Open access

Insulin Resistance and Alzheimer’s Disease

Written By

Sung Min Son, Hong Joon Shin and Inhee Mook-Jung

Submitted: 26 November 2010 Published: 04 November 2011

DOI: 10.5772/23409

From the Edited Volume

Topics in the Prevention, Treatment and Complications of Type 2 Diabetes

Edited by Mark B. Zimering

Chapter metrics overview

4,766 Chapter Downloads

View Full Metrics

1. Introduction

Alzheimer disease (AD) is known as a form of type III diabetes due to its similar cellular responses and pathogenesis. Insulin alters normal brain function and peripheral glucose metabolism, and conditions that are related to insulin dysregulation, such as obesity, diabetes mellitus, and cardiovascular disease, have potentially harmful effects on brain function. Many reports have demonstrated that insulin resistance increases age-related memory impairments and is a risk factor for AD. The molecular and cellular link between insulin resistance and AD, however, is unknown. We discuss the potential mechanisms of these metabolic disorders in the pathogenesis of AD. Glucose homeostasis is critical for energy maintenance, neurogenesis, neuronal survival, and synaptic plasticity, which are required for learning and memory. During insulin resistance, one develops reduced sensitivity to insulin, resulting in hyperinsulinemia, and this impairment in insulin signaling mediates the pathogenesis of AD, which manifests as brain inflammation, oxidative stress, alterations in amyloid beta (Aβ) levels, and cell death. Human and experimental animal studies have noted that drugs that modulate insulin resistance decrease the accumulation of Aβ in the brain and the cognitive impairments that are associated with AD. Therapeutic strategies that target the link between insulin resistance and AD might benefit the development of future AD drugs.

Advertisement

2. Insulin signaling

2.1. Introduction

Insulin is a peptide hormone that comprises 51 amino acids and is synthesized in and secreted from pancreatic β-cells in the islet of Langerhans (Huang et al., 2010). Its rapid release is generally triggered by increased levels of glucose in the blood, despite other mechanisms that release insulin; normal blood glucose levels range from 4.4 to 6.1 mmol/L (82 to 110 mg/dL).

Of its many functions, the most significant effects of insulin are its promotion of cellular growth and differentiation, activation or repression of transcription, and regulation of protein kinases and phosphatases (Saltiel and Pessin, 2002). Defects in insulin signaling can affect many diseases, such as type 2 diabetes mellitus (T2DM), metabolic syndrome, and Alzheimer disease (AD). Although most studies have examined the function of insulin and the dysregulation of insulin secretion or insulin receptor signaling in peripheral tissue, it has recently been reported to cause serious mental illness (Huang et al., 2010).

2.2. General effects of insulin signaling

Insulin increases glycogen and lipid synthesis in liver and muscle cells and simultaneously inhibits glycogenolysis and gluconeogenesis in the liver during feeding (Saltiel and Kahn, 2001). These actions are mediated by its binding to membrane-bound insulin receptors (IRs), members of the tyrosine kinase receptor family (Saltiel and Pessin, 2002). The binding of insulin to the alpha subunit of IR induces a conformational change, resulting in the autophosphorylation of tyrosine residues in the beta subunit of the receptor (Van Obberghen et al., 2001). This ligation leads to the subsequent tyrosine phosphorylation of insulin receptor substrates 1-4 (IRS1-4), Shc, Gab-1, Cbl, and APS (Saltiel and Pessin, 2002), and activates downstream cytoplasmic effectors, such as the lipid kinase phosphatidylinositol 3-kinase (PI 3-kinase) and mitogen-activated protein kinase (MAPK) (Cole et al., 2007; Plum et al., 2005).

During insulin-mediated glucose uptake, the phosphorylated residues of insulin receptor and IRS1, 2, and 3 are recognized by the Src homology 2 (SH2) domain (Bevan, 2001) of the p85 regulatory subunit of PI 3-kinase. The catalytic subunit of PI 3-kinase, p110, phosphorylates phosphatidylinositol (4,5) bisphosphate (PtdIns(4,5)P2), leading to the formation of Ptd(3,4,5)P3 (Lizcano and Alessi, 2002). Ptd(3,4,5)P3 then binds to the plasma membrane and associates with phosphoinositol-dependent kinase-1 (PDK-1) and induces the phosphorylation and activation of protein kinase B (PKB)/Akt, a serine-threonine kinase. Activated Akt enters the cytoplasm, where it evokes the phosphorylation and inactivation of glycogen synthase kinase 3 (GSK3), which promotes glycogen synthesis.

By activating PI3-kinase and AKT (Lizcano and Alessi, 2002), insulin affects the translocation of glucose transporter 4 (GLUT4) from intracellular stores to the plasma membrane: When insulin concentrations are low, GLUT4 exists in cytoplasmic vesicles. After insulin binds to its receptors, GLUT4-containing vesicles fuse with the plasma membrane, and the newly inserted GLUT4 takes up glucose efficiently. In parallel, there is a PI 3-kinase independent pathway that recruits GLUT4 to the plasma membrane, led by the phosphorylation of the proto-oncogene Cbl, which is associated with the adaptor protein c-Cbl-associated protein (CAP) (Saltiel and Kahn, 2001).

Insulin promotes the uptake and synthesis of fatty acids in the liver and prevents lipolysis by inhibiting the intracellular lipase that hydrolyzes triglycerides to release fatty acids. Although insulin promotes the synthesis of glycogen in the liver, further synthesis is suppressed when the liver is saturated with glycogen. Any additional glucose that is taken up is then shunted into pathways that lead to fatty acid synthesis (Saltiel and Kahn, 2001).

One study has indicated that lipid synthesis requires increased expression of the transcription factor steroid regulatory element-binding protein (SREBP)-1c (Shimomura et al., 1999), which is synthesized as a membrane protein in the endoplasmic reticulum (ER) (Sato, 2010). Insulin is believed to induce SREBP-1c proteolytic activity and, consequently, insulin-mediated lipogenesis (Sato, 2010). Further, insulin inhibits lipid metabolism by decreasing cellular concentrations of cAMP through activation of a cAMP-specific phosphodiesterase in adipocytes (Kitamura et al., 1999).

Insulin stimulates amino acid uptake into cells, inhibits protein degradation, and promotes protein synthesis (Saltiel and Kahn, 2001) by stimulating components of the translational machinery, including eukaryotic initiation factors (elFs) and eukaryotic elongation factors (eEFs) (Proud, 2006). This entails the activation of PKB, leading to the phosphorylation and inactivation of GSK3, which in turn promotes phosphorylation and inhibition of a guanine nucleotide exchange factor, eIF2B, to regulate the initiation of translation (Proud, 2006).

Moreover, PKB phosphorylates the tuberous sclerosis complex 1 (TSC1)-TSC2 complex, relieving its inhibition of the mammalian target of rapamycin (mTOR) (Proud, 2006), which promotes protein synthesis through p70 ribosomal S6 kinase (p70S6k) and inhibition of eIF-4E binding protein (4E-BP1), and thereby governs ribosomal protein translation (Proud, 2006).

2.3. Insulin in the brain

The brain is the primary consumer of glucose, requiring two-thirds of total circulating glucose daily (Peters, 2011); thus, the regulation of glucose homeostasis by insulin in the central nervous system (CNS) is paramount for normal brain function. Insulin, by a saturable transport process that is mediated by insulin receptor, can cross the blood-brain barrier (BBB), which governs the transduction between the CNS and peripheral tissues (Neumann et al., 2008). Although insulin is synthesized locally in the brain (Banks et al., 1997; Devaskar et al., 1994; Rulifson et al., 2002), its function is unknown.

IRs in the CNS are widely expressed throughout the brain in neurons and glial cells, at particularly high concentrations in the cerebral cortex, hippocampus, hypothalamus, and olfactory bulb (Neumann et al., 2008). They have disparate functions, molecular weights, and structures from IRs in peripheral organs (Heidenreich et al., 1983), but it is unknown whether the counter effects of insulin in the CNS compared with those in peripheral tissues are attributed to such differences. Insulin increases glucose and inhibits feeding in the CNS, but decreases glucose and stimulates feeding in peripheral tissue (Florant et al., 1991). A recent report observed that impaired brain insulin activity might affect unrestrained lipolysis that initiates and exacerbates type 2 diabetes mellitus in humans (Scherer et al., 2011).

2.4. Role of insulin in cognitive function

In humans, brain insulin enhances learning, memory, and, in particular, verbal memory (Benedict et al., 2004). Insulin signaling in the limbic system and hypothalamus is especially important for cognitive function (Zhao et al., 1999), independent of changes in peripheral glucose (Craft et al., 1999; Kern et al., 1999). These functions have been supported by evidence that insulin modulates the concentrations of neurotransmitters, which influence cognition, such as Ach (acetylcholine) (Kopf and Baratti, 1999). Insulin also alters synaptic plasticity by regulating the endocytosis of 3-hydroxy-5-methylisoxazole-4-propionic acid (AMPA) receptor, which causes long-term depression (LTD) of excitatory synaptic transmission in the hippocampus and cerebellum (Huang et al., 2003; Man et al., 2000; Wang and Linden, 2000), and by enhancing N-methyl-D-aspartate (NMDA) receptor-mediated synaptic transmission at hippocampal CA1 synapses (van der Heide et al., 2005).

2.5. Insulin-like growth factor (IGF)

Insulin-like growth factors are polypeptides that are similar in sequence to proinsulin (Clemmons, 2007). Insulin-like growth factor 1, which stimulates cell growth and proliferation, especially in nerve cells, binds to IGF receptor and IRs (Jones et al., 2009). IGF-1 functions similarly to insulin receptor. IGF-1, a tyrosine kinase, initiates signaling cascades through IRS and enhances insulin activity (Clemmons, 2007).

Deficiency of or irresponsiveness to IGF-1 causes not only growth failure but also dyslipidemia (Twickler et al., 2003), insulin resistance (Conti et al., 2002; Twickler et al., 2003), and obesity (Rasmussen et al., 1995). According to recent longitudinal studies, these phenotypes are closely related to a high risk of neurodegenerative disorders, such as dementia and AD (Luchsinger et al., 2004; Ott et al., 1996; Ronnemaa et al., 2008). Similarly, mice in which IGF-1 has been deleted develop increased Aβ levels in the brain, suggesting that IGF-1 promotes the clearance of Aβ (Carro et al., 2002). Moreover, IGF-1 mediates transient site-selective increases in tau phosphorylation—hyperphosphorylated tau is a pathological hallmark of AD—in primary cortical neurons via GSK-3 pathways (Lesort and Johnson, 2000).

Figure 1.

Schematic diagram of the effects of peripheral insulin resistance on insulin, insulin-degrading enzyme (IDE) and Aβ levels. Peripheral insulin resistance and hyperinsulinemia triggers an excess release of FFA from adipocytes to liver and muscles. A three-fold increase of FFAs reduce insulin-dependent skeletal muscle glucose uptake by 50% (Roden et al., 1996), thereby hampering further insulin signal transduction (Hotamisligil et al., 1996; Schinner et al., 2005). In addition, Increased peripheral FFA levels invoke elevations in TNFα in the periphery and in the CNS, which may result in increased accumulation of Aβ. Chronic elevations in plasma Aβ may result in increased transport of Aβ into the brain. In contrast, peripheral hyperinsulinemia decrease insulin transport into the CNS. Low insulin level in CNS, and low level of IDE may contribute to the formation of senile plaques via disability to degrade Aβ peptides (Jones et al., 2009), further promoting intraneuronal Aβ accumulation, which is the hallmark of AD. <modified from Craft’s paper (Craft, 2007 )>

2.6. Insulin-degrading enzyme (IDE)

Insulin-degrading enzyme is the chief enzyme that degrades excess insulin and other substrates, including Aβ, a peptide that is implicated in the pathogenesis of AD (Wang et al., 2010). IDE knockout mice experienced decreased Aβ degradation, hyperinsulinemia, and hyperglycemia (Farris et al., 2003). As insulin levels rise, IDE expression increases to prevent the chronic activation of insulin (Zhao et al., 2004). Reduced IDE activity might contribute to the formation of senile plaques through the inability to degrade Aβ peptides (Jones et al., 2009).

Advertisement

3. Insulin resistance

3.1. Definition of insulin resistance

Insulin resistance is a physiological condition in which one loses or reduces his sensitivity to insulin—that is, it becomes less effective at lowering glucose levels in the blood. There are several representative phenotypes of insulin resistance that are used to diagnose diabetes, such as hyperinsulinemia (elevated levels of insulin that are required to maintain normal glucose levels in the bloodstream) and hyperglycemia (elevated glucose levels in the bloodstream that result from the failure of insulin to mediate glucose uptake or insufficient production of insulin by β-cells). Thus, insulin resistance reduces glycogen storage in muscle and fat cells and decreases hepatic glucose uptake.

High cortisol and uric acid levels and low vitamin D levels (Chiu et al., 2004; Vuorinen-Markkola and Yki-Jarvinen, 1994) are believed to be dietary conditions that precipitate insulin resistance. In addition, insulin resistance is closely related to obesity, hypertension, polycystic ovarian syndrome, dyslipidemia, and atherosclerosis (Saltiel and Pessin, 2002).

3.2. Peripheral effects of insulin resistance

Advanced glycation end products (AGEs) production is a pathological phenotype that results from hyperglycemia (Jager et al., 2007). Activation of the jun-N-terminal kinase (JNK) pathway by the binding of AGEs to their receptor (RAGE) leads to serine phosphorylation of IRS proteins and disrupts the imbalance in glucose levels further (Han et al., 2011). In addition to excess AGE production, prolonged hyperglycemia activates the transcription factor nuclear factor kappa B (NF-kB), which regulates proinflammatory and antiapoptotic pathways by modulating the transcription of cytokine and antioxidant genes (Baeuerle and Baltimore, 1996); it also controls the expression of genes that mediate immune responses and proliferation.

Peripheral insulin resistance mediates the progression to hyperglycemia and affects other functions of insulin. In particular, it affects reduced uptake of circulating lipids and increased hydrolysis of stored triglycerides. Such dysfunctional lipid homeostasis has been identified as a significant trigger of insulin resistance (Jones et al., 2009)

Because insulin resistance is linked to metabolic syndrome, which is generally characterized as overweightedness and obesity, and is often observed in persons with hyperglycemia and visceral adiposity, visceral fat is believed to accelerate insulin insensitivity, since visceral fat is less sensitive to the antilipolytic activity of insulin than subcutaneous fat (Jones et al., 2009), thereby triggering excessive release of free fatty acids (FFAs) (Schulingkamp et al., 2000) into the bloodstream. FFAs increase 3-fold above basal levels, reducing insulin-dependent skeletal muscle glucose uptake by 50% (Roden et al., 1996). This phenomenon, a direct cause of insulin resistance, induces inhibits serine phosphorylation of IRS and decreases its chances of binding to insulin receptor, further hampering insulin signaling (Hotamisligil et al., 1996; Schinner et al., 2005).

3.3. Central insulin resistance

As discussed, an abnormality in peripheral insulin is a risk factor for memory loss and neurodegeneration. In addition, chronic exposure of neurons to high insulin levels has a negative impact on memory, according to animal model studies. Chronic hyperinsulinemia impairs BBB function and IR activity and reduces insulin transport to the brain (Strachan et al., 1997). In contrast, some studies have noted that acute increases in brain insulin enhance memory (Craft et al., 1999; Park et al., 2000).

Advertisement

4. Link between insulin resistance and Alzheimer disease

Many studies have suggested that patients with diabetes have a higher risk of AD. Luchsinger et al. found that hyperinsulinemia increased risk for AD (Luchsinger et al., 2004). Also, it has been shown that high levels of insulin and insulin resistance are associated with a higher risk of AD in Rotterdam study, a population-based cohort study (Ott et al., 1999; Schrijvers et al., 2010). Although relationship between diabetes and AD remains controversial (Profenno et al., 2010), the abnormalities in insulin metabolism, which influence the onset of AD mechanistically, are believed to mediate AD through their influence on the synthesis and degradation of Aβ, tau hyperphosphorylation, oxidative stress and/or inflammation.

4.1. Impaired insulin/IGF-1 system in the AD brain

By positron emission tomography (PET) of early-stage AD and mild cognitive impairment (MCI), glucose uptake and metabolism decline significantly in the cortex (Jagust et al., 1991). According to another study, cerebral glucose utilization and blood flow fall by 45% and 18%, respectively, in the early stages of AD. In late-stage AD, cerebral blood flow is reduced by 65% (Hoyer and Nitsch, 1989). These data indicate that brain metabolism is altered in the AD brain, resembling T2DM. In addition, AD patients experience decreases in insulin in the CSF, reflecting dysregulation in insulin transport to the brain (Messier and White, 1987).

The AD brain also exhibits reduced expression of IR and IGF-1R, IRS, and trophic factors (Steen et al., 2005). IGF-1R expression decreases in AD brains in proportion to disease severity, and IGF-1 mRNA levels decline in late-stage AD (Frolich et al., 1998). IRS1/2 expression is decreased in the AD brain (Bosco et al., 2011; Squire, 1986). Because IRS1 and 2 regulate insulin and IGF-1 signaling, these findings suggest that insulin/IGF-1 signal transduction is altered in the AD brain.

There is much evidence of altered insulin signaling in AD. Inhibition of insulin/IGF-1 signaling in AD blocks the Wnt pathway (Doble and Woodgett, 2003), which mediates normal physiological processes in animals (Gogolla et al., 2009). Further, decreased PI3K signaling reduces GLUT4 translocation, inhibiting glucose uptake (Johnston et al., 2003). Since the brain depends on glucose as an energy source, altered glucose metabolism reduces the synthesis of acetyl-CoA and, ultimately, acetylcholine, an important neurotransmitter (Gibson et al., 1981). This decrease disrupts synaptic transmission and, consequently, impairs memory (Craft et al., 2003).

4.2. Insulin resistance and APP processing

Recent studies have indicated that abnormalities in insulin metabolism mediate the onset of AD through their influence on the synthesis and degradation of Aβ peptides (Table 1). Insulin significantly upregulates extracellular levels of Aβ40 and Aβ42 peptides through acceleration of amyloid precursor protein (APP)/Aβ trafficking from the Golgi/trans-Golgi network (TGN), a major site of Aβ generation, to the plasma membrane (Gasparini et al., 2001). Also, certain downstream signaling pathways of IR might also regulate the generation of Aβ peptides by modulating the expression or activity of β-secretase or γ-secretase (Phiel et al., 2003; Zhang et al., 2011), major components of APP processing (Edbauer et al., 2003; Vassar et al., 1999).

Under AD-like conditions, the expression levels of the neurotrophin receptors TrkA (tyrosine kinase receptor A) and p75NTR (p75 neurotrophin receptor) change markedly, increasing Aβ production by stabilizing β-secretase through the activation of sphingomyelinase (Puglielli et al., 2003). Chronic treatment of neuronal cells with IGF-1 (which induces insulin resistance) results in alteration in expression of TrkA and p75NTR, as in AD brains, increasing Aβ production (Costantini et al., 2006). Aβ is a ligand of p75NTR (Yaar et al., 1997), and Aβ stimulates p75NTR-mediated cell death in vitro and in vivo (Coulson, 2006; Sotthibundhu et al., 2008).

Figure 2.

Amyloid plaque and neurofibrillary tangles as hallmarks of Alzheimer disease (AD). AD is a progressive neurodegenerative disease characterized by senile plaques, neurofibrillary tangles and neuronal loss. Abnormal aggregates of amyloid-beta peptide (Aβ) are found in extracellular senile plaques and associated with neurodegeneration in AD. Neurofibrillary tangles are aggregates of the microtubule-associated protein (MAP) tau protein which is hyperphosphorylated by kinases and accumulated inside the neurons themselves.

In AD brains, Aβ accumulates on not only alterations in Aβ generation but also dysfunction of Aβ clearance. Several mechanisms of Aβ clearance have been examined: 1) Enzymatic degradation by microglia or endopeptidases, such as IDE (Qiu and Folstein, 2006), neprilysin (Iwata et al., 2000), endothelin-converting enzyme (ECE) (Eckman et al., 2003), angiotensin-converting enzyme (ACE) (Lehmann et al., 2005), matrix metalloproteinase-9 (MMP-9), and plasmin (Nalivaeva et al., 2008); 2) Influx into receptor-mediated transport across the BBB primarily via RAGE (Deane et al., 2003; Han et al., 2011); and 3) Receptor-mediated BBB transport by binding to low-density lipoprotein receptor-related protein-1 (LRP1) after ligation with apolipoprotein E (APOE) or α2-macroglobulin (α2M), to be delivered to peripheral sites of degradation (Shibata et al., 2000).

With regard to insulin resistance, insulin induces the accumulation of Aβ by limiting Aβ degradation through direct competition for IDE. IDE is a zinc metalloendopeptidase that preferentially cleaves proteins that tend to form β-pleated sheet-rich amyloid fibrils, such as Aβ peptides (Qiu and Folstein, 2006). IDE activity in the brain correlates negatively with Aβ levels (Farris et al., 2003), and IDE expression is decreased in the AD brain (Cook et al., 2003). Insulin regulates the levels of IDE, and insulin resistance reduces Aβ clearance by down-regulating IDE expression or competing with it for binding (Farris et al., 2003; Ho et al., 2004). A recent report has indicated that Aβ oligomers disrupt IR signaling, resulting from a rapid and substantial loss of neuronal IRs on dendrites by Aβ oligomer (Zhao et al., 2008). There is also evidence that Aβ reduces insulin binding to IRs and induces insulin resistance (Xie et al., 2002).

4.3. Insulin resistance and tau phosphorylation

AD is a multifactorial disease in which protein alterations, oxidative stress, inflammation, dysregulated immunity, impaired neuronal-glial communication, and increase of neurotoxic agents triggering neuronal death. AD is defined by Aβ pathology (Aβ plaques) and tau pathology [neurofibrillary tangles (NFTs)], but whether tau mediates AD pathology has been discussed for many years. Nevertheless, there is increasing evidence that when tau proteins assume pathological forms, they compromise neuronal function and affect cell death. These results suggest that tau is an important mediator of Aβ toxicity and AD pathology (Pritchard et al., 2011).

Microtubule (MT)-associated protein (MAP) tau is the major MT-associated phosphor-protein in normal neuronal cells (Cleveland et al., 1977). Normally, tau proteins assemble with tubulin to stabilize microtubules and vesicular transport. Neurofibrillary tangles are hyperphosphorylated and aggregated forms of tau proteins. When it is hyperphosphorylated, tau becomes insoluble and lacks affinity for microtubules, leading to neurodegeneration (Iqbal et al., 2005).

The kinases that regulate tau phosphorylation are grouped according to specificity: 1) proline-directed protein kinases, such as cyclin-dependent kinase 5 (CDK5), glycogen synthase kinase 3β (GSK3β), MAPK1, p38, JNK, and ERK2; and 2) nonproline-directed protein kinases, such as cAMP-dependent protein kinase A (PKA), PKC, CaMKII, CKII, and MAP/microtubule affinity-regulating kinase (MARK) (Baudier et al., 1987; Drewes et al., 1992; Goedert et al., 1997; Hanger et al., 1992; Morishima-Kawashima et al., 1995).

Tau hyperphosphorylation can also be induced by decreased dephosphorylation. Several protein phosphatases (PPs), including PP1, PP2A, PP2B, and PP5, catalyze tau dephosphorylation (Drewes et al., 1993; Liu et al., 2005). The appearance of tau hyperphosphorylation in the brain reflects an early stage of AD (Maccioni et al., 2010).

Insulin and IGF-1 are related to the phosphorylation state of tau (Table 1). Transient insulin treatment increases tau phosphorylation temporarily, correlating with the sequential activation and deactivation of GSK3β in SH-SY5Y cells (Lesort et al., 1999). Further, tau phosphorylation in primary cortical neurons is regulated by insulin and IGF-1 (Lesort and Johnson, 2000). In IRS-2 knockout mice, defects in IRS-2 signaling promotes neuronal death and induces the hyperphosphorylation of tau, suggesting that tau-mediated neurodegeneration is regulated by the IRS-2 branch of the insulin-IGF signaling pathway (Schubert et al., 2003).

Neuron-specific deletion of insulin receptor (neuronal/brain-specific insulin receptor knockout (NIRKO)) in mice leads to tau hyperphosphorylation (Schubert et al., 2004). However, the patterns of phosphorylation in NIRKO mice and IRS-2 knockout mice differ, suggesting that tau phosphorylation is regulated not only by insulin resistance, but also by other factors (e.g., hyperinsulinemia, hyperglycemia, inflammation) (Schubert et al., 2003; Schubert et al., 2004). There are other evidences on the relationship between insulin signaling and tau hyperphosphorylation. In IGF-1 knockout mice, tau is hyperphosphorylated at Ser202 and Ser396, implicating IGF-1 as protective against tau hyperphosphorylation (Cheng et al., 2005). Separately, the peripheral administration of insulin induced site-specific tau phosphorylation (at Ser202), which occurred rapidly after acute insulin administration but prolonged (Freude et al., 2005).

4.4. Insulin resistance and oxidative stress

The increase in oxidative stress and the resulting activation of many signaling pathways are associated with AD (Markesbery and Carney, 1999; Nunomura et al., 2001).

Oxidative stress is manifested by damage to proteins (Smith et al., 1991; Smith et al., 1996), lipids (Sayre et al., 1997), and nucleic acids (DNA, RNA) (Gabbita et al., 1998; Mecocci et al., 1994; Nunomura et al., 1999). In AD, neurons that show increased oxidative damage have significantly more mtDNA, indicating that mtDNA sustains high oxidative stress in AD (Hirai et al., 2001).

Mitochondria from the brains of AD patients experience structural damage and are the site of Aβ accumulation (Gouras et al., 2005; Hirai et al., 2001). Further, important mitochondrial enzymes become impaired in the brains of AD transgenic mice (Caspersen et al., 2005). Dysregulated mitochondria release oxidizing free radicals, with peroxidation of membrane lipids and the output of toxic aldehydes that cause considerable oxidative stress in AD (Smith et al., 1996).

Oxidative stress is a common feature in AD and insulin resistance. In Drosophila, oxidative stress increases signaling through c-Jun N-terminal kinase (JNK), which is a related pathway of AD and insulin resistance (Ozcan et al., 2004). Hydrogen peroxide-induced JNK activation induces γ-secretase activation, and increases Aβ generation (Shen et al., 2008). Insulin resistance leads to greater oxidative stress, DNA damage, mitochondrial dysfunction, and, ultimately, cell death (de la Monte, 2009). In AD patient brains, the expression of pro-oxidant enzymes that catalyze the generation of reactive nitrogen (RNS) and oxygen species (ROS), such as nitric oxide synthase (NOS) and NADPH oxidase (NOX), rises significantly, which accelerates insulin resistance in the brain (de la Monte and Wands, 2006).

4.5. Insulin resistance and inflammation

There are many reports that inflammation mediates the progress of AD. In brains of AD model mice and AD patients, Aβ plaque and NFTs activate astrocytes and microglia, resulting in the release of inflammatory molecules (cytokines and chemokines) and the production of complement, and ultimately causing neuroinflammation (Eikelenboom et al., 2002; Hoozemans et al., 2002; McGeer and McGeer, 2003; Tansey et al., 2007). In fact, in AD, neurons themselves induce the production of inflammatory molecules, such as interleukin (IL)-1, IL-6, TNF-α, and complement proteins (Li et al., 2000; Tchelingerian et al., 1996; Yu et al., 2002).Further, Aβ can attract and activate microglia, affecting the recruitment of microglia to Aβ plaques, and increase the secretion of proinflammatory molecules, such as IL-1, IL-6, and IL-8 (Rogers and Lue, 2001). Astrocytes are also activated by Aβ peptide to produce proinflammatory cytokines and chemokines in AD (Smits et al., 2002). Conversely, inflammatory cytokines, such as IL-6, regulate APP processing, resulting in elevation of Aβ42 levels (Papassotiropoulos et al., 2001).

Figure 3.

Schematic representation of molecular pathways linking insulin resistance and Alzheimer disease. Peripheral insulin resistance leads to decrease insulin signaling in CNS, followed by alteration in brain metabolism. Increased Aβ toxicity, Tau hyperphosphorylation, oxidative stress and neuroinflammation are attributed to central insulin resistance, which leads to neurodegeneration.

Chronic administration of insulin induces inflammatory responses, although low doses of insulin is anti-inflammatory (Krogh-Madsen et al., 2004), suggesting that insulin contributes to inflammation. Several reports have noted that insulin modulates many inflammatory networks. Co-administration of insulin and lipopolysaccharide has a synergistic effect on the release of inflammatory cytokines, such as IL-1β and IL-6 (Soop et al., 2002). Insulin can regulate norepinephrine uptake in the locus coeruleus by modulating the expression of norepinephrine transporter protein (NET) (Figlewicz et al., 1993). Because norepinephrine is an endogenous anti-inflammatory neuromodulator that prevents IL-1β expression (Heneka et al., 2002), chronic administration of insulin might have abnormal inflammatory effects.

Advertisement

5. Clinical studies on insulin resistance and AD

Much evidence suggests that insulin resistance and peripheral hyperinsulinemia are risk factors for AD, implicating them as novel targets (Craft, 2005; Watson and Craft, 2003). For example, optimal doses of insulin and glucose in patients with AD improve declines in cognition (Watson et al., 2005). Using hyperinsulinemic-euglycemic clamps, intravenous insulin infusion in older subjects and in AD patients improves memory (Craft et al., 2003). Recently, intranasal administration of insulin was shown to improve cognitive function and modulate APP processing, resulting in increased Aβ40/Aβ42 ratio (Reger et al., 2008).

1. Aβ metabolism
Insulin increases the extracellular level of Aβ by promoting its secretion. (Gasparini et al., 2001)
Insulin increases the extracellular level of Aβ by inhibiting its degradation via insulin-degrading enzyme (IDE). (Gasparini et al., 2001)
Chronic treatment with IGF-1 increases Aβ production by switching from TrkA to p75NTR. (Costantini et al., 2006)
IGF-1 reduces BACE-1 expression in PC12 cells via activation of PI3K and ERK1/2 signaling pathways. (Zhang et al., 2011)
IGF-1 enhanced APP phosphorylation at Thr668, contributing to the neuronal degeneration in AD. (Araki et al., 2009; Chang et al., 2006)
Infusion of insulin under glucose clamp originated a rapid increase in CSF Aβ levels in older subject (Watson and Craft, 2003)
Systemic administration of IGF-1 increases CSF Aβ levels. (Carro et al., 2002)
IGF-1 increases neuronal excitability and release of Aβ is enhanced by neuronal activity. (Gomez, 2008)
2. Tau hyperphosphorylation
Insulin and IGF-1 treatment results in increase in tau phosphorylation in primary cortical neurons. (Lesort and Johnson, 2000)
Peripheral administration of insulin induces site-specific tau phosphorylation (Ser202). (Freude et al., 2005)
In IGF-1 knockout mice, tau is hyperphosphorylated at Ser202 and Ser396. (Cheng et al., 2005)
In IRS-2 knockout mice, IRS-2 regulates tau-mediated neurodegeneration. (Schubert et al., 2003)
Neuronal/brain-specific insulin receptor knockout (NIRKO) in mice leads to tau hyperphosphorylation. (Schubert et al., 2004)

Table 1.

The direct role for insulin or IGF-1 as a possible mediator of AD.

In addition to insulin, insulin-sensitizing compounds have been used as pharmacological treatments. Thiazolidinediones (TZDs) act as agonists of nuclear receptor PPAR-γ (peroxisome proliferator-activated receptor-gamma) and improve insulin sensitivity by reducing circulating insulin, FFA levels, and glucose uptake. TZDs are also anti-inflammatory drugs that decrease the levels of many inflammatory mediators (Jiang et al., 2008; Neumann et al., 2008; Rojo et al., 2008). Pioglitazone and rosiglitazone are some of the TZDs that are available for clinical use. The dual function of TZDs also affects hallmarks of AD pathogenesis, such as Aβ levels and tau hyperphosphorylation. PPAR-γ agonists, such as pioglitazone and indomethacin, can regulate Aβ42 levels and Aβ plaque loads (Heneka et al., 2005; Sastre et al., 2003).

Troglitazone decreases tau phosphorylation at Ser202 and Ser396/404 through a PPAR-γ-dependent/independent mechanism (d'Abramo et al., 2006). In a recent report, rosiglitazone reduced tau phosphorylation through JNK inhibition (Yoon et al., 2010).

TZDs improve memory in animal models and patients with AD. Rosiglitazone, a TZD, attenuates learning and memory deficits in Tg2576 mouse, an animal model of AD (Pedersen et al., 2006). Recently, pioglitazone was noted to preserve cognitive function in AD (Sato et al., 2009). This pilot study on 42 patients who were treated with 15-30 mg pioglitazone daily or placebo observed cognitive improvement in the pioglitazone groups. In another study, 511 AD patients were assigned to receive 2, 4, or 8 mg rosiglitazone (rosiglitazone group) or nothing (control group) (Risner et al., 2006); the rosiglitazone group improved cognition significantly, and this result was restricted to non-APOE ε4 carriers.

Insulin-sensitizing hormones, such as ghrelin, also affect AD pathogenesis. Ghrelin is a 28-amino-acid endogenous hormone that is a ligand of growth hormone secretagogue receptor 1a (GHSR-1a) (Kojima et al., 1999). Several studies have shown that ghrelin reduces tau hyperphosphorylation in high-glucose-induced neurons (Chen et al., 2010), and ghrelin enhances hippocampus-dependent memory in rodents (Carlini et al., 2010). Recently, Moon et al. observed that ghrelin ameliorates cognitive dysfunction in oligomeric Aβ42-injected mice (Moon et al., 2011). These and other reports demonstrate that brain insulin resistance leads to AD pathologies and thus suggest that insulin-sensitizing agents, such as the TZDs and ghrelin, are useful therapeutic agents.

Advertisement

6. Conclusion

Insulin resistance is characterized by chronic peripheral insulin elevations, central reduction of insulin levels and abnormality of insulin activity. Insulin modulates not only peripheral glucose metabolism but also normal brain function, and thereby conditions related to insulin dysregulation, such as obesity and diabetes mellitus, have deleterious effects on brain function. According to recent longitudinal studies, insulin resistance increases the risk of memory deficit and AD. Potential mechanisms linking insulin resistance to AD pathogenesis include the alteration in APP processing, defects in signal transduction related to both neuronal function (e.g. GSK3β) and cellular toxicity including oxidative stress and inflammation. Identification of underlying mechanisms for relevance of insulin resistance to AD is exigent goal to develop effective therapeutic strategies targeting AD. Many evidences have noted that agents modulating insulin resistance, including TZDs and insulin-sensitizing hormones, alleviate cognitive impairments associated with AD. Therapeutic strategies focused on preventing or regulating insulin resistance may thus benefit the development of drugs for AD.

Advertisement

7. Acknowledgement

This work was supported by Proteomics (FPR08B1-200), WCU-Neurocytomics, and KHID-ROAD grants.

References

  1. 1. Araki W. Kume H. Oda A. Tamaoka A. Kametani F. 2009 IGF-1 promotes beta-amyloid production by a secretase-independent mechanism. Biochem Biophys Res Commun 380 111 4 .
  2. 2. Baeuerle P. A. Baltimore D. 1996 NF-kappa B: ten years after. Cell 87 13 20 .
  3. 3. Banks W. A. Jaspan J. B. Huang W. Kastin A. J. 1997 Transport of insulin across the blood-brain barrier: saturability at euglycemic doses of insulin. Peptides 18 1423 9 .
  4. 4. Baudier J. Lee S. H. Cole R. D. 1987 Separation of the different microtubule-associated tau protein species from bovine brain and their mode II phosphorylation by Ca2+/phospholipid-dependent protein kinase C. J Biol Chem 262 17584 90 .
  5. 5. Benedict C. Hallschmid M. Hatke A. Schultes B. Fehm H. L. Born J. Kern W. 2004 Intranasal insulin improves memory in humans. Psychoneuroendocrinology 29 1326 34 .
  6. 6. Bevan P. 2001 Insulin signalling. J Cell Sci 114 1429 30 .
  7. 7. Bosco D. Fava A. Plastino M. Montalcini T. Pujia A. 2011 Possible implications of Insulin Resistance and Glucose metabolism in Alzheimer’s disease pathogenesis. J Cell Mol Med.
  8. 8. Carlini V. P. Ghersi M. Schioth H. B. de Barioglio S. R. 2010 Ghrelin and memory: differential effects on acquisition and retrieval. Peptides 31 1190 3 .
  9. 9. Carro E. Trejo J. L. Gomez-Isla T. Le Roith D. Torres-Aleman I. 2002 Serum insulin-like growth factor I regulates brain amyloid-beta levels. Nat Med 8 1390 7 .
  10. 10. Caspersen C. Wang N. Yao J. Sosunov A. Chen X. Lustbader J. W. Xu H. W. Stern D. Mc Khann G. Yan S. D. 2005 Mitochondrial Abeta: a potential focal point for neuronal metabolic dysfunction in Alzheimer’s disease. FASEB J 19 2040 1 .
  11. 11. Chang K. A. Kim H. S. Ha T. Y. Ha J. W. Shin K. Y. Jeong Y. H. Lee J. P. Park C. H. Kim S. Baik T. K. et al. 2006 Phosphorylation of amyloid precursor protein (APP) at Thr668 regulates the nuclear translocation of the APP intracellular domain and induces neurodegeneration. Mol Cell Biol 26 4327 38 .
  12. 12. Chen Y. Cao C. P. Li C. R. Wang W. Zhang D. Han L. L. Zhang X. Q. Kim A. Kim S. Liu G. L. 2010 Ghrelin modulates insulin sensitivity and tau phosphorylation in high glucose-induced hippocampal neurons. Biol Pharm Bull 33 1165 9 .
  13. 13. Cheng C. M. Tseng V. Wang J. Wang D. Matyakhina L. Bondy C. A. 2005 Tau is hyperphosphorylated in the insulin-like growth factor-I null brain. Endocrinology 146 5086 91 .
  14. 14. Chiu K. C. Chu A. Go V. L. Saad M. F. 2004 Hypovitaminosis D is associated with insulin resistance and beta cell dysfunction. Am J Clin Nutr 79 820 5 .
  15. 15. Clemmons D. R. 2007 Modifying IGF1 activity: an approach to treat endocrine disorders, atherosclerosis and cancer. Nat Rev Drug Discov 6 821 33 .
  16. 16. Cleveland D. W. Hwo S. Y. Kirschner M. W. 1977 Physical and chemical properties of purified tau factor and the role of tau in microtubule assembly.J Mol Biol116 227 47 .
  17. 17. Cole A. R. Astell A. Green C. Sutherland C. 2007 Molecular connexions between dementia and diabetes. Neurosci Biobehav Rev 31 1046 63 .
  18. 18. Conti E. Andreotti F. Sestito A. Riccardi P. Menini E. Crea F. Maseri A. Lanza G. A. 2002 Reduced levels of insulin-like growth factor-1 in patients with angina pectoris, positive exercise stress test, and angiographically normal epicardial coronary arteries. Am J Cardiol 89 973 5 .
  19. 19. Cook D. G. Leverenz J. B. Mc Millan P. J. Kulstad J. J. Ericksen S. Roth R. A. Schellenberg G. D. Jin L. W. Kovacina K. S. Craft S. 2003 Reduced hippocampal insulin-degrading enzyme in late-onset Alzheimer’s disease is associated with the apolipoprotein E-epsilon4 allele. Am J Pathol 162 313 9 .
  20. 20. Costantini C. Scrable H. Puglielli L. 2006 An aging pathway controls the TrkA to 75NTR receptor switch and amyloid beta-peptide generation. EMBO J 25, 1997-2006.
  21. 21. Coulson E. J. 2006 Does the 75 neurotrophin receptor mediate Abeta-induced toxicity in Alzheimer’s disease? J Neurochem 98, 654-60.
  22. 22. Craft S. 2005 Insulin resistance syndrome and Alzheimer’s disease: age- and obesity-related effects on memory, amyloid, and inflammation. Neurobiol Aging 26 Suppl 1 65 9 .
  23. 23. Craft S. 2007 Insulin resistance and Alzheimer’s disease pathogenesis: potential mechanisms and implications for treatment. Curr Alzheimer Res 4 147 52 .
  24. 24. Craft S. Asthana S. Cook D. G. Baker L. D. Cherrier M. Purganan K. Wait C. Petrova A. Latendresse S. Watson G. S. et al. 2003 Insulin dose-response effects on memory and plasma amyloid precursor protein in Alzheimer’s disease: interactions with apolipoprotein E genotype. Psychoneuroendocrinology 28 809 22 .
  25. 25. Craft S. Asthana S. Newcomer J. W. Wilkinson C. W. Matos I. T. Baker L. D. Cherrier M. Lofgreen C. Latendresse S. Petrova A. et al. 1999 Enhancement of memory in Alzheimer disease with insulin and somatostatin, but not glucose. Arch Gen Psychiatry 56 1135 40.
  26. 26. d’Abramo C. Ricciarelli R. Pronzato M. A. Davies P. 2006 Troglitazone, a peroxisome proliferator-activated receptor-gamma agonist, decreases tau phosphorylation in CHOtau4R cells. J Neurochem 98 1068 77 .
  27. 27. de la Monte S. M. 2009 Insulin resistance and Alzheimer’s disease. BMB Rep 42 475 81 .
  28. 28. de la Monte S. M. Wands J. R. 2006 Molecular indices of oxidative stress and mitochondrial dysfunction occur early and often progress with severity of Alzheimer’s disease. J Alzheimers Dis 9 167 81 .
  29. 29. Deane R. Du Yan. S. Submamaryan R. K. La Rue B. Jovanovic S. Hogg E. Welch D. Manness L. Lin C. Yu J. et al. 2003 RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat Med 9 907 13 .
  30. 30. Devaskar S. U. Giddings S. J. Rajakumar P. A. Carnaghi L. R. Menon R. K. Zahm D. S. 1994 Insulin gene expression and insulin synthesis in mammalian neuronal cells. J Biol Chem 269 8445 54 .
  31. 31. Doble B. W. Woodgett J. R. 2003 GSK-3: tricks of the trade for a multi-tasking kinase. J Cell Sci 116 1175 86 .
  32. 32. Drewes G. Lichtenberg-Kraag B. Doring F. Mandelkow E. M. Biernat J. Goris J. Doree M. Mandelkow E. 1992 Mitogen activated protein (MAP) kinase transforms tau protein into an Alzheimer-like state. EMBO J 11 2131 8 .
  33. 33. Drewes G. Mandelkow E. M. Baumann K. Goris J. Merlevede W. Mandelkow E. 1993 Dephosphorylation of tau protein and Alzheimer paired helical filaments by calcineurin and phosphatase-2A. FEBS Lett 336 425 32 .
  34. 34. Eckman E. A. Watson M. Marlow L. Sambamurti K. Eckman C. B. 2003 Alzheimer’s disease beta-amyloid peptide is increased in mice deficient in endothelin-converting enzyme. J Biol Chem 278 2081 4 .
  35. 35. Edbauer D. Winkler E. Regula J. T. Pesold B. Steiner H. Haass C. 2003 Reconstitution of gamma-secretase activity. Nat Cell Biol 5 486 8 .
  36. 36. Eikelenboom P. Bate C. Van Gool W. A. Hoozemans J. J. Rozemuller J. M. Veerhuis R. Williams A. 2002 Neuroinflammation in Alzheimer’s disease and prion disease. Glia 40 232 9 .
  37. 37. Farris W. Mansourian S. Chang Y. Lindsley L. Eckman E. A. Frosch M. P. Eckman C. B. Tanzi R. E. Selkoe D. J. Guenette S. 2003 Insulin-degrading enzyme regulates the levels of insulin, amyloid beta-protein, and the beta-amyloid precursor protein intracellular domain in vivo. Proc Natl Acad Sci U S A 100 4162 7 .
  38. 38. Figlewicz D. P. Szot P. Israel P. A. Payne C. Dorsa D. M. 1993 Insulin reduces norepinephrine transporter mRNA in vivo in rat locus coeruleus. Brain Res 602 161 4 .
  39. 39. Florant G. L. Singer L. Scheurink A. J. Park C. R. Richardson R. D. Woods S. C. 1991 Intraventricular insulin reduces food intake and body weight of marmots during the summer feeding period. Physiol Behav 49 335 8 .
  40. 40. Freude S. Plum L. Schnitker J. Leeser U. Udelhoven M. Krone W. Bruning J. C. Schubert M. 2005 Peripheral hyperinsulinemia promotes tau phosphorylation in vivo. Diabetes 54 3343 8 .
  41. 41. Frolich L. Blum-Degen D. Bernstein H. G. Engelsberger S. Humrich J. Laufer S. Muschner D. Thalheimer A. Turk A. Hoyer S. et al. 1998 Brain insulin and insulin receptors in aging and sporadic Alzheimer’s disease. J Neural Transm 105 423 38 .
  42. 42. Gabbita S. P. Lovell M. A. Markesbery W. R. 1998 Increased nuclear DNA oxidation in the brain in Alzheimer’s disease. J Neurochem 71 2034 40 .
  43. 43. Gasparini L. Gouras G. K. Wang R. Gross R. S. Beal M. F. Greengard P. Xu H. 2001 Stimulation of beta-amyloid precursor protein trafficking by insulin reduces intraneuronal beta-amyloid and requires mitogen-activated protein kinase signaling. J Neurosci 21 2561 70 .
  44. 44. Gibson G. E. Peterson C. Jenden D. J. 1981 Brain acetylcholine synthesis declines with senescence. Science 213 674 6 .
  45. 45. Goedert M. Hasegawa M. Jakes R. Lawler S. Cuenda A. Cohen P. 1997 Phosphorylation of microtubule-associated protein tau by stress-activated protein kinases. FEBS Lett 409 57 62 .
  46. 46. Gogolla N. Galimberti I. Deguchi Y. Caroni P. 2009 Wnt signaling mediates experience-related regulation of synapse numbers and mossy fiber connectivities in the adult hippocampus. Neuron 62 510 25 .
  47. 47. Gomez J. M. 2008 Growth hormone and insulin-like growth factor-I as an endocrine axis in Alzheimer’s disease. Endocr Metab Immune Disord Drug Targets 8 143 51 .
  48. 48. Gouras G. K. Almeida C. G. Takahashi R. H. 2005 Intraneuronal Abeta accumulation and origin of plaques in Alzheimer’s disease. Neurobiol Aging26 1235 44 .
  49. 49. Han S. H. Kim Y. H. Mook-Jung I. 2011 RAGE: The beneficial and deleterious effects by diverse mechanisms of actions. Mol Cells 31 91 7 .
  50. 50. Hanger D. P. Hughes K. Woodgett J. R. Brion J. P. Anderton B. H. 1992 Glycogen synthase kinase-3 induces Alzheimer’s disease-like phosphorylation of tau: generation of paired helical filament epitopes and neuronal localisation of the kinase. Neurosci Lett 147 58 62 .
  51. 51. Heidenreich K. A. Zahniser N. R. Berhanu P. Brandenburg D. Olefsky J. M. 1983 Structural differences between insulin receptors in the brain and peripheral target tissues. J Biol Chem 258 8527 30 .
  52. 52. Heneka M. T. Galea E. Gavriluyk V. Dumitrescu-Ozimek L. Daeschner J. O’Banion M. K. Weinberg G. Klockgether T. Feinstein D. L. 2002 Noradrenergic depletion potentiates beta-amyloid-induced cortical inflammation: implications for Alzheimer’s disease. J Neurosci 22 2434 42 .
  53. 53. Heneka M. T. Sastre M. Dumitrescu-Ozimek L. Hanke A. Dewachter I. Kuiperi C. O’Banion K. Klockgether T. Van Leuven F. Landreth G. E. 2005 Acute treatment with the PPARgamma agonist pioglitazone and ibuprofen reduces glial inflammation and Abeta1-42 levels in APPV717I transgenic mice. Brain 128 1442 53 .
  54. 54. Hirai K. Aliev G. Nunomura A. Fujioka H. Russell R. L. Atwood C. S. Johnson A. B. Kress Y. Vinters H. V. Tabaton M. et al. 2001 Mitochondrial abnormalities in Alzheimer’s disease. J Neurosci 21 3017 23 .
  55. 55. Ho L. Qin W. Pompl P. N. Xiang Z. Wang J. Zhao Z. Peng Y. Cambareri G. Rocher A. Mobbs C. V. et al. 2004 Diet-induced insulin resistance promotes amyloidosis in a transgenic mouse model of Alzheimer’s disease. FASEB J 18 902 4 .
  56. 56. Hoozemans J. J. Veerhuis R. Rozemuller A. J. Eikelenboom P. 2002 The pathological cascade of Alzheimer’s disease: the role of inflammation and its therapeutic implications. Drugs Today (Barc) 38 429 43 .
  57. 57. Hotamisligil G. S. Peraldi P. Budavari A. Ellis R. White M. F. Spiegelman B. M. 1996 IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science 271 665 8 .
  58. 58. Hoyer S. Nitsch R. 1989 Cerebral excess release of neurotransmitter amino acids subsequent to reduced cerebral glucose metabolism in early-onset dementia of Alzheimer type. J Neural Transm 75 227 32 .
  59. 59. Huang C. C. Lee C. C. Hsu K. S. 2010 The role of insulin receptor signaling in synaptic plasticity and cognitive function. Chang Gung Med J 33 115 25 .
  60. 60. Huang C. C. You J. L. Lee C. C. Hsu K. S. 2003 Insulin induces a novel form of postsynaptic mossy fiber long-term depression in the hippocampus. Mol Cell Neurosci 24 831 41 .
  61. 61. Iqbal K. Alonso Adel. C. Chen S. Chohan M. O. El -Akkad E. Gong C. X. Khatoon S. Li B. Liu F. Rahman A. et al. 2005 Tau pathology in Alzheimer disease and other tauopathies. Biochim Biophys Acta 1739 198 210 .
  62. 62. Iwata N. Tsubuki S. Takaki Y. Watanabe K. Sekiguchi M. Hosoki E. Kawashima-Morishima M. Lee H. J. Hama E. Sekine-Aizawa Y. et al. 2000 Identification of the major Abeta1-42-degrading catabolic pathway in brain parenchyma: suppression leads to biochemical and pathological deposition. Nat Med 6 143 50 .
  63. 63. Jager J. Gremeaux T. Cormont M. Le Marchand-Brustel Y. Tanti J. F. 2007 Interleukin-1beta-induced insulin resistance in adipocytes through down-regulation of insulin receptor substrate-1 expression. Endocrinology 148 241 51 .
  64. 64. Jagust W. J. Seab J. P. Huesman R. H. Valk P. E. Mathis C. A. Reed B. R. Coxson P. G. Budinger T. F. 1991 Diminished glucose transport in Alzheimer’s disease: dynamic PET studies. J Cereb Blood Flow Metab 11 323 30 .
  65. 65. Jiang Q. Heneka M. Landreth G. E. 2008 The role of peroxisome proliferator-activated receptor-gamma (PPARgamma) in Alzheimer’s disease: therapeutic implications. CNS Drugs 22 1 14 .
  66. 66. Johnston A. M. Pirola L. Van Obberghen E. 2003 Molecular mechanisms of insulin receptor substrate protein-mediated modulation of insulin signalling. FEBS Lett 546 32 6 .
  67. 67. Jones A. Kulozik P. Ostertag A. Herzig S. 2009 Common pathological processes and transcriptional pathways in Alzheimer’s disease and type 2 diabetes. J Alzheimers Dis 16 787 808 .
  68. 68. Kern W. Born J. Schreiber H. Fehm H. L. 1999 Central nervous system effects of intranasally administered insulin during euglycemia in men. Diabetes 48 557 63 .
  69. 69. Kitamura T. Kitamura Y. Kuroda S. Hino Y. Ando M. Kotani K. Konishi H. Matsuzaki H. Kikkawa U. Ogawa W. et al. 1999 Insulin-induced phosphorylation and activation of cyclic nucleotide phosphodiesterase 3B by the serine-threonine kinase Akt. Mol Cell Biol 19 6286 96 .
  70. 70. Kojima M. Hosoda H. Date Y. Nakazato M. Matsuo H. Kangawa K. 1999 Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 402 656 60 .
  71. 71. Kopf S. R. Baratti C. M. 1999 Effects of posttraining administration of insulin on retention of a habituation response in mice: participation of a central cholinergic mechanism. Neurobiol Learn Mem 71 50 61 .
  72. 72. Krogh-Madsen R. Plomgaard P. Keller P. Keller C. Pedersen B. K. 2004 Insulin stimulates interleukin-6 and tumor necrosis factor-alpha gene expression in human subcutaneous adipose tissue. Am J Physiol Endocrinol Metab 286, E234 8 .
  73. 73. Lehmann D. J. Cortina-Borja M. Warden D. R. Smith A. D. Sleegers K. Prince J. A. van Duijn C. M. Kehoe P. G. 2005 Large meta-analysis establishes the ACE insertion-deletion polymorphism as a marker of Alzheimer’s disease. Am J Epidemiol 162 305 17 .
  74. 74. Lesort M. Johnson G. V. 2000 Insulin-like growth factor-1 and insulin mediate transient site-selective increases in tau phosphorylation in primary cortical neurons. Neuroscience 99 305 16 .
  75. 75. Lesort M. Jope R. S. Johnson G. V. 1999 Insulin transiently increases tau phosphorylation: involvement of glycogen synthase kinase-3beta and Fyn tyrosine kinase. J Neurochem 72 576 84 .
  76. 76. Li Y. Barger S. W. Liu L. Mrak R. E. Griffin W. S. 2000 S100beta induction of the proinflammatory cytokine interleukin-6 in neurons. J Neurochem 74 143 50 .
  77. 77. Liu F. Grundke-Iqbal I. Iqbal K. Gong C. X. 2005 Contributions of protein phosphatases 1 PP2A, PP2B and PP5 to the regulation of tau phosphorylation. Eur J Neurosci 22, 1942-50.
  78. 78. Lizcano J. M. Alessi D. R. 2002 The insulin signalling pathway. Curr Biol 12, R236 8 .
  79. 79. Luchsinger J. A. Tang M. X. Shea S. Mayeux R. 2004 Hyperinsulinemia and risk of Alzheimer disease. Neurology 63 1187 92 .
  80. 80. Maccioni R. B. Farias G. Morales I. Navarrete L. 2010 The revitalized tau hypothesis on Alzheimer’s disease. Arch Med Res 41 226 31 .
  81. 81. Man H. Y. Lin J. W. Ju W. H. Ahmadian G. Liu L. Becker L. E. Sheng M. Wang Y. T. 2000 Regulation of AMPA receptor-mediated synaptic transmission by clathrin-dependent receptor internalization. Neuron 25 649 62 .
  82. 82. Markesbery W. R. Carney J. M. 1999 Oxidative alterations in Alzheimer’s disease. Brain Pathol 9 133 46 .
  83. 83. Mc Geer E. G. Mc Geer P. L. 2003 Inflammatory processes in Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry 27 741 9 .
  84. 84. Mecocci P. Mac Garvey. U. Beal M. F. 1994 Oxidative damage to mitochondrial DNA is increased in Alzheimer’s disease. Ann Neurol 36 747 51 .
  85. 85. Messier C. White N. M. 1987 Memory improvement by glucose, fructose, and two glucose analogs: a possible effect on peripheral glucose transport. Behav Neural Biol 48 104 27 .
  86. 86. Moon M. Choi J. G. Nam D. W. Hong H. S. Choi Y. J. Oh M. S. Mook-Jung I. 2011 Ghrelin ameliorates cognitive dysfunction and neurodegeneration in intrahippocampal amyloid-beta1-42 oligomer-injected mice. J Alzheimers Dis 23 147 59 .
  87. 87. Morishima-Kawashima M. Hasegawa M. Takio K. Suzuki M. Yoshida H. Titani K. Ihara Y. 1995 Proline-directed and non-proline-directed phosphorylation of PHF-tau. J Biol Chem 270 823 9 .
  88. 88. Nalivaeva N. N. Fisk L. R. Belyaev N. D. Turner A. J. 2008 Amyloid-degrading enzymes as therapeutic targets in Alzheimer’s disease. Curr Alzheimer Res 5 212 24 .
  89. 89. Neumann K. F. Rojo L. Navarrete L. P. Farias G. Reyes P. Maccioni R. B. 2008 Insulin resistance and Alzheimer’s disease: molecular links & clinical implications. Curr Alzheimer Res 5 438 47 .
  90. 90. Nunomura A. Perry G. Aliev G. Hirai K. Takeda A. Balraj E. K. Jones P. K. Ghanbari H. Wataya T. Shimohama S. et al. 2001 Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol 60 759 67 .
  91. 91. Nunomura A. Perry G. Pappolla M. A. Wade R. Hirai K. Chiba S. Smith M. A. 1999 RNA oxidation is a prominent feature of vulnerable neurons in Alzheimer’s disease. J Neurosci 19 1959 64 .
  92. 92. Ott A. Stolk R. P. Hofman A. van Harskamp F. Grobbee D. E. Breteler M. M. 1996 Association of diabetes mellitus and dementia: the Rotterdam Study. Diabetologia 39 1392 7 .
  93. 93. Ott A. Stolk R. P. van Harskamp F. Pols H. A. Hofman A. Breteler M. M. 1999 Diabetes mellitus and the risk of dementia: The Rotterdam Study. Neurology 53 1937 42 .
  94. 94. Ozcan U. Cao Q. Yilmaz E. Lee A. H. Iwakoshi N. N. Ozdelen E. Tuncman G. Gorgun C. Glimcher L. H. Hotamisligil G. S. 2004 Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 306 457 61 .
  95. 95. Papassotiropoulos A. Hock C. Nitsch R. M. 2001 Genetics of interleukin 6: implications for Alzheimer’s disease. Neurobiol Aging 22 863 71 .
  96. 96. Park C. R. Seeley R. J. Craft S. Woods S. C. 2000 Intracerebroventricular insulin enhances memory in a passive-avoidance task. Physiol Behav 68 509 14 .
  97. 97. Pedersen W. A. Mc Millan P. J. Kulstad J. J. Leverenz J. B. Craft S. Haynatzki G. R. 2006 Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Exp Neurol 199 265 73 .
  98. 98. Peters A. 2011 The selfish brain: Competition for energy resources. Am J Hum Biol 23 29 34 .
  99. 99. Phiel C. J. Wilson C. A. Lee V. M. Klein P. S. 2003 GSK-3alpha regulates production of Alzheimer’s disease amyloid-beta peptides. Nature 423 435 9 .
  100. 100. Plum L. Schubert M. Bruning J. C. 2005 The role of insulin receptor signaling in the brain. Trends Endocrinol Metab 16 59 65 .
  101. 101. Pritchard S. M. Dolan P. J. Vitkus A. Johnson G. V. 2011 The Toxicity of Tau in Alzheimer Disease: Turnover, Targets and Potential Therapeutics. J Cell Mol Med.
  102. 102. Profenno L. A. Porsteinsson A. P. Faraone S. V. 2010 Meta-analysis of Alzheimer’s disease risk with obesity, diabetes, and related disorders. Biol Psychiatry 67 505 12 .
  103. 103. Proud C. G. 2006 Regulation of protein synthesis by insulin. Biochem Soc Trans 34 213 6 .
  104. 104. Puglielli L. Ellis B. C. Saunders A. J. Kovacs D. M. 2003 Ceramide stabilizes beta-site amyloid precursor protein-cleaving enzyme 1 and promotes amyloid beta-peptide biogenesis. J Biol Chem 278 19777 83 .
  105. 105. Qiu W. Q. Folstein M. F. 2006 Insulin, insulin-degrading enzyme and amyloid-beta peptide in Alzheimer’s disease: review and hypothesis. Neurobiol Aging 27 190 8 .
  106. 106. Rasmussen M. H. Hvidberg A. Juul A. Main K. M. Gotfredsen A. Skakkebaek N. E. Hilsted J. Skakkebae N. E. 1995 Massive weight loss restores 24-hour growth hormone release profiles and serum insulin-like growth factor-I levels in obese subjects. J Clin Endocrinol Metab 80 1407 15 .
  107. 107. Reger M. A. Watson G. S. Green P. S. Wilkinson C. W. Baker L. D. Cholerton B. Fishel M. A. Plymate S. R. Breitner J. C. De Groodt W. et al. 2008 Intranasal insulin improves cognition and modulates beta-amyloid in early AD. Neurology 70 440 8 .
  108. 108. Risner M. E. Saunders A. M. Altman J. F. Ormandy G. C. Craft S. Foley I. M. Zvartau-Hind M. E. Hosford D. A. Roses A. D. 2006 Efficacy of rosiglitazone in a genetically defined population with mild-to-moderate Alzheimer’s disease. Pharmacogenomics J 6 246 54 .
  109. 109. Roden M. Price T. B. Perseghin G. Petersen K. F. Rothman D. L. Cline G. W. Shulman G. I. 1996 Mechanism of free fatty acid-induced insulin resistance in humans. J Clin Invest 97 2859 65 .
  110. 110. Rogers J. Lue L. F. 2001 Microglial chemotaxis, activation, and phagocytosis of amyloid beta-peptide as linked phenomena in Alzheimer’s disease. Neurochem Int 39 333 40 .
  111. 111. Rojo L. E. Fernandez J. A. Maccioni A. A. Jimenez J. M. Maccioni R. B. 2008 Neuroinflammation: implications for the pathogenesis and molecular diagnosis of Alzheimer’s disease. Arch Med Res 39 1 16 .
  112. 112. Ronnemaa E. Zethelius B. Sundelof J. Sundstrom J. Degerman-Gunnarsson M. Berne C. Lannfelt L. Kilander L. 2008 Impaired insulin secretion increases the risk of Alzheimer disease. Neurology 71 1065 71 .
  113. 113. Rulifson E. J. Kim S. K. Nusse R. 2002 Ablation of insulin-producing neurons in flies: growth and diabetic phenotypes. Science 296 1118 20 .
  114. 114. Saltiel A. R. Kahn C. R. 2001 Insulin signalling and the regulation of glucose and lipid metabolism. Nature 414 799 806 .
  115. 115. Saltiel A. R. Pessin J. E. 2002 Insulin signaling pathways in time and space. Trends Cell Biol 12 65 71 .
  116. 116. Sastre M. Dewachter I. Landreth G. E. Willson T. M. Klockgether T. van Leuven F. Heneka M. T. 2003 Nonsteroidal anti-inflammatory drugs and peroxisome proliferator-activated receptor-gamma agonists modulate immunostimulated processing of amyloid precursor protein through regulation of beta-secretase. J Neurosci 23 9796 804 .
  117. 117. Sato R. 2010 Sterol metabolism and SREBP activation. Arch Biochem Biophys 501 177 81 .
  118. 118. Sato T. Hanyu H. Hirao K. Kanetaka H. Sakurai H. Iwamoto T. 2009 Efficacy of PPAR-gamma agonist pioglitazone in mild Alzheimer disease. Neurobiol Aging.
  119. 119. Sayre L. M. Zelasko D. A. Harris P. L. Perry G. Salomon R. G. Smith M. A. 1997 4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased in Alzheimer’s disease. J Neurochem 68 2092 7 .
  120. 120. Scherer T. O’Hare J. Diggs-Andrews K. Schweiger M. Cheng B. Lindtner C. Zielinski E. Vempati P. Su K. Dighe S. et al. 2011 Brain insulin controls adipose tissue lipolysis and lipogenesis. Cell Metab 13 183 94 .
  121. 121. Schinner S. Scherbaum W. A. Bornstein S. R. Barthel A. 2005 Molecular mechanisms of insulin resistance. Diabet Med 22 674 82 .
  122. 122. Schrijvers E. M. Witteman J. C. Sijbrands E. J. Hofman A. Koudstaal P. J. Breteler M. M. 2010 Insulin metabolism and the risk of Alzheimer disease: the Rotterdam Study. Neurology 75 1982 7 .
  123. 123. Schubert M. Brazil D. P. Burks D. J. Kushner J. A. Ye J. Flint C. L. Farhang-Fallah J. Dikkes P. Warot X. M. Rio C. et al. 2003 Insulin receptor substrate-2 deficiency impairs brain growth and promotes tau phosphorylation. J Neurosci 23 7084 92 .
  124. 124. Schubert M. Gautam D. Surjo D. Ueki K. Baudler S. Schubert D. Kondo T. Alber J. Galldiks N. Kustermann E. et al. 2004 Role for neuronal insulin resistance in neurodegenerative diseases. Proc Natl Acad Sci U S A 101 3100 5 .
  125. 125. Schulingkamp R. J. Pagano T. C. Hung D. Raffa R. B. 2000 Insulin receptors and insulin action in the brain: review and clinical implications. Neurosci Biobehav Rev 24 855 72 .
  126. 126. Shen C. Chen Y. Liu H. Zhang K. Zhang T. Lin A. Jing N. 2008 Hydrogen peroxide promotes Abeta production through JNK-dependent activation of gamma-secretase. J Biol Chem 283 17721 30 .
  127. 127. Shibata M. Yamada S. Kumar S. R. Calero M. Bading J. Frangione B. Holtzman D. M. Miller C. A. Strickland D. K. Ghiso J. et al. 2000 Clearance of Alzheimer’s amyloid-ss(1-40) peptide from brain by LDL receptor-related protein-1 at the blood-brain barrier. J Clin Invest 106 1489 99 .
  128. 128. Shimomura I. Bashmakov Y. Ikemoto S. Horton J. D. Brown M. S. Goldstein J. L. 1999 Insulin selectively increases SREBP-1c mRNA in the livers of rats with streptozotocin-induced diabetes. Proc Natl Acad Sci U S A 96 13656 61 .
  129. 129. Smith C. D. Carney J. M. Starke-Reed P. E. Oliver C. N. Stadtman E. R. Floyd R. A. Markesbery W. R. 1991 Excess brain protein oxidation and enzyme dysfunction in normal aging and in Alzheimer disease. Proc Natl Acad Sci U S A 88 10540 3 .
  130. 130. Smith M. A. Perry G. Richey P. L. Sayre L. M. Anderson V. E. Beal M. F. Kowall N. 1996 Oxidative damage in Alzheimer’s. Nature 382 120 1 .
  131. 131. Smits H. A. Rijsmus A. van Loon J. H. Wat J. W. Verhoef J. Boven L. A. Nottet H. S. 2002 Amyloid-beta-induced chemokine production in primary human macrophages and astrocytes. J Neuroimmunol 127 160 8 .
  132. 132. Soop M. Duxbury H. Agwunobi A. O. Gibson J. M. Hopkins S. J. Childs C. Cooper R. G. Maycock P. Little R. A. Carlson G. L. 2002 Euglycemic hyperinsulinemia augments the cytokine and endocrine responses to endotoxin in humans. Am J Physiol Endocrinol Metab 282, E1276 85 .
  133. 133. Sotthibundhu A. Sykes A. M. Fox B. Underwood C. K. Thangnipon W. Coulson E. J. 2008 Beta-amyloid(1-42) induces neuronal death through the 75 neurotrophin receptor. J Neurosci 28, 3941-6.
  134. 134. Squire L. R. 1986 Mechanisms of memory. Science 232 1612 9 .
  135. 135. Steen E. Terry B. M. Rivera E. J. Cannon J. L. Neely T. R. Tavares R. Xu X. J. Wands J. ands, J. R. and de la Monte, S. M. 2005 Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease--is this type 3 diabetes? J Alzheimers Dis 7 63 80 .
  136. 136. Strachan M. W. Deary I. J. Ewing F. M. Frier B. M. 1997 Is type II diabetes associated with an increased risk of cognitive dysfunction? A critical review of published studies. Diabetes Care 20 438 45 .
  137. 137. Tansey M. G. Mc Coy M. K. Frank-Cannon T. C. 2007 Neuroinflammatory mechanisms in Parkinson’s disease: potential environmental triggers, pathways, and targets for early therapeutic intervention. Exp Neurol 208 1 25 .
  138. 138. Tchelingerian J. L. Le Saux F. Jacque C. 1996 Identification and topography of neuronal cell populations expressing TNF alpha and IL-1 alpha in response to hippocampal lesion. J Neurosci Res 43 99 106 .
  139. 139. Twickler M. T. Cramer M. J. Koppeschaar H. P. 2003 Unraveling Reaven’s syndrome X: serum insulin-like growth factor-I and cardiovascular disease. Circulation 107, e190 2 ; author reply e190-2.
  140. 140. van der Heide L. P. Kamal A. Artola A. Gispen W. H. Ramakers G. M. 2005 Insulin modulates hippocampal activity-dependent synaptic plasticity in a N-methyl-d-aspartate receptor and phosphatidyl-inositol-3-kinase-dependent manner. J Neurochem 94 1158 66 .
  141. 141. Van Obberghen E. Baron V. Delahaye L. Emanuelli B. Filippa N. Giorgetti-Peraldi S. Lebrun P. Mothe-Satney I. Peraldi P. Rocchi S. et al. 2001 Surfing the insulin signaling web. Eur J Clin Invest 31 966 77 .
  142. 142. Vassar R. Bennett B. D. Babu-Khan S. Kahn S. Mendiaz E. A. Denis P. Teplow D. B. Ross S. Amarante P. Loeloff R. et al. 1999 Beta-secretase cleavage of Alzheimer’s amyloid precursor protein by the transmembrane aspartic protease BACE. Science 286 735 41 .
  143. 143. Vuorinen-Markkola H. Yki-Jarvinen H. 1994 Hyperuricemia and insulin resistance. J Clin Endocrinol Metab 78 25 9 .
  144. 144. Wang S. Wang R. Chen L. Bennett D. A. Dickson D. W. Wang D. S. 2010 Expression and functional profiling of neprilysin, insulin-degrading enzyme, and endothelin-converting enzyme in prospectively studied elderly and Alzheimer’s brain. J Neurochem 115 47 57 .
  145. 145. Wang Y. T. Linden D. J. 2000 Expression of cerebellar long-term depression requires postsynaptic clathrin-mediated endocytosis. Neuron 25 635 47 .
  146. 146. Watson G. S. Cholerton B. A. Reger M. A. Baker L. D. Plymate S. R. Asthana S. Fishel M. A. Kulstad J. J. Green P. S. Cook D. G. et al. 2005 Preserved cognition in patients with early Alzheimer disease and amnestic mild cognitive impairment during treatment with rosiglitazone: a preliminary study. Am J Geriatr Psychiatry 13 950 8 .
  147. 147. Watson G. S. Craft S. 2003 The role of insulin resistance in the pathogenesis of Alzheimer’s disease: implications for treatment. CNS Drugs 17 27 45 .
  148. 148. Xie L. Helmerhorst E. Taddei K. Plewright B. Van Bronswijk W. Martins R. 2002 Alzheimer’s beta-amyloid peptides compete for insulin binding to the insulin receptor. J Neurosci 22, RC221 EOF .
  149. 149. Yaar M. Zhai S. Pilch P. F. Doyle S. M. Eisenhauer P. B. Fine R. E. Gilchrest B. A. 1997 Binding of beta-amyloid to the 75 neurotrophin receptor induces apoptosis. A possible mechanism for Alzheimer’s disease. J Clin Invest 100, 2333-40.
  150. 150. Yoon S. Y. Park J. S. Choi J. E. Choi J. M. Lee W. J. Kim S. W. Kim D. H. 2010 Rosiglitazone reduces tau phosphorylation via JNK inhibition in the hippocampus of rats with type 2 diabetes and tau transfected SH-SY5Y cells.Neurobiol Dis 40 449 55 .
  151. 151. Yu J. X. Bradt B. M. Cooper N. R. 2002 Constitutive expression of proinflammatory complement components by subsets of neurons in the central nervous system. J Neuroimmunol 123 91 101 .
  152. 152. Zhang H. Gao Y. Dai Z. Meng T. Tu S. Yan Y. 2011 IGF-1 reduces BACE-1 expression in PC12 cells via activation of PI3-K/Akt and MAPK/ERK1/2 signaling pathways. Neurochem Res 36 49 57 .
  153. 153. Zhao L. Teter B. Morihara T. Lim G. P. Ambegaokar S. S. Ubeda O. J. Frautschy S. A. Cole G. M. 2004 Insulin-degrading enzyme as a downstream target of insulin receptor signaling cascade: implications for Alzheimer’s disease intervention. J Neurosci 24 11120 6 .
  154. 154. Zhao W. Chen H. Xu H. Moore E. Meiri N. Quon M. J. Alkon D. L. 1999 Brain insulin receptors and spatial memory. Correlated changes in gene expression, tyrosine phosphorylation, and signaling molecules in the hippocampus of water maze trained rats. J Biol Chem 274 34893 902 .
  155. 155. Zhao W. Q. De Felice F. G. Fernandez S. Chen H. Lambert M. P. Quon M. J. Krafft G. A. Klein W. L. 2008 Amyloid beta oligomers induce impairment of neuronal insulin receptors. FASEB J 22 246 60 .

Written By

Sung Min Son, Hong Joon Shin and Inhee Mook-Jung

Submitted: 26 November 2010 Published: 04 November 2011