Elsevier

Neuropharmacology

Volume 76, Part A, January 2014, Pages 57-67
Neuropharmacology

Liraglutide can reverse memory impairment, synaptic loss and reduce plaque load in aged APP/PS1 mice, a model of Alzheimer's disease

https://doi.org/10.1016/j.neuropharm.2013.08.005Get rights and content

Highlights

  • Liraglutide protected 14-month-old APP/PS1 mice in the key markers for AD.

  • The overall plaque load in the brain was reduced by 33%.

  • The chronic inflammation response was reduced by 30%.

  • Neuronal progenitor cell count in the dentate gyrus was increased by 50%.

  • Total brain APP and beta-amyloid oligomer levels were reduced.

Abstract

Type 2 diabetes is a risk factor in the development of Alzheimer's disease (AD). It has been shown that insulin signalling is desensitised in the brains of AD patients. The incretin hormone Glucagon-like peptide-1 (GLP-1) facilitates insulin signalling, and long-lasting analogues such as liraglutide (Victoza®) are on the market as type 2 diabetes treatments. We have previously shown that liraglutide improved cognitive function, reduced amyloid plaque deposition, inflammation, overall APP and oligomer levels and enhanced LTP when injected peripherally for two months in 7 month old APPswe/PS1ΔE9 (APP/PS1) mice. This showed that liraglutide has preventive effects at the early stage of AD development.

The current study investigated whether Liraglutide would have restorative effects in late-stage Alzheimer's disease in mice. Accordingly, 14-month-old APP/PS1 and littermate control mice were injected with Liraglutide (25 nmol/kg bw) ip. for 2 months. Spatial memory was improved by Liraglutide-treatment in APP/PS1 mice compared with APP/PS1 saline-treated mice. Overall plaque load was reduced by 33%, and inflammation reduced by 30%, while neuronal progenitor cell count in the dentate gyrus was increased by 50%. LTP was significantly enhanced in APP/PS1 liraglutide-treated mice compared with APP/PS1 saline mice, corroborated with increased synapse numbers in hippocampus and cortex. Total brain APP and beta-amyloid oligomer levels were reduced in Liraglutide-treated APP/PS1 mice while IDE levels were increased. These results demonstrate that Liraglutide not only has preventive properties, but also can reverse some of the key pathological hallmarks of AD. Liraglutide is now being tested in clinical trials in AD patients.

This article is part of the Special Issue entitled ‘The Synaptic Basis of Neurodegenerative Disorders’.

Introduction

Type 2 diabetes (T2DM) has been identified as a risk factor for Alzheimer disease (AD) (Hoyer, 2004, Hölscher, 2005, Craft, 2007, Perry et al., 2007). Impaired insulin signalling has been linked to cerebral degenerative processes in aged T2DM patients (Zhao et al., 2004) and AD patients (Ohara et al., 2011) and insulin receptor desensitisation is observed in the Alzheimer's disease brain (Craft, 2007, Talbot et al., 2012). The desensitisation could play a role in the development of neurodegenerative disorders, as insulin is a growth factor with neuroprotective properties (Gasparini et al., 2002, Hoyer, 2004, Holscher, 2011).

GLP-1 is an incretin hormone (Lovshin and Drucker, 2009). Currently, the GLP-1 receptor agonists exendin-4 (Exenatide, Byetta®), liraglutide (Victoza®) and lixisenatide (Lyxumia®) are approved for treatment of T2DM (Lovshin and Drucker, 2009, Wohlfart et al., 2013). These analogues are injected subcutaneously and are well tolerated.

GLP-1 also acts as a growth factor in the brain, and has been shown to induce neurite outgrowth and to protect against oxidative injury (Perry et al., 2007). Furthermore, mice that overexpress GLP-1 receptors in the hippocampus exhibited increased neurite growth and improved learning (During et al., 2003). The deletion of the GLP-1 receptor impairs learning and of developing long-term potentiation of synaptic transmission (LTP) in the hippocampus (Abbas et al., 2009). Moreover, Liraglutide and exendin-4 have been shown to reduce endogenous levels of beta-amyloid in the brain (Perry et al., 2003, McClean et al., 2011). GLP-1 analogues induce the proliferation of neuronal progenitor cells in the brains of mice (During et al., 2003, Hunter and Holscher, 2012). This induction of cell proliferation has the potential to facilitate the repair of neuronal networks in cortical tissue and could have beneficial effects in patients with AD (Greenberg and Jin, 2006). We have previously shown that injection of Val(8)GLP-1 ip. for 3 weeks rescued LTP in the hippocampus of a APP/PS1 mouse model of AD (Gengler et al., 2012). Importantly, GLP-1 and GLP-1 analogues cross the blood brain barrier (BBB) (Kastin et al., 2002, Perry and Greig, 2002, McClean et al., 2011, Gengler et al., 2012, Hunter and Holscher, 2012). Our recent work testing liraglutide in 7–9 month old APPswe/PS1ΔE9 mice demonstrated that at an intermediate stage of disease progression, when mice are beginning to display behavioural deficits, 8 weeks treatment with liraglutide ip. improved learning and memory, enhanced LTP, reduced beta amyloid plaque load and chronic inflammation, reduced total levels of APP and toxic oligomers (McClean et al., 2011). Building on this encouraging study, the current study aims to address whether liraglutide has restorative effects at a late stage of disease progression with a high amyloid plaque load, memory and synaptic failure and synaptic loss, or only when taken at an early stage to prevent further deterioration. Accordingly, 16 month-old APPswe/PS1ΔE9 and wild-type mice were tested after having been injected ip. with liraglutide for 8 weeks. We analysed the effects on memory, synaptic plasticity of hippocampus, synaptic density, plaque deposition and inflammation, neurogenesis in dentate gyrus, and levels of total APP and soluble aggregated beta amyloid.

Section snippets

Animals

APPswe/PS1ΔE9 mice with a C57Bl/6 background were obtained from the Jackson lab (http://research.jax.org/repository/alzheimers.html). Heterozygous males were bred with wild-type C57/Bl6 females bought locally (Harlan, UK). Offspring were ear punched and genotyped using PCR with primers specific for the APP-sequence (Forward “GAATTCCGACATGACTCAGG”, Reverse: “GTTCTGCTGCATCTTGGACA”). For details see Gengler et al. (2010). Mice not expressing the transgene were used as wild-type controls. Male

Object recognition memory is restored by liraglutide treatment in aged APP/PS1 mice

Path length, speed and anxiety measures in the open field, after a single administration of liraglutide (25 nmol/kg bw) or saline 30 min prior to introduction to the apparatus, were similar in liraglutide and saline-treated wild-type and APP/PS1 mice (data not shown). In an object recognition task the acute response to administration of Liraglutide or saline was assessed (30 min prior to exposure to open field on day 1 and 30 min before exposure to familiar object on day 2). A one-way ANOVA of

Discussion

The aim of this study was to assess whether liraglutide could have a restorative effect on the pathophysiology of Alzheimer's disease progression in 14–16 month-old APP/PS1 mice, given the recent findings demonstrating a preventive effect after two months liraglutide-treatment from age 7–9 months in APP/PS1 mice, in improving cognition and reducing many AD-associated biomarkers (McClean et al., 2011). The results presented here demonstrate that ten weeks liraglutide treatment results in

Acknowledgements

This project was funded by Alzheimer's Society (grant number 97) with support from The Henry Smith Charity. We thank Prof. P Flatt and Dr. VA Gault the UU diabetes research group for advice and support.

Dr. Holscher is a named inventor on a patent application by Ulster University for the use of GLP-1 analogues in neurodegenerative diseases.

References (52)

  • P.E. Schulz et al.

    Using paired-pulse facilitation to probe the mechanisms for long-term potentiation

    Journal of Physiology (Paris)

    (1995)
  • D.J. Selkoe

    Clearing the brain's amyloid cobwebs

    Neuron

    (2001)
  • M.L. Tsai et al.

    Seizures induced by GABAB-receptor blockade in early-life induced long-term GABA(B) receptor hypofunction and kindling facilitation

    Epilepsy Research

    (2008)
  • W.Q. Zhao et al.

    Insulin and the insulin receptor in experimental models of learning and memory

    European Journal of Pharmacology

    (2004)
  • W.Q. Zhao et al.

    Insulin receptor dysfunction impairs cellular clearance of neurotoxic oligomeric a{beta}

    The Journal of Biological Chemistry

    (2009)
  • L.D. Baker et al.

    Insulin resistance and Alzheimer-like reductions in regional cerebral glucose metabolism for cognitively normal adults with prediabetes or early type 2 diabetes

    Archives of Neurology

    (2010)
  • M. Blurton-Jones et al.

    Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease

    Proceedings of the National Academy of Sciences of the United States of America

    (2009)
  • T.R. Bomfim et al.

    An anti-diabetes agent protects the mouse brain from defective insulin signaling caused by Alzheimer's disease-associated Abeta oligomers

    The Journal of Clinical Investigation

    (2012)
  • L. Bondolfi et al.

    Amyloid-associated neuron loss and gliogenesis in the neocortex of amyloid precursor protein transgenic mice

    The Journal of Neuroscience: The Official Journal of the Society for Neuroscience

    (2002)
  • J. Born et al.

    Sniffing neuropeptides: a transnasal approach to the human brain

    Nature Neuroscience

    (2002)
  • A.L. Carvalho et al.

    Role of the brain-derived neurotrophic factor at glutamatergic synapses

    British Journal of Pharmacology

    (2008)
  • S. Cohen-Cory et al.

    Effects of brain-derived neurotrophic factor on optic axon branching and remodelling in vivo

    Nature

    (1995)
  • S. Craft

    Insulin resistance and Alzheimer's disease pathogenesis: potential mechanisms and implications for treatment

    Current Alzheimer Research

    (2007)
  • F.G. De Felice et al.

    Protection of synapses against Alzheimer's-linked toxins: insulin signaling prevents the pathogenic binding of Abeta oligomers

    Proceedings of the National Academy of Sciences of the United States of America

    (2009)
  • R. Deane et al.

    Role of the blood-brain barrier in the pathogenesis of Alzheimer's disease

    Current Alzheimer Research

    (2007)
  • J.E. Donahue et al.

    Apolipoprotein E, amyloid-beta, and blood-brain barrier permeability in Alzheimer disease

    Journal of Neuropathology and Experimental Neurology

    (2008)
  • Cited by (256)

    • Alternative role of glucagon-like Peptide-1 receptor agonists in neurodegenerative diseases

      2023, European Journal of Pharmacology
      Citation Excerpt :

      The effects of GLP-1 analogs could potentially improve neuronal connectivity and cognitive impairments in patients with AD. Other mechanisms contribute to the beneficial effects of liraglutide, including increasing formation of neuronal progenitor and stem cells (Hamilton et al., 2011; Parthsarathy and Holscher, 2013); reducing activated microglial numbers and extent of neuroinflammation, including late-stage AD mice (McClean and Holscher, 2014a; Paladugu et al., 2021); promoting autophagy via the AMPK/mTOR (Kong et al., 2018); improving mitochondrial function through the cAMP/PKA pathway in astrocytes and supporting neuronal survival (Xie et al., 2021); decreasing oxidative stress and lipid peroxidation levels by upregulating superoxidase dismutase and glutathione peroxidase (An et al., 2021); reducing oxidative/nitrosative stress and inflammation (Duarte et al., 2020); decreasing iron overload and oxidative stress (An et al., 2021); increasing the expression of Bcl-2; and reducing the expression of Bax with reduced hyperphosphorylated tau protein and Akt/GSK-3β signaling pathway in human SH-SY5Y cells under oxidative stress (Zheng et al., 2019). The benefits of liraglutide have been demonstrated in various AD models, such as Aβ1-42 intracerebroventricular injection (Qi et al., 2016); aged APP/PS1 mice (McClean and Holscher, 2014a); senescence-accelerated mouse prone eight mice (Hansen et al., 2015); various DM models, including streptozotocin-induced AD model (Xiong et al., 2013); hyperhomocysteinemia-induced AD-like pathology (Zhang et al., 2019b); 5xFAD mouse models of AD (Paladugu et al., 2021); and infusion of amyloid-β oligomers into the intracerebroventriculum of macaques (Batista et al., 2018).

    View all citing articles on Scopus
    View full text