Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2015

Open Access 01-12-2015 | Research

Insulin improves memory and reduces chronic neuroinflammation in the hippocampus of young but not aged brains

Authors: Linda Adzovic, Ashley E Lynn, Heather M D’Angelo, Alexis M Crockett, Roxanne M Kaercher, Sarah E Royer, Sarah C Hopp, Gary L Wenk

Published in: Journal of Neuroinflammation | Issue 1/2015

Login to get access

Abstract

The role of insulin in the brain is still not completely understood. In the periphery, insulin can decrease inflammation induced by lipopolysaccharide (LPS); however, whether insulin can reduce inflammation within the brain is unknown. Experiments administrating intranasal insulin to young and aged adults have shown that insulin improves memory. In our animal model of chronic neuroinflammation, we administered insulin and/or LPS directly into the brain via the fourth ventricle for 4 weeks in young rats; we then analyzed their spatial memory and neuroinflammatory response. Additionally, we administered insulin or artificial cerebral spinal fluid (aCSF), in the same manner, to aged rats and then analyzed their spatial memory and neuroinflammatory response. Response to chronic neuroinflammation in young rats was analyzed in the presence or absence of insulin supplementation. Here, we show for the first time that insulin infused (i.c.v.) to young rats significantly attenuated the effects of LPS by decreasing the expression of neuroinflammatory markers in the hippocampus and by improving performance in the Morris water pool task. In young rats, insulin infusion alone significantly improved their performance as compared to all other groups. Unexpectedly, in aged rats, the responsiveness to insulin was completely absent, that is, spatial memory was still impaired suggesting that an age-dependent insulin resistance may contribute to the cognitive impairment observed in neurodegenerative diseases. Our data suggest a novel therapeutic effect of insulin on neuroinflammation in the young but not the aged brain.
Literature
2.
go back to reference Lynch AM, Lynch MA. The age-related increase in IL-1 type I receptor in rat hippocampus is coupled with an increase in caspase-3 activation. Eur J Neurosci. 2002;15:1779–88.CrossRefPubMed Lynch AM, Lynch MA. The age-related increase in IL-1 type I receptor in rat hippocampus is coupled with an increase in caspase-3 activation. Eur J Neurosci. 2002;15:1779–88.CrossRefPubMed
3.
go back to reference Campuzano O, Castillo-Ruiz MM, Acarin L, Castellano B, Gonzalez B. Increased levels of proinflammatory cytokines in the aged rat brain attenuate injury-induced cytokine response after excitotoxic damage. J Neurosci Res. 2009;87:2484–97.CrossRefPubMed Campuzano O, Castillo-Ruiz MM, Acarin L, Castellano B, Gonzalez B. Increased levels of proinflammatory cytokines in the aged rat brain attenuate injury-induced cytokine response after excitotoxic damage. J Neurosci Res. 2009;87:2484–97.CrossRefPubMed
4.
go back to reference Ye SM, Johnson RW. Increased interleukin-6 expression by microglia from brain of aged mice. J Neuroimmunol. 1999;9:139–48.CrossRef Ye SM, Johnson RW. Increased interleukin-6 expression by microglia from brain of aged mice. J Neuroimmunol. 1999;9:139–48.CrossRef
5.
go back to reference Colton C, Wilcock DM. Assessing activation states in microglia. CNS Neurol Disord Drug Targets. 2010;9:174–91.CrossRefPubMed Colton C, Wilcock DM. Assessing activation states in microglia. CNS Neurol Disord Drug Targets. 2010;9:174–91.CrossRefPubMed
6.
go back to reference Smith JA, Das A, Ray SK, Banik NL. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 2012;87:10–20.CrossRefPubMed Smith JA, Das A, Ray SK, Banik NL. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull. 2012;87:10–20.CrossRefPubMed
7.
go back to reference Wenk GL, Hauss-Wegrzyniak B. Animal models of chronic neuroinflammation as a model of Alzheimer’s disease. In: Bondy S, Campbell A, editors. Inflammatory events in neurodegeneration. Scottsdale, AZ: Prominent Press; 2001. p. 83–7. Wenk GL, Hauss-Wegrzyniak B. Animal models of chronic neuroinflammation as a model of Alzheimer’s disease. In: Bondy S, Campbell A, editors. Inflammatory events in neurodegeneration. Scottsdale, AZ: Prominent Press; 2001. p. 83–7.
8.
go back to reference Bilbo SD. Early-life infection is a vulnerability factor for aging-related glial alterations and cognitive decline. Neurobiol Learn Mem. 2010;94:57–64.PubMedCentralCrossRefPubMed Bilbo SD. Early-life infection is a vulnerability factor for aging-related glial alterations and cognitive decline. Neurobiol Learn Mem. 2010;94:57–64.PubMedCentralCrossRefPubMed
9.
go back to reference Eikelenboom P, van Exel E, Hoozemans JJ, Veerhuis R, Rozemuller AJ, van Gool WA. Neuroinflammation - an early event in both the history and pathogenesis of Alzheimer’s disease. Neurodegener Dis. 2010;7:38–41.CrossRefPubMed Eikelenboom P, van Exel E, Hoozemans JJ, Veerhuis R, Rozemuller AJ, van Gool WA. Neuroinflammation - an early event in both the history and pathogenesis of Alzheimer’s disease. Neurodegener Dis. 2010;7:38–41.CrossRefPubMed
10.
go back to reference Heneka MT, Nadrigny F, Regen T, Martinez-Hernandez A, Dumitrescu-Ozimek L, Terwel D, et al. Locus ceruleus controls Alzheimer’s disease pathology by modulating microglial functions through norepinephrine. Proc Natl Acad Sci U S A. 2010;107:6058–63.PubMedCentralCrossRefPubMed Heneka MT, Nadrigny F, Regen T, Martinez-Hernandez A, Dumitrescu-Ozimek L, Terwel D, et al. Locus ceruleus controls Alzheimer’s disease pathology by modulating microglial functions through norepinephrine. Proc Natl Acad Sci U S A. 2010;107:6058–63.PubMedCentralCrossRefPubMed
11.
go back to reference Watson GS, Craft S. Modulation of memory by insulin and glucose: neuropsychological observations in Alzheimer’s disease. Eur J Pharmacol. 2004;490:97–113.CrossRefPubMed Watson GS, Craft S. Modulation of memory by insulin and glucose: neuropsychological observations in Alzheimer’s disease. Eur J Pharmacol. 2004;490:97–113.CrossRefPubMed
12.
go back to reference Arnold SE, Arvanitakis Z, Bakshi KP, Bennett DA, Fuino RL, Han LY. 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;122:1316–38.PubMedCentralCrossRefPubMed Arnold SE, Arvanitakis Z, Bakshi KP, Bennett DA, Fuino RL, Han LY. 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;122:1316–38.PubMedCentralCrossRefPubMed
13.
go back to reference Najem D, Bamji-Mirza M, Chang N, Liu QY, Zhang W. Insulin resistance, neuroinflammation, and Alzheimer’s disease. Rev Neurosci. 2014;25:509–25.CrossRefPubMed Najem D, Bamji-Mirza M, Chang N, Liu QY, Zhang W. Insulin resistance, neuroinflammation, and Alzheimer’s disease. Rev Neurosci. 2014;25:509–25.CrossRefPubMed
14.
go back to reference Jeschke MG, Klein D, Bolder U, Einspanier R. Insulin attenuates the systemic inflammatory response in endotoxemic rats. Endocrinol. 2004;145:4084–93.CrossRef Jeschke MG, Klein D, Bolder U, Einspanier R. Insulin attenuates the systemic inflammatory response in endotoxemic rats. Endocrinol. 2004;145:4084–93.CrossRef
15.
go back to reference Dandona P, Aljada A, Mohanty P, Ghanim H, Hamouda W, Assian E, et al. Insulin inhibits intranuclear nuclear factor κB and stimulates IκB in mononuclear cells in obese subjects: evidence for an anti-inflammatory effect? J Clin Endocrinol Metab. 2001;86:3257–65.PubMed Dandona P, Aljada A, Mohanty P, Ghanim H, Hamouda W, Assian E, et al. Insulin inhibits intranuclear nuclear factor κB and stimulates IκB in mononuclear cells in obese subjects: evidence for an anti-inflammatory effect? J Clin Endocrinol Metab. 2001;86:3257–65.PubMed
16.
go back to reference Leverence JT, Medhora M, Konduri GG, Sampath V. Lipopolysaccharide-induced cytokine expression in alveolar epithelial cells: role of PKCζ-mediated p47phox phosphorylation. Chemico-Biol Inter. 2011;189:72–81.CrossRef Leverence JT, Medhora M, Konduri GG, Sampath V. Lipopolysaccharide-induced cytokine expression in alveolar epithelial cells: role of PKCζ-mediated p47phox phosphorylation. Chemico-Biol Inter. 2011;189:72–81.CrossRef
17.
go back to reference Dallot E, Méhats C, Oger S, Leroy MJ, Breuiller-Fouché MA. Role for PKCζ in the LPS-induced translocation NF-κB p65 subunit in cultured myometrial cells. Biochimie. 2005;87:513–21.CrossRefPubMed Dallot E, Méhats C, Oger S, Leroy MJ, Breuiller-Fouché MA. Role for PKCζ in the LPS-induced translocation NF-κB p65 subunit in cultured myometrial cells. Biochimie. 2005;87:513–21.CrossRefPubMed
18.
go back to reference Li H, Liu B, Huang J, Chen H, Guo X, Yuan Z. Insulin inhibits lipopolysaccharide-induced nitric oxide synthase expression in rat primary astrocytes. Brain Res. 2013;1506:1–11.CrossRefPubMed Li H, Liu B, Huang J, Chen H, Guo X, Yuan Z. Insulin inhibits lipopolysaccharide-induced nitric oxide synthase expression in rat primary astrocytes. Brain Res. 2013;1506:1–11.CrossRefPubMed
19.
go back to reference Martins JO, Ferracini M, Ravanelli N, Landgraf RG, Jancar S. Insulin suppresses LPS-induced iNOS and COX-2 expression and NF-κB activation in alveolar macrophages. Cell Physiol Biochem. 2008;22:279–86.CrossRefPubMed Martins JO, Ferracini M, Ravanelli N, Landgraf RG, Jancar S. Insulin suppresses LPS-induced iNOS and COX-2 expression and NF-κB activation in alveolar macrophages. Cell Physiol Biochem. 2008;22:279–86.CrossRefPubMed
20.
go back to reference Huang XH, Doerner AM, Pan WW, Smith L, Huang S, Papdimor TJ, et al. An atypical protein kinase C (PKCζ) plays a critical role in lipopolysaccharide-activated NF-κB in human peripheral blood monocytes and macrophages. J Immunol. 2009;182:5810–5.CrossRefPubMed Huang XH, Doerner AM, Pan WW, Smith L, Huang S, Papdimor TJ, et al. An atypical protein kinase C (PKCζ) plays a critical role in lipopolysaccharide-activated NF-κB in human peripheral blood monocytes and macrophages. J Immunol. 2009;182:5810–5.CrossRefPubMed
21.
go back to reference Aye IL, Jansson T, Powell TL. Interleukin-1β inhibits insulin signaling and prevents insulin-stimulated system amino acid transport in primary human trophoblasts. Mol Cell Endocrinol. 2013;381:46–55.PubMedCentralCrossRefPubMed Aye IL, Jansson T, Powell TL. Interleukin-1β inhibits insulin signaling and prevents insulin-stimulated system amino acid transport in primary human trophoblasts. Mol Cell Endocrinol. 2013;381:46–55.PubMedCentralCrossRefPubMed
22.
go back to reference Gual P, Le Marchand-Brustel Y, Tanti JF. Positive and negative regulation of insulin signaling through IRS-1 phosphorylation. Biochimie. 2005;87:99–109.CrossRefPubMed Gual P, Le Marchand-Brustel Y, Tanti JF. Positive and negative regulation of insulin signaling through IRS-1 phosphorylation. Biochimie. 2005;87:99–109.CrossRefPubMed
23.
24.
go back to reference Balakrishnan K, Verdile G, Mehta PD, Beilby J, Nolan D, Galvao DA, et al. Plasma Aβ42 correlates positively with increased body fat in healthy individuals. J Alz Dis. 2005;8:269–82. Balakrishnan K, Verdile G, Mehta PD, Beilby J, Nolan D, Galvao DA, et al. Plasma Aβ42 correlates positively with increased body fat in healthy individuals. J Alz Dis. 2005;8:269–82.
25.
go back to reference Biessels G, Kappelle LJ. Increased risk of Alzheimer’s disease in type II diabetes: insulin resistance of the brain or insulin-induced amyloid pathology? Biochem Soc Trans. 2005;33:1041–4.CrossRefPubMed Biessels G, Kappelle LJ. Increased risk of Alzheimer’s disease in type II diabetes: insulin resistance of the brain or insulin-induced amyloid pathology? Biochem Soc Trans. 2005;33:1041–4.CrossRefPubMed
26.
go back to reference Casserly I, Topol E. Convergence of atherosclerosis and Alzheimer’s disease: inflammation, cholesterol, and misfolded proteins. Lancet. 2004;363:1139–46.CrossRefPubMed Casserly I, Topol E. Convergence of atherosclerosis and Alzheimer’s disease: inflammation, cholesterol, and misfolded proteins. Lancet. 2004;363:1139–46.CrossRefPubMed
27.
28.
go back to reference Wang KC, Woung LC, Tsai MT, Liu CC, Su YH, Li CY. Risk of Alzheimer’s disease in relation to diabetes: a population-based cohort study. Neuroepidemiol. 2012;38:237–44.CrossRef Wang KC, Woung LC, Tsai MT, Liu CC, Su YH, Li CY. Risk of Alzheimer’s disease in relation to diabetes: a population-based cohort study. Neuroepidemiol. 2012;38:237–44.CrossRef
29.
go back to reference Havrankova J, Roth J, Brownstein M. Insulin receptors are widely distributed in the central nervous system of the rat. Nature. 1978;272:827–9.CrossRefPubMed Havrankova J, Roth J, Brownstein M. Insulin receptors are widely distributed in the central nervous system of the rat. Nature. 1978;272:827–9.CrossRefPubMed
30.
go back to reference Zhao WQ, Alkon DL. Role of insulin and insulin receptor in learning and memory. Mol Cell Endocrinol. 2001;177:125–34.CrossRefPubMed Zhao WQ, Alkon DL. Role of insulin and insulin receptor in learning and memory. Mol Cell Endocrinol. 2001;177:125–34.CrossRefPubMed
31.
go back to reference Clarke DW, Mudd L, Boyd Jr FT. Insulin is released from rat brain neuronal cells in culture. J Neurochem. 1986;47:831–6.CrossRefPubMed Clarke DW, Mudd L, Boyd Jr FT. Insulin is released from rat brain neuronal cells in culture. J Neurochem. 1986;47:831–6.CrossRefPubMed
32.
go back to reference Valenciano AI, Corrochano S, de Pablo F, de la Villa P, de la Rosa EJ. Proinsulin/insulin is synthesized locally and prevents caspase- and cathepsin-mediated cell death in the embryonic mouse retina. J Neurochem. 2006;99:524–36.CrossRefPubMed Valenciano AI, Corrochano S, de Pablo F, de la Villa P, de la Rosa EJ. Proinsulin/insulin is synthesized locally and prevents caspase- and cathepsin-mediated cell death in the embryonic mouse retina. J Neurochem. 2006;99:524–36.CrossRefPubMed
33.
go back to reference Dou JT, Chen M, Dufour F, Alkon DL, Zhao WQ. Insulin receptor signaling in long-term memory consolidation following spatial learning. Learn Mem. 2005;12:646–55.PubMedCentralCrossRefPubMed Dou JT, Chen M, Dufour F, Alkon DL, Zhao WQ. Insulin receptor signaling in long-term memory consolidation following spatial learning. Learn Mem. 2005;12:646–55.PubMedCentralCrossRefPubMed
34.
go back to reference Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci. 2002;5:514–6.CrossRefPubMed Born J, Lange T, Kern W, McGregor GP, Bickel U, Fehm HL. Sniffing neuropeptides: a transnasal approach to the human brain. Nat Neurosci. 2002;5:514–6.CrossRefPubMed
35.
go back to reference Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born J, et al. Intranasal insulin improves memory in humans. Psychoneuroendocrinol. 2004;29:1326–34.CrossRef Benedict C, Hallschmid M, Hatke A, Schultes B, Fehm HL, Born J, et al. Intranasal insulin improves memory in humans. Psychoneuroendocrinol. 2004;29:1326–34.CrossRef
36.
go back to reference Reger MA, Watson GS, Green PS, Wilkinson CW, Baker LD, Cholerton B, et al. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurol. 2008;70:440–8.CrossRef Reger MA, Watson GS, Green PS, Wilkinson CW, Baker LD, Cholerton B, et al. Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurol. 2008;70:440–8.CrossRef
37.
go back to reference Adzovic L, Domenici L. Insulin induces phosphorylation of the AMPA receptor subunit GluR1, reversed by ZIP, and overexpression of protein kinase M zeta, reversed by amyloid beta. J Neurochem. 2014;31:582–7.CrossRef Adzovic L, Domenici L. Insulin induces phosphorylation of the AMPA receptor subunit GluR1, reversed by ZIP, and overexpression of protein kinase M zeta, reversed by amyloid beta. J Neurochem. 2014;31:582–7.CrossRef
38.
go back to reference Kamal A, Ramakers GM, Gispen WH, Biessels GJ. Effect of chronic intracerebroventricular insulin administration in rats on the peripheral glucose metabolism and synaptic plasticity of CA1 hippocampal neurons. Brain Res. 2012;1435:99–104.CrossRefPubMed Kamal A, Ramakers GM, Gispen WH, Biessels GJ. Effect of chronic intracerebroventricular insulin administration in rats on the peripheral glucose metabolism and synaptic plasticity of CA1 hippocampal neurons. Brain Res. 2012;1435:99–104.CrossRefPubMed
39.
go back to reference Marchalant Y, Cerbai F, Brothers H, Wenk GL. Cannabinoid receptor stimulation is anti-inflammatory and improves memory in old rats. Neurobiol Aging. 2008;29:1894–901.PubMedCentralCrossRefPubMed Marchalant Y, Cerbai F, Brothers H, Wenk GL. Cannabinoid receptor stimulation is anti-inflammatory and improves memory in old rats. Neurobiol Aging. 2008;29:1894–901.PubMedCentralCrossRefPubMed
40.
go back to reference Marchalant Y, Brothers HM, Norman GJ, Karolina K, DeVries C, Wenk GL. Cannabinoids attenuate the effects of aging upon neuroinflammation and neurogenesis. Neurobiol Dis. 2009;34:300–7.CrossRefPubMed Marchalant Y, Brothers HM, Norman GJ, Karolina K, DeVries C, Wenk GL. Cannabinoids attenuate the effects of aging upon neuroinflammation and neurogenesis. Neurobiol Dis. 2009;34:300–7.CrossRefPubMed
41.
go back to reference Hauss-Wegrzyniak B, Dobrzanski P, Stoehr JD, Wenk GL. Chronic neuroinflammation in rats reproduces components of the neurobiology of Alzheimer’s disease. Brain Res. 1998;780:294–303.CrossRefPubMed Hauss-Wegrzyniak B, Dobrzanski P, Stoehr JD, Wenk GL. Chronic neuroinflammation in rats reproduces components of the neurobiology of Alzheimer’s disease. Brain Res. 1998;780:294–303.CrossRefPubMed
42.
go back to reference Hauss-Wegrzyniak B, Galons JP, Wenk GL. Quantitative volumetric analysis of brain magnetic resonance imaging from rat with chronic neuroinflammation and correlation with histology. Exp Neurol. 2000;165:347–54.CrossRefPubMed Hauss-Wegrzyniak B, Galons JP, Wenk GL. Quantitative volumetric analysis of brain magnetic resonance imaging from rat with chronic neuroinflammation and correlation with histology. Exp Neurol. 2000;165:347–54.CrossRefPubMed
43.
go back to reference Hauss-Wegrzyniak B, Lynch MA, Vraniak PD, Wenk GL. Chronic brain inflammation results in cell loss in the entorhinal cortex and impaired LTP in perforant path-granule cell synapses. Exp Neurol. 2002;341:336–41.CrossRef Hauss-Wegrzyniak B, Lynch MA, Vraniak PD, Wenk GL. Chronic brain inflammation results in cell loss in the entorhinal cortex and impaired LTP in perforant path-granule cell synapses. Exp Neurol. 2002;341:336–41.CrossRef
44.
go back to reference Rosi S, Ramirez-Amaya V, Hauss-Wegrzyniak B, Wenk GL. Chronic brain inflammation leads to a decline in hippocampal NMDA R1 receptors. J Neuroinflam. 2004;1:12–8.CrossRef Rosi S, Ramirez-Amaya V, Hauss-Wegrzyniak B, Wenk GL. Chronic brain inflammation leads to a decline in hippocampal NMDA R1 receptors. J Neuroinflam. 2004;1:12–8.CrossRef
45.
go back to reference Rosi S, Ramirez-Amaya V, Vazdarjanova A, Esperanza E, Larkin P, Fike JR, et al. Accuracy of hippocampal network activity is disrupted by neuroinflammation: rescue by memantine. Brain. 2009;132:2464–86.PubMedCentralCrossRefPubMed Rosi S, Ramirez-Amaya V, Vazdarjanova A, Esperanza E, Larkin P, Fike JR, et al. Accuracy of hippocampal network activity is disrupted by neuroinflammation: rescue by memantine. Brain. 2009;132:2464–86.PubMedCentralCrossRefPubMed
46.
go back to reference Rosi S, Ramirez-Amaya V, Vazdarjanova A, Worley PF, Barnes CA, Wenk GL. Neuroinflammation alters the hippocampal pattern of behaviorally induced Arc expression. J Neurosci. 2005;25:723–31.CrossRefPubMed Rosi S, Ramirez-Amaya V, Vazdarjanova A, Worley PF, Barnes CA, Wenk GL. Neuroinflammation alters the hippocampal pattern of behaviorally induced Arc expression. J Neurosci. 2005;25:723–31.CrossRefPubMed
47.
go back to reference Wenk GL, Hauss-Wegrzyniak B, Willard LB. Pathological and biochemical studies of chronic neuroinflammation may lead to therapies for Alzheimer’s disease. In: Patterson P, Kordon C, Christen Y, editors. Research and perspectives in neurosciences: neuro-immune neurodegenerative and psychiatric disorders and neural injury. Heidelberg: Springer-Verlag; 2000. p. 73–7. Wenk GL, Hauss-Wegrzyniak B, Willard LB. Pathological and biochemical studies of chronic neuroinflammation may lead to therapies for Alzheimer’s disease. In: Patterson P, Kordon C, Christen Y, editors. Research and perspectives in neurosciences: neuro-immune neurodegenerative and psychiatric disorders and neural injury. Heidelberg: Springer-Verlag; 2000. p. 73–7.
48.
go back to reference Marchalant Y, Rosi S, Wenk GL. Anti-inflammatory property of the cannabinoid agonist WIN-55212-2 in a rodent model of chronic brain inflammation. Neurosci. 2007;144:1516–22.CrossRef Marchalant Y, Rosi S, Wenk GL. Anti-inflammatory property of the cannabinoid agonist WIN-55212-2 in a rodent model of chronic brain inflammation. Neurosci. 2007;144:1516–22.CrossRef
49.
go back to reference Figlewicz DP, Bennett J, Evans SB, Kaiyala K, Sipols AJ, Benoit SC. Intraventricular insulin and leptin reverse place preference conditioned with high-fat diet in rats. Behav Neurosci. 2004;118:479–87.CrossRefPubMed Figlewicz DP, Bennett J, Evans SB, Kaiyala K, Sipols AJ, Benoit SC. Intraventricular insulin and leptin reverse place preference conditioned with high-fat diet in rats. Behav Neurosci. 2004;118:479–87.CrossRefPubMed
50.
go back to reference Bardou I, Brothers HM, Kaercher RM, Hopp SC, Wenk GL. Differential effects of duration and age upon the consequences of neuroinflammation in the hippocampus. Neurobiol Aging. 2013;34:2293–301.PubMedCentralCrossRefPubMed Bardou I, Brothers HM, Kaercher RM, Hopp SC, Wenk GL. Differential effects of duration and age upon the consequences of neuroinflammation in the hippocampus. Neurobiol Aging. 2013;34:2293–301.PubMedCentralCrossRefPubMed
51.
go back to reference Martins JO, Ferracini M, Ravanelli N, Landgraf RG, Jancar S. Insulin inhibits LPS-induced signaling pathways in alveolar macrophages. Cell Physiol Biochem. 2007;21:297–304.CrossRef Martins JO, Ferracini M, Ravanelli N, Landgraf RG, Jancar S. Insulin inhibits LPS-induced signaling pathways in alveolar macrophages. Cell Physiol Biochem. 2007;21:297–304.CrossRef
52.
go back to reference Bamji-Mirza M, Callaghan D, Najem D, Shen S, Hasim MS, Yang Z, et al. Stimulation of insulin signaling and inhibition of JNK-AP1 activation protect cells from amyloid-β-induced signaling dysregulation and inflammatory response. J Alz Dis. 2014;40:105–22. Bamji-Mirza M, Callaghan D, Najem D, Shen S, Hasim MS, Yang Z, et al. Stimulation of insulin signaling and inhibition of JNK-AP1 activation protect cells from amyloid-β-induced signaling dysregulation and inflammatory response. J Alz Dis. 2014;40:105–22.
53.
go back to reference Brothers HM, Bardou I, Hopp SC, Kaercher RM, Corona AW, Fenn AM, et al. Riluzole partially rescues age-associated, but not LPS-induced, loss of glutamate transporters and spatial memory. J Neuroimm Pharmacol. 2013;8:1098–105.CrossRef Brothers HM, Bardou I, Hopp SC, Kaercher RM, Corona AW, Fenn AM, et al. Riluzole partially rescues age-associated, but not LPS-induced, loss of glutamate transporters and spatial memory. J Neuroimm Pharmacol. 2013;8:1098–105.CrossRef
54.
go back to reference Leitges M, Sanz L, Martin P, Duran A, Braun U, García JF, et al. Targeted disruption of the zeta PKC gene results in the impairment of the NF-κB pathway. Mol Cell. 2001;8:771–80.CrossRefPubMed Leitges M, Sanz L, Martin P, Duran A, Braun U, García JF, et al. Targeted disruption of the zeta PKC gene results in the impairment of the NF-κB pathway. Mol Cell. 2001;8:771–80.CrossRefPubMed
55.
go back to reference Frölich L, Blum–Degen D, Bernstein HG, Englesberger S, Humrich J, Laufer S, et al. Insulin and insulin receptors in the brain in aging and in sporadic Alzheimer’s disease. J Neural Transm. 1998;105:423–38.CrossRefPubMed Frölich L, Blum–Degen D, Bernstein HG, Englesberger S, Humrich J, Laufer S, et al. Insulin and insulin receptors in the brain in aging and in sporadic Alzheimer’s disease. J Neural Transm. 1998;105:423–38.CrossRefPubMed
56.
go back to reference Townsend M, Metha T, Selkoe DJ. Soluble A-beta inhibits specific signal transduction cascades common to the insulin receptor pathway. J Biol Chem. 2007;282:33305–12.CrossRefPubMed Townsend M, Metha T, Selkoe DJ. Soluble A-beta inhibits specific signal transduction cascades common to the insulin receptor pathway. J Biol Chem. 2007;282:33305–12.CrossRefPubMed
Metadata
Title
Insulin improves memory and reduces chronic neuroinflammation in the hippocampus of young but not aged brains
Authors
Linda Adzovic
Ashley E Lynn
Heather M D’Angelo
Alexis M Crockett
Roxanne M Kaercher
Sarah E Royer
Sarah C Hopp
Gary L Wenk
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2015
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-015-0282-z

Other articles of this Issue 1/2015

Journal of Neuroinflammation 1/2015 Go to the issue