. Windham BG, et al. Associations between inflammation and cognitive function in African Americans and European Americans. J. Am. Geriatrics Soc. 2014;62:2303–2310. doi: 10.1111/jgs.13165.
    1. Huell M, Strauss S, Volk B, Berger M, Bauer J. Interleukin-6 is present in early stages of plaque formation and is restricted to the brains of Alzheimer’s disease patients. Acta Neuropathol. 1995;89:544–551. doi: 10.1007/BF00571510.
    1. Quintanilla RA, Orellana DI, Gonzalez-Billault C, Maccioni RB. Interleukin-6 induces Alzheimer-type phosphorylation of tau protein by deregulating the cdk5/p35 pathway. Exp. Cell Res. 2004;295:245–257. doi: 10.1016/j.yexcr.2004.01.002.
    1. Burton MD, Johnson RW. Interleukin-6 trans-signaling in the senescent mouse brain is involved in infection-related deficits in contextual fear conditioning. Brain Behav. Immun. 2012;26:732–738. doi: 10.1016/j.bbi.2011.10.008.
    1. Heyser CJ, Masliah E, Samimi A, Campbell IL, Gold LH. Progressive decline in avoidance learning paralleled by inflammatory neurodegeneration in transgenic mice expressing interleukin 6 in the brain. Proc. Natl Acad. Sci. USA. 1997;94:1500–1505. doi: 10.1073/pnas.94.4.1500.
    1. Brosseron F, Krauthausen M, Kummer M, Heneka MT. Body fluid cytokine levels in mild cognitive impairment and Alzheimer’s disease: a comparative overview. Mol. Neurobiol. 2014;50:534–544. doi: 10.1007/s12035-014-8657-1.
    1. Swardfager W, et al. A meta-analysis of cytokines in Alzheimer’s disease. Biol. Psychiatry. 2010;68:930–941. doi: 10.1016/j.biopsych.2010.06.012.
    1. McKhann GM, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement. 2011;7:263–269. doi: 10.1016/j.jalz.2011.03.005.
    1. Brucki SM, Nitrini R, Caramelli P, Bertolucci PH, Okamoto IH. Suggestions for utilization of the mini-mental state examination in Brazil. Arquivos de. neuro-psiquiatria. 2003;61:777–781. doi: 10.1590/S0004-282X2003000500014.
    1. Machado TH, et al. Normative data for healthy elderly on the phonemic verbal fluency task - FAS. Dement. Neuropsychol. 2009;3:55–60. doi: 10.1590/S1980-57642009DN30100011.
    1. Nitrini R, et al. Performance of illiterate and literate nondemented elderly subjects in two tests of long-term memory. J. Int. Neuropsychol. Soc. 2004;10:634–638. doi: 10.1017/S1355617704104062.
    1. Beato RG, Nitrini R, Formigoni AP, Caramelli P. Brazilian version of the Frontal Assessment Battery (FAB): preliminary data on administration to healthy elderly. Dement. Neuropsychologia. 2007;1:59–65. doi: 10.1590/S1980-57642008DN10100010.
    1. Pauli WM, Nili AN, Tyszka JM. A high-resolution probabilistic in vivo atlas of human subcortical brain nuclei. Sci. data. 2018;5:180063. doi: 10.1038/sdata.2018.63.
    1. Klein A, Tourville J. 101 labeled brain images and a consistent human cortical labeling protocol. Front. Neurosci. 2012;6:171. doi: 10.3389/fnins.2012.00171.
    1. Pascoal TA, et al. Abeta-induced vulnerability propagates via the brain’s default mode network. Nat. Commun. 2019;10:2353. doi: 10.1038/s41467-019-10217-w.
    1. Cascino GD. Temporal lobe epilepsy: more than hippocampal pathology. Epilepsy Curr. 2005;5:187–189. doi: 10.1111/j.1535-7511.2005.00059.x.
    1. Deulofeu Fontanillas F, et al. Massive hemoptysis secondary to mycotic aortic aneurysm. de. Med. interna. 1989;6:373–375.
    1. Pasternak O, Kubicki M, Shenton ME. In vivo imaging of neuroinflammation in schizophrenia. Schizophrenia Res. 2016;173:200–212. doi: 10.1016/j.schres.2015.05.034.
    1. Souza PVS, Bortholin T, Naylor FGM, Pinto W, Oliveira ASB. Teaching NeuroImages: early-onset dementia and demyelinating neuropathy disclosing cerebrotendinous xanthomatosis. Neurology. 2017;89:e134. doi: 10.1212/WNL.0000000000004363.
    1. Virel A, et al. Magnetic resonance imaging as a tool to image neuroinflammation in a rat model of Parkinson’s disease–phagocyte influx to the brain is promoted by bilberry-enriched diet. Eur. J. Neurosci. 2015;42:2761–2771. doi: 10.1111/ejn.13044.
    1. Thaler JP, et al. Obesity is associated with hypothalamic injury in rodents and humans. J. Clin. Investig. 2012;122:153–162. doi: 10.1172/JCI59660.
    1. Lambert MP, et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc. Natl Acad. Sci. USA. 1998;95:6448–6453. doi: 10.1073/pnas.95.11.6448.
    1. Figueiredo CP, et al. Memantine rescues transient cognitive impairment caused by high-molecular-weight abeta oligomers but not the persistent impairment induced by low-molecular-weight oligomers. J. Neurosci.: . J. Soc. Neurosci. 2013;33:9626–9634. doi: 10.1523/JNEUROSCI.0482-13.2013.
    1. Seixas da Silva GS, et al. Amyloid-beta oligomers transiently inhibit AMP-activated kinase and cause metabolic defects in hippocampal neurons. J. Biol. Chem. 2017;292:7395–7406. doi: 10.1074/jbc.M116.753525.
    1. De Felice FG, et al. Protection of synapses against Alzheimer’s-linked toxins: insulin signaling prevents the pathogenic binding of Abeta oligomers. Proc. Natl Acad. Sci. USA. 2009;106:1971–1976. doi: 10.1073/pnas.0809158106.
    1. Batista AF, et al. The diabetes drug liraglutide reverses cognitive impairment in mice and attenuates insulin receptor and synaptic pathology in a non-human primate model of Alzheimer’s disease. J. Pathol. 2018;245:85–100. doi: 10.1002/path.5056.
    1. Donato J, Jr., Frazao R, Fukuda M, Vianna CR, Elias CF. Leptin induces phosphorylation of neuronal nitric oxide synthase in defined hypothalamic neurons. Endocrinology. 2010;151:5415–5427. doi: 10.1210/en.2010-0651.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262.
    1. Berkseth KE, et al. Hypothalamic gliosis associated with high-fat diet feeding is reversible in mice: a combined immunohistochemical and magnetic resonance imaging study. Endocrinology. 2014;155:2858–2867. doi: 10.1210/en.2014-1121.
    1. Briellmann RS, Kalnins RM, Berkovic SF, Jackson GD. Hippocampal pathology in refractory temporal lobe epilepsy: T2-weighted signal change reflects dentate gliosis. Neurology. 2002;58:265–271. doi: 10.1212/WNL.58.2.265.
    1. Callen DJ, Black SE, Gao F, Caldwell CB, Szalai JP. Beyond the hippocampus: MRI volumetry confirms widespread limbic atrophy in AD. Neurology. 2001;57:1669–1674. doi: 10.1212/WNL.57.9.1669.
    1. Ishii M, Iadecola C. Metabolic and non-cognitive manifestations of Alzheimer’s disease: the hypothalamus as both culprit and target of pathology. Cell Metab. 2015;22:761–776. doi: 10.1016/j.cmet.2015.08.016.
    1. Loskutova N, Honea RA, Brooks WM, Burns JM. Reduced limbic and hypothalamic volumes correlate with bone density in early Alzheimer’s disease. J. Alzheimer’s Dis. 2010;20:313–322. doi: 10.3233/JAD-2010-1364.
    1. Iwahara N, et al. Role of suppressor of cytokine signaling 3 (SOCS3) in altering activated microglia phenotype in APPswe/PS1dE9 mice. J. Alzheimer’s Dis. 2017;55:1235–1247. doi: 10.3233/JAD-160887.
    1. Gjerum L, et al. A visual rating scale for cingulate island sign on 18F-FDG-PET to differentiate dementia with Lewy bodies and Alzheimer’s disease. J. Neurological Sci. 2019;410:116645. doi: 10.1016/j.jns.2019.116645.
    1. Wong D, et al. Reduced hippocampal glutamate and posterior cingulate N-acetyl aspartate in mild cognitive impairment and Alzheimer’s disease is associated with episodic memory performance and white matter integrity in the cingulum: a pilot study. J. Alzheimer’s Dis. 2020;73:1385–1405. doi: 10.3233/JAD-190773.
    1. Ferreira ST, Klein WL. The Abeta oligomer hypothesis for synapse failure and memory loss in Alzheimer’s disease. Neurobiol. Learn. Mem. 2011;96:529–543. doi: 10.1016/j.nlm.2011.08.003.
    1. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356. doi: 10.1126/science.1072994.
    1. Sukoff Rizzo SJ, et al. Evidence for sustained elevation of IL-6 in the CNS as a key contributor of depressive-like phenotypes. Transl. Psychiatry. 2012;2:e199. doi: 10.1038/tp.2012.120.
    1. Chiba T, et al. Amyloid-beta causes memory impairment by disturbing the JAK2/STAT3 axis in hippocampal neurons. Mol. Psychiatry. 2009;14:206–222. doi: 10.1038/mp.2008.105.
    1. Ott A, et al. Association of diabetes mellitus and dementia: the Rotterdam Study. Diabetologia. 1996;39:1392–1397. doi: 10.1007/s001250050588.
    1. Ott A, et al. Diabetes mellitus and the risk of dementia: the Rotterdam Study. Neurology. 1999;53:1937–1942. doi: 10.1212/WNL.53.9.1937.
    1. Whitmer RA, et al. Central obesity and increased risk of dementia more than three decades later. Neurology. 2008;71:1057–1064. doi: 10.1212/01.wnl.0000306313.89165.ef.
    1. Spilt A, et al. Not all age-related white matter hyperintensities are the same: a magnetization transfer imaging study. AJNR Am. J. Neuroradiol. 2006;27:1964–1968.
    1. Blessing EM, Beissner F, Schumann A, Brunner F, Bar KJ. A data-driven approach to mapping cortical and subcortical intrinsic functional connectivity along the longitudinal hippocampal axis. Hum. brain Mapp. 2016;37:462–476. doi: 10.1002/hbm.23042.
    1. Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991;82:239–259. doi: 10.1007/BF00308809.
    1. Ogomori K, et al. Beta-protein amyloid is widely distributed in the central nervous system of patients with Alzheimer’s disease. Am. J. Pathol. 1989;134:243–251.
    1. Standaert DG, Lee VM, Greenberg BD, Lowery DE, Trojanowski JQ. Molecular features of hypothalamic plaques in Alzheimer’s disease. Am. J. Pathol. 1991;139:681–691.
    1. Fronczek R, et al. Hypocretin (orexin) loss in Alzheimer’s disease. Neurobiol. aging. 2012;33:1642–1650. doi: 10.1016/j.neurobiolaging.2011.03.014.
    1. Musiek ES, Xiong DD, Holtzman DM. Sleep, circadian rhythms, and the pathogenesis of Alzheimer disease. Exp. Mol. Med. 2015;47:e148. doi: 10.1038/emm.2014.121.
    1. Kincheski GC, et al. Chronic sleep restriction promotes brain inflammation and synapse loss, and potentiates memory impairment induced by amyloid-beta oligomers in mice. Brain, Behav., Immun. 2017;64:140–151. doi: 10.1016/j.bbi.2017.04.007.
    1. Balschun D, et al. Interleukin-6: a cytokine to forget. FASEB J.: . Publ. Federation Am. Societies Exp. Biol. 2004;18:1788–1790. doi: 10.1096/fj.04-1625fje.
    1. Song DK, et al. Central beta-amyloid peptide-induced peripheral interleukin-6 responses in mice. J. Neurochem. 2001;76:1326–1335. doi: 10.1046/j.1471-4159.2001.00121.x.
    1. Craft S, Zallen G, Baker LD. Glucose and memory in mild senile dementia of the Alzheimer type. J. Clin. Exp. Neuropsychol. 1992;14:253–267. doi: 10.1080/01688639208402827.
    1. Wright CB, et al. Interleukin-6 is associated with cognitive function: the Northern Manhattan Study. J. Stroke Cerebrovasc. Dis.: . J. Natl Stroke Assoc. 2006;15:34–38. doi: 10.1016/j.jstrokecerebrovasdis.2005.08.009.
    1. Pavlov VA, Tracey KJ. The vagus nerve and the inflammatory reflex–linking immunity and metabolism. Nat. Rev. Endocrinol. 2012;8:743–754. doi: 10.1038/nrendo.2012.189.
    1. Gyengesi E, et al. Chronic microglial activation in the GFAP-IL6 mouse contributes to age-dependent cerebellar volume loss and impairment in motor function. Front. Neurosci. 2019;13:303. doi: 10.3389/fnins.2019.00303.
    1. Ledo JH, et al. Amyloid-beta oligomers link depressive-like behavior and cognitive deficits in mice. Mol. psychiatry. 2013;18:1053–1054. doi: 10.1038/mp.2012.168.
    1. Chakrabarty P, et al. Massive gliosis induced by interleukin-6 suppresses Abeta deposition in vivo: evidence against inflammation as a driving force for amyloid deposition. FASEB. J.: . Publ. Federation Am. Societies Exp. Biol. 2010;24:548–559.
    1. Forny-Germano L, et al. Alzheimer’s disease-like pathology induced by amyloid-beta oligomers in nonhuman primates. J. Neurosci.: . J. Soc. Neurosci. 2014;34:13629–13643. doi: 10.1523/JNEUROSCI.1353-14.2014.
    1. Bomfim TR, et al. An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer’s disease-associated Abeta oligomers. J. Clin. Investig. 2012;122:1339–1353. doi: 10.1172/JCI57256.
    1. Lyra ESNM, et al. Understanding the link between insulin resistance and Alzheimer’s disease: Insights from animal models. Exp. Neurol. 2019;316:1–11. doi: 10.1016/j.expneurol.2019.03.016.
    1. Mao YF, et al. Intranasal insulin alleviates cognitive deficits and amyloid pathology in young adult APPswe/PS1dE9 mice. Aging cell. 2016;15:893–902. doi: 10.1111/acel.12498.
    1. Talbot K, 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. Investig. 2012;122:1316–1338. doi: 10.1172/JCI59903.
    1. H. H. Ruiz et al. 2016 Increased susceptibility to metabolic dysregulation in a mouse model of Alzheimer’s disease is associated with impaired hypothalamic insulin signaling and elevated BCAA levels Alzheimer’s Dement.: J. Alzheimer’s Assoc. 12 851–861.
    1. Marciniak E, et al. Tau deletion promotes brain insulin resistance. J. Exp. Med. 2017;214:2257–2269. doi: 10.1084/jem.20161731.
    1. Rui L, Yuan M, Frantz D, Shoelson S, White MF. SOCS-1 and SOCS-3 block insulin signaling by ubiquitin-mediated degradation of IRS1 and IRS2. J. Biol. Chem. 2002;277:42394–42398. doi: 10.1074/jbc.C200444200.
    1. Jorgensen SB, et al. Deletion of skeletal muscle SOCS3 prevents insulin resistance in obesity. Diabetes. 2013;62:56–64. doi: 10.2337/db12-0443.
    1. Pedroso JA, et al. Inactivation of SOCS3 in leptin receptor-expressing cells protects mice from diet-induced insulin resistance but does not prevent obesity. Mol. Metab. 2014;3:608–618. doi: 10.1016/j.molmet.2014.06.001.
    1. Hashioka S, Klegeris A, Qing H, McGeer PL. STAT3 inhibitors attenuate interferon-gamma-induced neurotoxicity and inflammatory molecule production by human astrocytes. Neurobiol. Dis. 2011;41:299–307. doi: 10.1016/j.nbd.2010.09.018.
    1. Reichenbach N, et al. Inhibition of Stat3-mediated astrogliosis ameliorates pathology in an Alzheimer’s disease model. EMBO Mol. Med. 2019;11:e9665. doi: 10.15252/emmm.201809665.
    1. Nicolas CS, et al. The Jak/STAT pathway is involved in synaptic plasticity. Neuron. 2012;73:374–390. doi: 10.1016/j.neuron.2011.11.024.
    1. Chen E, et al. A novel role of the STAT3 pathway in brain inflammation-induced human neural progenitor cell differentiation. Curr. Mol. Med. 2013;13:1474–1484. doi: 10.2174/15665240113139990076.
    1. Saxe MD, et al. Ablation of hippocampal neurogenesis impairs contextual fear conditioning and synaptic plasticity in the dentate gyrus. Proc. Natl Acad. Sci. USA. 2006;103:17501–17506. doi: 10.1073/pnas.0607207103.
    1. Greco SJ, et al. Leptin reduces pathology and improves memory in a transgenic mouse model of Alzheimer’s disease. J. Alzheimer’s Dis. 2010;19:1155–1167. doi: 10.3233/JAD-2010-1308.
    1. Bell RD, Zlokovic BV. Neurovascular mechanisms and blood-brain barrier disorder in Alzheimer’s disease. Acta Neuropathologica. 2009;118:103–113. doi: 10.1007/s00401-009-0522-3.
    1. Thal DR, Griffin WS, de Vos RA, Ghebremedhin E. Cerebral amyloid angiopathy and its relationship to Alzheimer’s disease. Acta Neuropathologica. 2008;115:599–609. doi: 10.1007/s00401-008-0366-2.
    1. Starr JM, et al. Increased blood-brain barrier permeability in type II diabetes demonstrated by gadolinium magnetic resonance imaging. J. Neurol. Neurosurg. Psychiatry. 2003;74:70–76. doi: 10.1136/jnnp.74.1.70.
    1. Stevens MJ, Feldman EL, Greene DA. The aetiology of diabetic neuropathy: the combined roles of metabolic and vascular defects. Diabet. Med.: J. Br. Diabet. Assoc. 1995;12:566–579. doi: 10.1111/j.1464-5491.1995.tb00544.x.
    1. Ting EY, Yang AC, Tsai SJ. Role of Interleukin-6 in depressive disorder. Int. J. Mol. Sci. 2020;21:2194. doi: 10.3390/ijms21062194.
    1. Ownby RL, Crocco E, Acevedo A, John V, Loewenstein D. Depression and risk for Alzheimer disease: systematic review, meta-analysis, and metaregression analysis. Arch. Gen. Psychiatry. 2006;63:530–538. doi: 10.1001/archpsyc.63.5.530.
    1. Butchart J, et al. Etanercept in Alzheimer disease: a randomized, placebo-controlled, double-blind, phase 2 trial. Neurology. 2015;84:2161–2168. doi: 10.1212/WNL.0000000000001617.
    1. Miguel-Alvarez M, et al. Non-steroidal anti-inflammatory drugs as a treatment for Alzheimer’s disease: a systematic review and meta-analysis of treatment effect. Drugs Aging. 2015;32:139–147. doi: 10.1007/s40266-015-0239-z.

Source: PubMed

3
Abonneren