Elsevier

Neuropharmacology

Volume 85, October 2014, Pages 113-120
Neuropharmacology

Insulin protects against Aβ-induced spatial memory impairment, hippocampal apoptosis and MAPKs signaling disruption

https://doi.org/10.1016/j.neuropharm.2014.01.036Get rights and content

Highlights

  • Intra-hippocampal insulin treatment protects against Aβ-induced memory impairment.

  • Intra-hippocampal insulin treatment prevents Aβ-induced hippocampal apoptosis.

  • Intra-hippocampal insulin treatment prevents Aβ-induced hippocampal P38 activation.

  • Intra-hippocampal insulin treatment prevents Aβ-induced hippocampal ERK activation.

Abstract

Alzheimer disease (AD) is a progressive neurodegenerative disease characterized by extracellular deposits of beta amyloid (Aβ) and neuronal loss particularly in the hippocampus. Accumulating evidences have implied that insulin signaling impairment plays a key role in the pathology of AD; as much as it is considered as type 3 Diabetes. MAPKs are a group of signaling molecules which are involved in pathobiology of AD. Therefore this study was designed to investigate if intrahippocampal insulin hinders Aβ-related memory deterioration, hippocampal apoptosis and MAPKs signaling alteration induced by Aβ. Adult male Sprague-Dawely rats weighing 250–300 g were used in this study. The canules were implanted bilaterally into CA1 region. Aβ25-35 was administered during first 4 days after surgery (5 μg/2.5 μL/daily). Insulin treatment (0.5 or 6 mU) was done during days 4–9. The animal's learning and memory capability was assessed on days 10–13 using Morris water maze. After finishing of behavioral studies the hippocampi was isolated and the amount of hippocampal cleaved caspase 3 (the landmark of apoptosis) and the phosphorylated (activated) forms of P38, JNK and ERK was analyzed by western blot. The results showed that insulin in 6 but not 0.5 mU reversed the memory loss induced by Aβ25-35. Western blot analysis revealed that Aβ25-35 induced elevation of caspase-3 and all 3 MAPks subfamily activity, while insulin in 6 mu restored ERK and P38 activation but has no effect on JNK. This study disclosed that intrahippocampal insulin treatment averts not only Aβ-induced memory deterioration but also hippocampal caspase-3, ERK and P38 activation.

Introduction

For several decades insulin-independent uptake of glucose, led scientists to regard brain as an insulin-independent tissue; however the discovery of insulin and its receptor within the brain revolutionized that old misconception (Havrankova et al., 1978a, Havrankova et al., 1978b). Since then numerous evidences have verified the expression of insulin receptors (IRs) in various brain structures (van Houten et al., 1979). Distribution of IRs in brain structures is not ubiquitous and some regions like the hippocampus have higher IR density (Hill et al., 1986). Insulin has been shown to play various important roles in central nervous system (CNS). It has also been shown that impaired insulin signaling is involved in pathology of Alzheimer Disease (AD); in a way that this disease is named as “Diabetes type-3” (reviewed in (Cholerton et al., 2013, Ghasemi et al., 2013)). Even some trial studies suggest that intranasal insulin treatment might be helpful in treating AD (Freiherr et al., 2013, Reger et al., 2006, Reger et al., 2008). Intranasal administration of insulin has been shown to achieve direct CNS delivery because it travels through olfactory nerve (both extracellularly and intracellularly), while has no systemic effect (Chapman et al., 2013).

AD is one of the most prevalent neurodegenerative diseases estimating to affect about 30 million people in the world (Holtzman et al., 2011). This disease is characterized by accumulation of extracellular senile plaques and intracellular neurofibrillary tangles (NFT) along with neuronal loss particularly in the hippocampus (Serrano-Pozo et al., 2011). Hippocampus is one of the most important brain structures playing a pivotal role in spatial learning and memory (Li et al., 2012) and is the most important part of the brain affected by AD pathology (Fellgiebel and Yakushev, 2011).

MAPKs are a group of serine–threonine kinases playing roles in a variety of cellular activities and are divided into 3 main subgroups; extracellular signal-regulated kinases (ERKs), Jun N-terminal kinases (JNKs) and P38 MAPK (Jin et al., 2006). Theses kinases are assumed to contribute in the development of AD as there is a relationship between beta amyloid (Aβ) and them (reviewed in (Ghribi et al., 2003, Kim and Choi, 2010)). For instance MAPKs are reported to take part in tau hyperphosphorylation which in turn participates in AD pathology (Ferrer et al., 2001, Ferrer et al., 2003, Puig et al., 2004). MAPKs are also illustrated to play roles in APP metabolism and Aβ production (Cho et al., 2007, Colombo et al., 2009). Additionally they are documented to contribute in apoptosis and LTP disruption caused by Aβ (Ramin et al., 2011, Wang et al., 2004). Then MAPKs are considered to be engaged in AD pathology.

Considering the protective effects of insulin against AD, the present study aimed to clarify if intra-hippocampal insulin administration is protective against Aβ-induced spatial memory impairment, hippocampal apoptosis and MAPKs activity alteration.

Section snippets

Materials

Amyloid β-Protein 25-35 (Aβ25-35, A4559) was purchased from Sigma, USA; Western blot antibodies including caspase-3 (9665), beta-actin (4970), phospho-P38 (9211), phospho-JNK (4671), phospho-ERK (4377) and secondary HRP-conjugated (7074) were purchased from Cell Signaling Technology Company, USA. Amersham ECL select (RPN2235) reagent kit was purchased from GE healthcare, UK and PVDF membrane was purchased from Millipore. Other reagents were obtained from usual commercial sources.

Animals

Adult male

Spatial learning and memory test

The effects of vehicle, Aβ25-35 or (and) insulin administration on water maze spatial learning and memory is represented in Fig. 1. This figure shows that there is a negative linear correlation between escape latency and training days in all groups (the relative changes of escape latencies on day2/day1 and day3/day1 are significant in all groups), indicating that all groups have learnt the platform location. However Aβ25-35 administration slowed down the learning capability. Fig. 1A shows the

Discussion

Accumulation of Aβ on molecular level and memory loss in phenotypic level are known as the most important features of AD (reviewed in (Holtzman et al., 2011)). Several lines of evidences have shown that Aβ accumulation plays a key role in the memory impairment seen in AD (Saura et al., 2005). Furthermore converging evidences have shown that insulin signaling disturbances, particularly central insulin resistance, contribute in the physiopathology of AD (reviewed in (Ghasemi et al., 2013)). The

Acknowledgments

This work was derived from the thesis of Rasoul Ghasemi and supported by a grant (No. 90-5783) from Shiraz University of Medical Sciences, Shiraz, Iran. The help from Dr. Zahra Bagheri in statistical analysis is greatly appreciated.

References (64)

  • C.C. Lee et al.

    Insulin rescues amyloid beta-induced impairment of hippocampal long-term potentiation

    Neurobiol. Aging

    (2009)
  • H. Li et al.

    Unilateral lesion of dorsal hippocampus in adult rats impairs contralateral long-term potentiation in vivo and spatial memory in the early postoperative phase

    Behav. Brain Res.

    (2012)
  • M. Moosavi et al.

    The effect of intrahippocampal insulin microinjection on spatial learning and memory

    Horm. Behav.

    (2006)
  • M. Moosavi et al.

    Insulin protects against stress-induced impairments in water maze performance

    Behav. Brain Res.

    (2007)
  • M. Ramin et al.

    Inhibition of JNK phosphorylation reverses memory deficit induced by beta-amyloid (1-42) associated with decrease of apoptotic factors

    Behav. Brain Res.

    (2011)
  • M. Reger et al.

    Effects of intranasal insulin on cognition in memory-impaired older adults: modulation by APOE genotype

    Neurobiol. Aging

    (2006)
  • A.A. Rensink et al.

    Insulin inhibits amyloid beta-induced cell death in cultured human brain pericytes

    Neurobiol. Aging

    (2004)
  • M. Schafer et al.

    Insulin-specific sensitization of cultured cerebrocortical neurons to glutamate excitotoxicity

    Brain Res.

    (1992)
  • M. Stanciu et al.

    Persistent activation of ERK contributes to glutamate-induced oxidative toxicity in a neuronal cell line and primary cortical neuron cultures

    J. Biol. Chem.

    (2000)
  • T. Takadera et al.

    Toxic effect of a beta-amyloid peptide (beta 22-35) on the hippocampal neuron and its prevention

    Neurosci. Lett.

    (1993)
  • M. Townsend et al.

    Soluble Abeta inhibits specific signal transduction cascades common to the insulin receptor pathway

    J. Biol. Chem.

    (2007)
  • N. Yamamoto et al.

    Ketamine reduces amyloid beta-protein degradation by suppressing neprilysin expression in primary cultured astrocytes

    Neurosci. Lett.

    (2013)
  • R. Yang et al.

    Anti-amnesic effect of neurosteroid PREGS in Abeta25-35-injected mice through sigma1 receptor- and alpha7nAChR-mediated neuroprotection

    Neuropharmacology

    (2012)
  • A. Zarifkar et al.

    Agmatine prevents LPS-induced spatial memory impairment and hippocampal apoptosis

    Eur. J. Pharmacol.

    (2010)
  • X. Zhu et al.

    Differential activation of neuronal ERK, JNK/SAPK and p38 in Alzheimer disease: the 'two hit' hypothesis

    Mech. Ageing Dev.

    (2001)
  • N.R. Bhat et al.

    Hydrogen peroxide activation of multiple mitogen-activated protein kinases in an oligodendrocyte cell line: role of extracellular signal-regulated kinase in hydrogen peroxide-induced cell death

    J. Neurochem.

    (1999)
  • P.M. Canas et al.

    Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by beta-amyloid peptides via p38 mitogen-activated protein kinase pathway

    J. Neurosci.

    (2009)
  • C.D. Chapman et al.

    Intranasal treatment of central nervous system dysfunction in humans

    Pharm. Res.

    (2013)
  • H.J. Cho et al.

    IFN-gamma-induced BACE1 expression is mediated by activation of JAK2 and ERK1/2 signaling pathways and direct binding of STAT1 to BACE1 promoter in astrocytes

    Glia

    (2007)
  • S. Craft et al.

    Intranasal insulin therapy for Alzheimer disease and amnestic mild cognitive impairment: a pilot clinical trial

    Arch. Neurol.

    (2012)
  • P. Dandona et al.

    Anti-inflammatory effects of insulin

    Curr. Opin. Clin. Nutr. Metab. Care

    (2007)
  • A.I. Duarte et al.

    Oxidative stress affects synaptosomal gamma-aminobutyric acid and glutamate transport in diabetic rats: the role of insulin

    Diabetes

    (2004)
  • Cited by (56)

    • Mitochondrial dysfunction: A potential target for Alzheimer's disease intervention and treatment

      2021, Drug Discovery Today
      Citation Excerpt :

      In addition, impaired insulin responsiveness is correlated with dysfunctional mitochondrial glucose utilization in the 3xTg AD mouse model [66]. Also, insulin administration in the hippocampus can improve Aβ-induced memory impairment and can activate major hippocampal kinases (cellular signal-regulated kinases (ERK) and P38) [67,68]. More recently, in 2017, Craft et al. evaluated the long-acting insulin drug detemir, administered intranasally for 3 weeks, in adults with AD or amnestic mild cognitive impairment [69].

    • Intranasal insulin and orexins to treat age-related cognitive decline

      2021, Physiology and Behavior
      Citation Excerpt :

      However, this improvement in memory performance was only seen in young, but not aged, rats. Several studies have also demonstrated that intrahippocampal insulin administration dose-dependently enhances spatial working memory rats [134-136]. Additionally, it was found that intrahippocampal insulin reversed the spatial memory deficit induced by a high-fat diet in C57BL/6 J mice [137]; however, it was not sufficient to restore spatial learning and memory deficits induced by scopolamine in rats [138].

    View all citing articles on Scopus
    View full text