Diabetes mellitus and Alzheimer’s disease: GSK-3β as a potential link

Diabetes mellitus and Alzheimer’s disease: GSK-3β as a potential link

Accepted Manuscript Title: Diabetes mellitus and Alzheimer’s disease: GSK-3␤ as a potential link Authors: Ying Zhang, Nan-qu Huang, Fei Yan, Hai Jin, ...

675KB Sizes 0 Downloads 77 Views

Accepted Manuscript Title: Diabetes mellitus and Alzheimer’s disease: GSK-3␤ as a potential link Authors: Ying Zhang, Nan-qu Huang, Fei Yan, Hai Jin, Shao-yu Zhou, Jing-shan Shi, Feng Jin PII: DOI: Reference:

S0166-4328(17)31529-2 https://doi.org/10.1016/j.bbr.2017.11.015 BBR 11174

To appear in:

Behavioural Brain Research

Received date: Revised date: Accepted date:

13-9-2017 8-10-2017 13-11-2017

Please cite this article as: Zhang Ying, Huang Nan-qu, Yan Fei, Jin Hai, Zhou Shaoyu, Shi Jing-shan, Jin Feng.Diabetes mellitus and Alzheimer’s disease: GSK-3␤ as a potential link.Behavioural Brain Research https://doi.org/10.1016/j.bbr.2017.11.015 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Diabetes mellitus and Alzheimer's disease: GSK-3β as a potential link

IP T

Ying Zhang1†, Nan-qu Huang2†, Fei Yan1, Hai Jin3 , Shao-yu Zhou1,4, Jing-shan Shi1, Feng Jin1* 1. Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China 2. Drug Clinical Trial Institution, The First People’s Hospital of Zunyi, Guizhou, China 3. Institute of Digestive Diseases of Affiliated Hospital, Zunyi Medical University, Guizhou, China 4. Department of Environmental Health, Indiana University Bloomington, Indiana, United States. * Corresponding author. †

These authors have contributed equally to this work.

Highlights

SC R

E-mail address: [email protected].

TE D

M

A

N

U

• GSK-3β is involved in the physiological and pathological progress of DM and AD respectively. • In DM, GSK-3β is one of the key factors leading to insulin deficiency and insulin resistance. • In AD, GSK-3β plays an important role in hyperphosphorylation of microtubuleassociated protein tau. • GSK-3β as a potential link between DM and AD is reviewed. • GSK-3β as a link between DM and AD further supports that AD is regarded as Type 3 diabetes. Abstract: It is well known that Alzheimer’s disease (AD) is closely related to diabetes mellitus (DM), and AD is also regarded as Type 3 diabetes (T3D). However, the exact link between AD and DM is still unclear. Recently, more and more evidence has shown

EP

that glycogen synthase kinase-3β (GSK-3β) may be the potential link between DM and AD. In DM, GSK-3β is the crucial enzyme of glycogen synthesis, which plays a key

CC

role in regulating blood glucose. More importantly, GSK-3β is one of the key factors leading to insulin deficiency and insulin resistance, and insulin resistance is an

A

important hallmark of the occurrence and development of DM. In AD, GSK-3β plays an important role in hyperphosphorylation of microtubule-associated protein tau (tau), which is one of the pathological features in AD. GSK-3β is one of the important kinases of tau phosphorylation and is involved in the insulin/phosphoinositide 3-kinase/protein kinase B (insulin/PI3K/Akt) signaling pathway. Dysfunction of the insulin/PI3K/Akt signaling pathway, which regulates glucose metabolism in the brain, can lead to tau hyperphosphorylation in the brain of AD patents. Additionally, insulin resistance in DM

may cause β-amyloid (Aβ) deposition, which will be cleared by tau, but excessive phosphorylation of tau will further aggravate the neurotoxicity; then damage the brain and affect the cognitive function. GSK-3β is considered as a common kinase in insulin signaling transduction and tau protein phosphorylation, so we have reasons to believe that GSK-3β is a potential link between DM and AD.

A

N

U

SC R

IP T

Abbreviations: 3xTg, triple-transgenic; Aβ, β-amyloid; AD, Alzheimer’s disease; ADAS-cog, Alzheimer's Disease Assessment Scale-cognitive subscale; Akt, protein kinase B; APP, amyloid precursor protein; CREB, cAMP-response element binding protein; DM, diabetes mellitus; FFAs, free fatty acids; GCA, Global Clinical Assessment; GDS, Geriatric Depression Scale; GPCRs, G protein-coupled receptors; GS, glycogen synthase; GSK-3β, glycogen synthase kinase-3β; IGF-1, insulin-like growth factor-1; iNOS, inducible nitric oxide synthase; IRS-1, insulin receptor substrate-1; JNK, C-Jun-N terminal kinase; MMSE, Mini-Mental Status Examination; NFTs, neurofibrillary tangles; NOD, non-obese diabetic; PDK1, phosphoinositidedependent protein kinase 1; PDX-1, duodenal homeobox 1; PI3K, phosphoinositide 3kinase; PP-1, protein phosphatase 1; PS1, presenilin 1; SPs, senile plaques; STAT3, signal transduction and transcriptional activator 3; T3D, Type 3 diabetes; tau, microtubule-associated protein tau; UCP2, uncoupling protein 2.

M

Key words: glycogen synthase kinase-3β; diabetes mellitus; Alzheimer’s disease; microtubule-associated protein tau; insulin resistance Introduction

TE D

1.

AD is a neurodegenerative disease which is pathologically characterized by typically senile plaques (SPs) formed of Aβ deposition, and neurofibrillary tangles (NFTs) composed of tau hyperphosphorylation [1, 2]. With increasingly aging

EP

population, the incidence of AD has increased year by year, therefore, prevention and control of AD has become globally focused [3]. Although a lot of research has been

CC

carried out on AD, the etiology and pathogenesis of AD are not yet fully understood. In our previous study about icariin (ICA, extracted from Chinese herb Berberidaceae

A

epimedium L.) protecting amyloid precursor protein/presenilin 1 (APP/PS1) doubletransgenic AD mice, we also found that 14-month-old AD model mice had significantly higher blood glucose levels than that of wild-type mice of the same age [4]. This suggests that glucose metabolism disorder is associated with AD. In addition, Vandal M, et al. found that there was obvious glucose intolerance in the triple-transgenic mouse model of AD (3xTg-AD) [5]. But, a randomized, double-blind, placebo-controlled trial showed that in patients with early AD, it is very common that they are not diagnosed

with pre-diabetes and diabetes [6]. Interestingly, AD-related proteins will in turn play a role in the signaling pathway of insulin. Many researchers found that the APL-1, an APP-related protein, is encoded by Caenorhabditis elegans gene which also affects the signaling pathway of insulin in AD patients [7]. In addition, researchers from Tohoku University in Japan found that, the way insulin signals work in the brain may be similar to the way insulin signals work in the pancreas of DM patients, suggesting that AD may be described as a diabetic disorder in the brain [8]. Therefore, AD is considered to be

IP T

brain-specific T3D, which is similar to but different from DM [9, 10]. Although the

specific mechanism is still unclear, recent studies have found that GSK-3β associated

2.

SC R

with insulin signaling pathway may be a potential link between AD and DM. Role of GSK-3β in DM

DM is a group of metabolic diseases characterized by high blood sugar, and hyperglycemia is caused by insulin secretion deficiency or its biological effect’s

U

damage. GSK-3β as one of the first kinases capable of phosphorylating glycogen

N

synthase (GS), is isolated from rabbit skeletal muscle and is one of the few protein

A

kinases that are inactivated by phosphorylation [11]. Among them, the high expression of GSK-3β was correlated with the decrease of insulin sensitivity, and it was involved

M

in blood glucose regulation, insulin deficiency and insulin resistance. Therefore, more and more attention has been paid to the study of the relationship between GSK-3β and

TE D

the occurrence of DM.

2.1 GSK-3β in blood glucose regulation Blood glucose regulation is not only an important part of the regulation of life’s

EP

activities but also an important condition for homeostasis in the body. The disorder of blood glucose regulation increased the incidence of DM. Insulin-based regulation is still the main path of blood glucose regulation, in which the insulin receptor-mediated

CC

signaling pathway plays a key role [12]. PI3K/Akt/GSK-3β signaling pathway is involved in the insulin signaling transduction, and GSK-3β is regulated and controlled

A

by insulin in this signaling pathway, which is related to the glycogen synthesis regulation. The insulin receptor binds insulin and has a tyrosine-protein kinase activity, and mediates the metabolic functions of insulin. Binding to insulin stimulates the association of the receptor with downstream mediators, including insulin receptor substrate-1 (IRS-1) and PI3K, the insulin receptor can activate PI3K either directly by binding to the p85 regulatory subunit, which produces PIP3, directly or indirectly leading to phosphorylation and activation of Akt, afterwards Akt phosphorylates the

Ser9 site of GSK-3β and inhibits its activity [13, 14]. Then failing to inhibit the activity of glycogen synthase, which promotes glycogen synthesis and reduces the blood glucose level. When insulin and insulin-like growth factor-1 (IGF-1) signal dysfunction, the body increases GSK-3β activity to eventually increase blood glucose, by phosphorylation of Tyr216 site of GSK-3β to inhibit the activity of PI3K/Akt [15], and also by blockade of Wnt signaling pathway to negatively regulate GSK-3β through PI3K/Akt dependent mechanism [16] (Figure 1). Studies have shown that heat shock-

IP T

induced glycogen synthesis in L6 skeletal muscle cells leads to an increase in glycogen-

associated protein phosphatase 1 (PP-1) and GS activity, accompanied by sustained

SC R

Akt/GSK-3β phosphorylation. When Wortmannin (PI3K inhibitor) inhibits Akt/GSK3β phosphorylation, it prevents 2-deoxyglucose uptake, and eliminates heat shockinduced glycogen synthesis as well [17]. In the DM model mice, it has been found that

excessive activation of GSK-3β results in the decrease of islet β cell proliferation [18].

U

Islet β cells are endocrine cells that secrete insulin in the body, which can regulate blood

N

glucose levels. Endogenous GSK-3β controls the growth of islet β cells by feedback

blood glucose.

M

2.2 GSK-3β and insulin deficiency

A

inhibition of the PI3K/Akt signaling pathway, then plays a critical role in regulating

Congenital or acquired dysfunction of islet β cells can lead to inadequate insulin

TE D

secretion, causing DM. The exact mechanism is unclear, but it may be related to glucose toxicity, lipid toxicity, inflammatory response, oxidative stress and other factors [1921]. GSK-3β is one of the key factors that mediate islet β cells apoptosis and is closely

EP

related to insulin deficiency. A previous study found that hyperglycemia-induced loss of islet β cells is associated with increased oxidative stress [22]. Gliclazide is a sulfonylurea oral anti-diabetic drug preventing human islet β cells from apoptosis

CC

induced by high glucose, which was due, at least in part, to its antioxidant activity [23]. Under diabetic conditions, the increase in oxidative stress leads to the activation

A

of the C-Jun-N terminal kinase (JNK) pathway, further stimulating the phosphorylation of GSK-3β [24, 25], while GSK-3β hyperactivity has anti-proliferative and proapoptotic properties effects on the body. GSK-3β phosphorylation can inhibit the activity of cAMP-response element binding protein (CREB), and then down-regulate the expression of Bcl-2 [26, 27], activation of caspase-3 and caspase-9, leading to reduction of islet β cells, eventually speeding up the process of DM. In addition, inducible nitric oxide synthase (iNOS) as the main medium of inflammation, can

activate JNK and GSK-3β, and inhibit the expression of pancreatic and duodenal homeobox 1 (PDX-1) and IRS-2 protein, finally leading to islet β cell exhaustion [28, 29]. Increased blood glucose levels induce mitochondrial superoxide production and activation of uncoupling protein 2 (UCP2), and reduce the ATP/ADP ratio, leading to impaired β cell function and reduced insulin secretion [30]. Among them, GSK-3β, as one of the rate-limiting enzymes, is a key factor in ATP production that inhibits glycogen synthesis. In addition, glucose tolerance and islet β cells were proliferated

IP T

relative to the control group in β-Gsk-3β-/- mice [31]. Therefore, excessive activation of GSK-3β is associated with apoptosis of islet β cells and reduction of insulin

SC R

secretion, which may lead to DM (Figure 1). 2.3 GSK-3β and insulin resistance

Insulin resistance refers to the decrease of glucose uptake and utilization efficiency drops for various reasons. In order to maintain the stability of blood sugar in the body,

U

islet β cells compensate with excessive secretion of insulin and this eventually leads to

N

hyperinsulinemia. It’s easy to lead to metabolic syndrome and T2D. Cells regulate

A

blood glucose through insulin receptors binding to insulin, and insulin resistance means that the body's cells (mainly muscle) are less sensitive to insulin. Impaired signal

M

transduction of the insulin/PI3K/Akt signaling pathway is one of the main features of insulin resistance, and insulin resistance will further lead to an increase in GSK-3β [32,

TE D

33]. Yokoo et al. found the increased phosphorylation of GSK-3β-Ser9 by suppression of constitutively active GSK-3 in cultured bovine adrenal chromaffin cells, and the stability of insulin receptor mRNA was reduced, which in turn affected the level of insulin receptor [34]. In the study of the role of STAT3 (signal transduction and

EP

transcriptional activator 3) in insulin signaling, L-STAT-/- mice had an increased GSK3β level compared with the control group. It was also found that suppression of GSK-

CC

3β with lithium chloride and L803-mts can restore glucose balance and improve glucose intolerance [35]. In addition, evidence has shown that insulin resistance is associated

A

with hepatitis C virus (HCV) infection, and GSK-3β is also involved in it [36]. HCV E2 protein inhibits glucose uptake and glycogen synthesis by affecting the expression of IRS-1, Akt protein and GSK-3β phosphorylation in Huh7 cells, leading to insulin resistance [37]. It's interesting that inhibition of GSK-3β activity in DM mice can prevent DM-related pathological changes [38]. What’s more, there is another view. Some researchers have suggested that the root cause of T2D metabolism disorder is the disorder of fat metabolism. A large amount of

fat is deposited in muscle, liver and islet β cells, leading to insulin resistance and islet cell dysfunction. Adipose tissue-derived cytokines can cause systemic insulin resistance, and GSK-3β plays an important role in it [39, 40]. In 1994, Benjamin WB et al found that the GSK-3β content in the cytoplasm was reduced during the differentiation of 3T3-L1 cells into adipocytes [41]. It's interesting that the constructed GSK-3β-specific RNAi adenovirus vector was transfected into HEK293A cells, and the expression of GSK-3β was down-regulated, which reduced the effect of free fatty acids

IP T

(FFAs) on human umbilical vein endothelial cells injury [42]. GSK-3β-specific RNAi and lithium chloride also inhibits the activity of GSK-3β in human L02 and HepG2

SC R

cells, and inhibits the upregulation of JNK phosphorylation and Bax uptake by saturated FFAs sodium palmitate and reduces lipoapoptosis [43]. This indicates that GSK-3β is one of the important factors involved in lipid metabolism.

Studies have shown that plasma levels of FFAs are elevated in most obese subjects,

U

whereas physiological elevation of plasma FFAs leads to insulin-induced glucose

N

transport and/or phosphorylation deficits, which in turn inhibits glucose uptake [44].

A

Leonard H Storlien et al found that insulin sensitivity in rat skeletal muscle was negatively correlated with triglyceride levels in muscle, and high-fat diets could lead to

M

insulin resistance [45]. Subsequent studies have shown that changes in plasma FFAs concentrations mediate insulin resistance, plasma insulin levels, and glucose

TE D

intolerance [46]. This suggests that obesity and T2D are associated with insulin resistance. In obese individuals, adipose tissue can release a large number of FFAs, glycerol, hormones, proinflammatory cytokines and others involved in the development

EP

of insulin resistance [47]. Compared with the control group, the expression of GSK-3β protein in the adipose tissue of the insulin-resistant rats induced by high-fat diet was significantly increased [48]. In DM mice, intraperitoneal injection of melatonin can

CC

improve glucose tolerance and insulin resistance, and increase liver glycogen synthesis, which is related to the level of GSK-3β phosphorylation [49]. HepG2 cells were treated

A

with palmitate to induce insulin resistance, and NO-1886 (ibrolipim, a lipoprotein lipase activator) reduced GSK-3β protein levels and stimulated glycogen synthesis, improved insulin resistance [50]. Glucose and FFAs together promote the islet β cell’s apoptosis by activating GSK-3β. When GSK-3β expression is reduced, islet β cell apoptosis decreases and instead its proliferation increases, as does glycogen synthesis, and insulin resistance and fat metabolic disorders. In summary, GSK-3β plays an important role in insulin resistance caused by exogenous infection and endogenous lipid

metabolism disorders [36, 37, 49, 50] (Figure 2). 3.

GSK-3β, tau phosphorylation and AD The function of GSK-3 was originally thought to be the phosphorylation of only

GS and therefore inactivated GS. However, later, a large number of studies have shown that GSK-3 can also phosphorylate tau protein [51, 52]. Two subtypes of GSK-3 were found in mammals: GSK-3α and GSK-3β, the latter of which plays a key role in tau protein phosphorylation. Excessive activation of GSK-3β promotes abnormal

IP T

hyperphosphorylation of tau protein, aggravates degeneration of neurons, interferes with normal synaptic plasticity, and accelerates AD pathology process in AD patients

SC R

[53]. GSK-3β plays an important role in the pathological changes of tau protein in AD

[54]. The activation of GSK-3β was demonstrated to promote abnormal hyperphosphorylation of tau and was involved in the occurrence and development of AD [55]. Microtubules composed of microtubule-associated proteins and tubulin are

U

important components of the cytoskeleton, and axonal cytoskeleton is closely related

N

to axonal growth. Spittaels K et al found that co-expression of GSK-3β decreased

A

axonal dilatation and reduced axonal lesions in the CNS of tau transgenic mice [56]. A decrease in the activity of this kinase inhibits the increase in motile mitochondria, which

M

in turn regulates mitochondrial axonal transport [57]. Therefore, phosphorylation of tau by GSK-3β may be critical for axonal elongation during development, also GSK-3β

TE D

phosphorylates the tau protein at multiple sites in intact cells. Furthermore, GSK-3βmediated phosphorylation of Ser404 protects the drosophila visual system [58], and studies have shown that phosphorylation-incompetent tau protein is more serious for

EP

synaptic damage than hyperphosphorylated tau protein, and surprisingly found that phosphorylation can prolong the longevity of drosophila [59]. In a study published in the Science, Lars Ittner et al proposed a new view that the reason for tau

CC

phosphorylation is actually to protect neurons from damage. In the early stages of the disease, the protein assists in the protective phosphorylation of tau and interferes with

A

the neurotoxic complex formed by Aβ; and they suggest that tau protein phosphorylation initially has a protective effect on neurons, but Aβ can attack the protective function until the function is gradually lost. At this stage, the level of toxicity will lead to neuronal destruction and cognitive impairment associated with AD [60]. Tau phosphorylation plays an important role in the brain, which may be an important mechanism of repair and resistance to injury, but tau hyperphosphorylation is also an important factor in the formation of NFTs. It is quite necessary to control the

phosphorylation of tau in a reasonable range by regulating GSK-3β signaling pathway in AD. 3.1 GSK-3β, an important kinase for phosphorylation of tau protein In 1975, the tau protein was first identified as a thermostable protein necessary for microtubule assembly, which has been shown to have a characterization of native unfolding proteins [61-63]. The tau protein interacts with tubulin to stabilize the microtubules and promote tubulin assembly into microtubules. This study found that

IP T

the tau protein binds to the surface of the microtubule and acts as a velocity "controller" to regulate the transport of dynein and kinesin. This transport is necessary to transfer

SC R

other proteins from the neuronal synapses to the cell body and maintain healthy neurons

[64]. It has been reported that there are about 30 Ser and Thr phosphorylation sites in normal tau proteins [65], and it is worth mentioning that the normal phosphorylation of tau protein is a necessary condition for its normal biological function. As early as 1997,

U

Lesort M et al found that neuronal cells rich in phosphorylated tau protein were more

N

tolerant to apoptosis [66]. Tau protein phosphorylation is regulated by many kinases

A

and phosphatases, and GSK-3β is one of the most important phosphorylated kinases catalyzing tau protein, which plays a key role in regulating tau phosphorylation levels

M

and stabilizing microtubules [67]. GSK-3β is highly expressed in the brain tissues, especially in neurons, and its expression in developing brain tissue is much higher than

TE D

that in adult brain tissue [68, 69]. Moreover, the enzyme plays an important role in the growth of axons during brain development [70]. In addition, tau and GSK-3β coimmunoprecipitation were found by chromatographic separation, indicating that the

EP

kinase binds to tau in the brain, which in turn leads to GSK-3β through tau associated with microtubules [71]. 3.2 GSK-3β, mutant PS1 and tau hyperphosphorylation

CC

PS1 is an important component of γ-secretase, which is highly expressed in the

hippocampus, inner olfactory cortex, cortex and cerebellum. PS1 protein mutation is

A

thought to be closely related to the pathogenesis of AD [72]. Mutant PS1 is easily cleaved by caspases, which increases Aβ content in neurons and then regulates phosphorylation of microtubule-associated protein tau [73]. Previously, Murayama M et al found that overexpression of PS1 reduced the level of cytoplasmic β-catenin in transfected cells, but the consequences of this interaction were unknown [74]. Subsequently, studies have found that wild-type PS1-induced overexpression increases the binding of β-catenin to GSK-3β and accelerates the turnover of endogenous β-

catenin, suggesting that GSK-3β-dependent degradation of β-catenin requires PS1 expression. In contrast, PS1 mutants interfere with this process, which not only attenuate the interaction between GSK-3β and β-catenin, but also interfere with the conversion of β-catenin [75]. Studies have shown that tau and GSK-3β bind to the same region of PS1, and PS1 mutations increase the binding of tau to GSK-3β and its taudirected kinase activity, while the activation of GSK-3β further leads to sustained enhancement of tau hyperphosphorylation and NFTs formation [76]. Further studies

IP T

showed that there were Congo red birefringence and Thioflavin T reactivity in the

neurons of familial Alzheimer’s disease (FAD) mutant (I213T) PS1 knock-in mouse

SC R

which are both histologic criteria of NFTs, indicating that PS1 mutations accelerate the formation of tau pathology and lead to the pathogenesis of AD [77]. GSK-3β-mediated phosphorylation induced PS1 structure changes, reducing the interaction of PS1 with β-catenin, thereby causing the β-catenin to stabilize [78], also it has been demonstrated

U

that PS1 is a substrate of inactive GSK-3β [79]. When the mouse overexpresses GSK-

N

3β, the hippocampal neurons show a decrease in nuclear β-catenin levels, tau

A

hyperphosphorylation, and further neurodegeneration [80]. In summary, the mutant PS1 may induce GSK-3β overexpression, resulting in tau hyperphosphorylation (Figure

M

3).

3.3 PI3K/Akt/GSK-3β signaling pathway in tau hyperphosphorylation disorders

of

the

PI3K/Akt/GSK-3β

TE D

The

pathway

also

lead

to

tau

hyperphosphorylation. PI3K has the activity of Ser/Thr kinase as well as that of phosphatidylinositol kinase; when PI3K is activated by signals from tyrosine kinases

EP

and G protein-coupled receptors (GPCRs), a second messenger PIP3 is generated on the cell membrane; PIP3 binds to the N-terminal PH domain of Akt and promotes phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylated Akt protein on

CC

the Thr308 and Ser473, thereby activating Akt. GSK-3β is a downstream molecule of the PI3K/Akt signaling pathway. Activated Akt binds to GSK-3β, induces GSK-3β to

A

transpose into cell membrane, phosphorylates its N-terminal Ser9 active site and inactivates GSK-3β, which in turn affects the downstream substrate of GSK-3β such as tau protein and insulin receptor. Studies have shown that the PI3K/Akt/GSK-3β signaling pathway plays a key role in neuroprotection and enhances cell survival by stimulating cell proliferation and inhibiting apoptosis [81, 82]. However, dysfunction of this signaling pathway will increase GSK-3β activity and lead to excessive phosphorylation of tau. The expression of GSK-3β and the phosphorylation of the tau

protein were detected by transfection of GSK-3β into rat brain. It was found that the abnormal phosphorylation of tau protein was co-located with GSK-3β, indicating that GSK-3β-induced hyperphosphorylation of tau protein was associated with the mechanism of neurodegenerative disease [83]. When the rats were injected with propofol, Akt decreased, GSK-3β phosphorylation increased, and hippocampal apoptosis and cognitive deficits were detected. Pretreatment of dexmedetomidine (Dex) in rats can restore PI3K/Akt/GSK-3β signaling to normal and reduce hippocampal cell

IP T

apoptosis and improve cognitive dysfunction [84]. It has been shown that arctigenin (Arctigenin, a lignan from Wikstroemia indica) has neuroprotective effects on AD mice,

SC R

which can improve learning and memory deficiencies by increasing the

phosphorylation of PI3K/Akt, decreasing the activity of GSK-3β, and decreasing levels of tau phosphorylation of Thr181, Thr231 and Ser404 sites in the hippocampus [85]. The study further confirms that the PI3K/Akt/GSK-3β signaling pathway is the key to

U

regulating the phosphorylation of tau protein. In both in vivo, and in vitro, after

N

HEK293/tau cells were treated with H2O2, the level of tau phosphorylation in Thr231

A

and Ser396 sites increased, but the level of tau phosphorylation decreased to normal after HEK293/tau cells were treated with TPPU, a drug that has been shown to have

M

neuroprotective effects. Additionally, TPPU up-regulated the expression of p-Akt, while decreasing the phosphorylation of GSK-3β [86]. Moreover, the neuroprotective

TE D

effect of ginkgolides (Ginkgolides are biologically active terpenic lactones present in Ginkgo biloba) is also related to this pathway. Ginkgolide activates the PI3K/Akt signaling pathway to inhibit the phosphorylation of GSK3β at the Ser9 site, thereby

EP

protecting cells from tau hyperphosphorylation related toxicity [87]. Thus, both in vivo and in vitro the tau hyperphosphorylation is associated with the PI3K/Akt/GSK-3β signaling pathway, and inhibition of the GSK-3β over-activation may alleviate tau

CC

hyperphosphorylation, therefore slowing the further aggravation of AD. 4.

GSK-3β, a potential link between DM and AD

A

We have known that GSK-3β plays a key role in DM, being the key to the dynamic

regulation of blood glucose, and also being one of the key factors leading to insulin deficiency and insulin resistance. In clinical trials, it was found that hypoglycemic agents can help AD patients by improving their brain activity. DM is an independent risk factor for cognitive impairment, and its damage to the CNS is getting more and more attention. In recent years, epidemiological studies have shown that DM, especially T2D, increases the risk of AD [88], indicating that DM may play an important role in

the development of AD pathogenesis. A reduction in insulin release and a decrease in insulin levels are critical to the development and progression of both T2D and AD. When insulin signaling is impaired by insulin resistance, it leads to DM, AD and other neurological diseases [89], also the decrease of glucose metabolism in AD patients was observed by 18F-FDG PET technique. Further studies have shown that insulin resistance is associated with a significant reduction in glucose metabolism in the brain of patients with AD, which in turn increased learning and memory dysfunction in AD

IP T

patients [90]. 4.1 Insulin resistance and Aβ deposition

SC R

We already know that GSK-3β plays an important role in insulin resistance caused

by exogenous infection and endogenous lipid metabolism disorders. The close relationship between insulin resistance and Aβ deposition is now described in this paragraph. Insulin resistance may first influence the clearance of normal Aβ by

U

affecting mitochondrial function, which in turn affects the clearance of the deposited

N

Aβ by inflammatory cascades. Some studies have suggested that Aβ deposition

A

involved in immune defense and anti-infection [91], but the formation of an inflammatory microenvironment will break this beneficial mechanism. Under this

M

microenvironment, the body will mistakenly believe that a large amount of Aβ is required to be removed as the invasive substance. Interestingly, under normal

TE D

circumstances this will not have too much effect on the body. However, it will eventually lead to a large amount of deposition of Aβ with age, physical dysfunction, and a decrease of Aβ clearance caused by insulin resistance. DM is also considered as

EP

a chronic mild inflammation-related diseases, and more and more evidence suggests that DM and its complications such as diabetic nephropathy, diabetic retinopathy and diabetic vascular disease and others are closely related to inflammation [92-94]. In

CC

addition, inflammation in AD plays an important role in the regeneration and apoptosis of hippocampal neurons [95]. Apart from the secretases and lysosomes which are two

A

cleavage enzymes for the removal of APP, the erroneously deposited Aβ requires other pathways to be cleared. Experiments have shown that early inflammatory response to activation of microglia-mediated phagocytosis can remove Aβ [96, 97], but sustained and hyperactivated inflammation will further lead to chronic long-term deposition of Aβ, and eventually increase the pathological damage of AD [98]. The inflammatory microenvironment are gradually forming in patients with insulin resistance, and uncontrollable cascade reaction will damage the health of patients step by step. In

conclusion, GSK-3β-involved insulin resistance may aggravate Aβ deposition by multiple routes (Figure 3). 4.2 GSK-3β and tau hyperphosphorylation The traditional viewpoint is that Aβ deposition forms plaques, leading to tau protein hyperphosphorylation, further leading to the death of cells and AD. Lee HG et al challenged this classic concept, suggesting that tau protein phosphorylation is a compensatory response in the process of neuronal antioxidant stress as a protective

IP T

function for neurons [99, 100]. Initial tau phosphorylation may be a protective response,

but excessive phosphorylation of tau will further aggravate the injury, and Aβ will

SC R

attack this protective function until this function is gradually lost. At this stage the level

of toxicity can lead to AD associated neuronal destruction and cognitive impairment. Aβ deposition is toxic to neurons, but the first step of tau phosphorylation is actually to reduce this toxicity. Tau phosphorylation, which is involved in GSK-3β, plays an

U

important regulatory role in the brain, but the mutant PS1 may cause GSK-3β

N

overexpression, and thus hyperphosphorylated tau which is an important factor in the

A

formation of NFTs. In addition to that, insulin resistance and insulin signaling pathways also play an important role in the regulation of tau phosphorylation. In the study of non-

M

obese diabetic (NOD) model mice, insulin dysfunction was found to cause AD-like tau hyperphosphorylation in the brain [101]. When insulin was used to treat db/db diabetic

TE D

mice, it was found that it not only restored the phosphorylation levels of insulin receptors and GSK3β, but also improved the learning ability of mice, which is related to reductions in the level of tau phosphorylation in the brain [102]. Kim’s study show

EP

that moderate exercise can inhibited the hyperphosphorylation of tau in the hippocampus by inhibiting the expression of GSK-3β in diabetic rats and found that the incidence of AD was reduced and that the PI3K/Akt/GSK-3β pathway was involved in

CC

it [103]. In rats, reducing oxidative stress through the PI3K/Akt/GSK-3β signaling pathway can improve diabetes-induced cognitive impairment [104]. In addition,

A

autopsies were performed on patients with DM and/or AD, and the levels and activity of PI3K/Akt signaling pathway were reduced in their frontal cortex, with a large amount of phosphorylated GSK-3β and abnormal tau hyperphosphorylated proteins detected. Interestingly, the defects of this signaling pathway are more severe in individuals who suffer both DM and AD (T2DM-AD) [105]. Excessive activation of GSK-3β is closely related to tau hyperphosphorylation, and inhibition of the GSK-3β over-activation may alleviate tau hyperphosphorylation, thereby slowing the further aggravation of AD

(Figure 3). GSK-3β is not only the important kinases of tau phosphorylation, but also an important kinase of tau hyperphosphorylation, in different status. GSK-3β, as a kinase in insulin signal transduction and tau protein phosphorylation, plays an important role both in the formation and aggravation of insulin resistance, and in the clearance and abnormal deposition of Aβ as well [105]. Therefore, we speculate that GSK-3β is the link between AD and DM. Progress in targeting the GSK-3β in AD

IP T

5.

We have reviewed the important role of GSK-3β in AD and DM, which as a

SC R

potential link between DM and AD, that is a good explanation of why the risk of AD in

diabetic patients more than twice as high as normal [106]. Therefore, a large number of inhibitors for GSK-3β have been studied, and some of the GSK-3β inhibitors have been treated as a therapeutic drug for AD into a Phase II clinical trial [107, 108]. GSK-3β

U

inhibitors can be divided into ATP competitive and non-ATP competitive, and the first

N

classes of GSK-3β inhibitors were classified as ATP competitive, but non-ATP

A

competitive GSK-3β inhibitors with high selectivity [109]. Due to the high selectivity and less toxicity of non-ATP competition, it is considered to be a more promising

M

research direction. This section focuses on the progress of GSK-3β inhibitors in AD, and we will review the progress of GSK-3β inhibitors in AD from basic and clinical

TE D

studies (Table 1). First, let's review basic research of GSK-3β inhibitors in AD: in cultured rat cortical cells, the caspase-dependent apoptosis induced by calcineurin inhibitors was prevented by GSK-3β inhibitor 1-Azakenpaullone [110]. And in c-Kit

EP

Wv/+mice (c-Kit Wv/+ mutation-induced β-cell dysfunction), inhibition of GSK-3β activity by 1-Azakenpaullone improves β-cell function, 1-Azakenpaullone could prevent the onset of diabetes by improving glucose tolerance and β-cell function [111].

CC

In tauopathies (a group of disorders characterized by aggregation of tau include AD and the fronto-temporal dementias), tau abnormalities significantly disrupt neuronal

A

function, before the classical pathological hallmarks of tauopathies. Use lithium and AR-A014418 (another GSK-3β inhibitor) significantly reduced the tau overexpression on axonal, and affect the abnormal behavior of Drosophila [112]. Treatment of primary cultures neurons with lithium and AR-A014418 decreased full-length fractalkine (a large cytokine protein), proving that changing the inflammatory environment may also be the reason for GSK-3β inhibitors's neuroprotective effect [113]. Furthermore the treatment of human neural progenitor cells with IM-12 (GSK-3β inhibitor) resulted in

an increase of neuronal cells, suggest that GSK-3β is closely related to neuronal proliferation [114]. Indirubin and its derivatives have been shown to possess potent inhibitory effects on GSK-3β, use it to treatment APP/PS1 mice, that the spatial memory deficits significantly attenuated, and this was accompanied by a marked decrease in several AD-like phenotypes [115]. In the previous article, we referred to the high fat diet and insulin resistance are closely linked. Treatment of mice (with high fat diet induced cognitive impairment) with Indirubin the learning and memory

IP T

performance of mice increased, suggested that GSK-3β inhibition could prove to be

beneficial in insulin resistance induced cognitive deficit [116]. In addition, Indirubin

SC R

suppresses Aβ-induced apoptosis by inhibiting tau hyperphosphorylation [117] Intrahippocampal TDZD-8 (a non-ATP competitive thiadiazolidine inhibitor of GSK3β) blocked tau hyperphosphorylation and normalized hippocampus-dependent

memory in db/db mice, suggest that the GSK-3β inhibitor plays a protective role in

U

T2D-induced memory impairment [118]. Then let's review clinical research GSK-3β

N

inhibitors in AD: now the clinical research of GSK-3β inhibitors is still relatively less.

A

But as early as 2013, a pilot study of GSK-3β inhibitor tideglusib in AD had been done. Tideglusib produced positive trends in MMSE (Mini-Mental Status Examination),

M

ADAS-cog (Alzheimer’s Disease Assessment Scale-cognitive subscale), GDS (Geriatric Depression Scale), and GCA (Global Clinical Assessment) without statistical

TE D

significance in this small sample, this trial provides preliminary evidence to support a benefit of Tideglusib in AD [119]. Then in a randomized trial, Tideglusib reduces progression of brain atrophy in progressive supranuclear palsy, that reduced the

EP

progression of atrophy in the whole brain, particularly in the parietal and occipital lobes [120]. But in a phase II trial of Tideglusib in AD, many indicators are still no statistical difference, even though BACE1 in CSF significantly decreased with treatment in a

CC

small subgroup of patients [107]. According to the review earlier in this article, although Tideglusib encountered some obstacles in Phase II clinical trials, we still

A

believe that this is a promising target. We think it should be perform PS1 mutation screening in the diabetic patients, because they have a high risk of AD, treatment of this diabetic patients with GSK-3β inhibitors will have a positive effect.

6.

Conclusion Since AD has been discovered, we have never slowed the pace of its exploration,

but AD is still a huge mountain that we are unable to overcome. With the annual incidence of AD increasing dramatically, we urgently need to find drugs that can prevent and/or improve it. Recently it was found that the risk of dementia in DM patients is higher than that of non-diabetic patients and it is also well known that T2D/insulin resistance is involved in AD [121], further evidenced by researchers from

IP T

Rhode Island Hospital and Brown Medical School who discovered that the brain was

also responsible for producing small amounts of insulin and brought up the concept of

SC R

T3D [9], which is a condition where the brain doesn't produce enough insulin or known as the brain diabetes [122, 123]. This breakthrough also attracted more attention to further investigate the link between T2D and AD. There are many complex links between DM and AD, such as neuroinflammation, mitochondrial dysfunction and

U

imbalance of intestinal flora, but the key relation is still unclear [98, 124-126]. In recent

N

years, tau as a candidate for the treatment of AD has gotten more attention. Tau

A

hyperphosphorylation and the resulting formation of neurofibrillary tangles as well as synaptic loss are closely related to memory deficit in patients with AD. Interestingly, a

M

large number of recent studies have shown that GSK-3β as a common kinase of insulin signal transduction and tau protein phosphorylation has played an indispensable role in

TE D

DM and AD, which may be the potential link between DM and AD. In DM, GSK-3β plays a key role in the regulation of blood glucose, which is the main rate limiting enzyme for inhibition of glycogen synthesis. More importantly, it is

EP

one of the key factors leading to insulin deficiency and insulin resistance, and insulin resistance is a hallmark in the occurrence and development of DM. In AD, GSK-3β plays an important role as a kinase in both phosphorylation and hyperphosphorylation

CC

of tau. Meanwhile insulin resistance in DM may cause Aβ deposition, which will be cleared by tau, but excessive phosphorylation of tau will further aggravate Aβ

A

neurotoxicity, damage the brain and affect cognitive function. GSK-3β may not only be a promising therapeutic target, but also an important clue through which we can climb over the mountain of AD. Disclosure statement The authors have no conflicts of interest to disclose. Acknowledgements I gratefully acknowledge my valued colleagues for their contributions to several

studies described in this review. This work was supported by the National Natural Science Foundation of China (81660599, 81360311, 81460548), postgraduate education innovation program of Guizhou Province (11374), the Zunyi Medical University Funds (2013F-686, F-738).

A

CC

EP

TE D

M

A

N

U

SC R

IP T

References [1] Araujo DM, Cotman CW. Beta-amyloid stimulates glial cells in vitro to produce growth factors that accumulate in senile plaques in Alzheimer's disease. Brain research. 1992;569:141-5. [2] Kidd M. Paired helical filaments in electron microscopy of Alzheimer's disease. Nature. 1963;197:192-3. [3] International AsD. World Alzheimer report 2015. http://www.worldalzreport2015.org/downloads/world-alzheimer-report-2015.pdf. 2015. [4] Jin F, Gong QH, Xu YS, Wang LN, Jin H, Li F, et al. Icariin, a phosphodiesterase-5 inhibitor, improves learning and memory in APP/PS1 transgenic mice by stimulation of NO/cGMP signalling. The international journal of neuropsychopharmacology. 2014;17:871-81. [5] Vandal M, White PJ, Chevrier G, Tremblay C, St-Amour I, Planel E, et al. Agedependent impairment of glucose tolerance in the 3xTg-AD mouse model of Alzheimer's disease. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2015;29:4273-84. [6] Turner RS, Thomas RG, Craft S, van Dyck CH, Mintzer J, Reynolds BA, et al. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology. 2015;85:1383-91. [7] Ewald CY, Raps DA, Li C. APL-1, the Alzheimer's Amyloid precursor protein in Caenorhabditis elegans, modulates multiple metabolic pathways throughout development. Genetics. 2012;191:493-507. [8] Tamaki C, Ohtsuki S, Terasaki T. Insulin facilitates the hepatic clearance of plasma amyloid beta-peptide (1 40) by intracellular translocation of low-density lipoprotein receptor-related protein 1 (LRP-1) to the plasma membrane in hepatocytes. Molecular pharmacology. 2007;72:850-5. [9] Steen E, Terry BM, Rivera EJ, Cannon JL, Neely TR, Tavares R, et al. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer's disease--is this type 3 diabetes? Journal of Alzheimer's disease : JAD. 2005;7:63-80. [10] de la Monte SM, Tong M, Lester-Coll N, Plater M, Jr., Wands JR. Therapeutic rescue of neurodegeneration in experimental type 3 diabetes: relevance to Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2006;10:89-109. [11] Hemmings BA, Cohen P. Glycogen synthase kinase-3 from rabbit skeletal muscle. Methods in enzymology. 1983;99:337-45. [12] Robergs RA, McMinn SB, Mermier C, Leadbetter G, 3rd, Ruby B, Quinn C. Blood glucose and glucoregulatory hormone responses to solid and liquid carbohydrate ingestion during exercise. Int J Sport Nutr. 1998;8:70-83. [13] Duan C, Li M, Rui L. SH2-B promotes insulin receptor substrate 1 (IRS1)- and IRS2-mediated activation of the phosphatidylinositol 3-kinase pathway in response to leptin. J Biol Chem. 2004;279:43684-91. [14] Bijur GN, Jope RS. Opposing actions of phosphatidylinositol 3-kinase and glycogen synthase kinase-3beta in the regulation of HSF-1 activity. J Neurochem.

A

CC

EP

TE D

M

A

N

U

SC R

IP T

2000;75:2401-8. [15] Bhat RV, Shanley J, Correll MP, Fieles WE, Keith RA, Scott CW, et al. Regulation and localization of tyrosine216 phosphorylation of glycogen synthase kinase-3beta in cellular and animal models of neuronal degeneration. Proceedings of the National Academy of Sciences of the United States of America. 2000;97:11074-9. [16] Gleason JE, Szyleyko EA, Eisenmann DM. Multiple redundant Wnt signaling components function in two processes during C. elegans vulval development. Dev Biol. 2006;298:442-57. [17] Moon B, Duddy N, Ragolia L, Begum N. Stimulation of glycogen synthesis by heat shock in L6 skeletal-muscle cells: regulatory role of site-specific phosphorylation of glycogen-associated protein phosphatase 1. The Biochemical journal. 2003;371:85766. [18] Liu Z, Tanabe K, Bernal-Mizrachi E, Permutt MA. Mice with beta cell overexpression of glycogen synthase kinase-3beta have reduced beta cell mass and proliferation. Diabetologia. 2008;51:623-31. [19] Nolan CJ. Lipotoxicity, beta cell dysfunction, and gestational diabetes. Cell metabolism. 2014;19:553-4. [20] Donath MY, Storling J, Maedler K, Mandrup-Poulsen T. Inflammatory mediators and islet beta-cell failure: a link between type 1 and type 2 diabetes. Journal of molecular medicine (Berlin, Germany). 2003;81:455-70. [21] Robertson R, Zhou H, Zhang T, Harmon JS. Chronic oxidative stress as a mechanism for glucose toxicity of the beta cell in type 2 diabetes. Cell biochemistry and biophysics. 2007;48:139-46. [22] Robertson RP. Chronic oxidative stress as a central mechanism for glucose toxicity in pancreatic islet beta cells in diabetes. The Journal of biological chemistry. 2004;279:42351-4. [23] Del Guerra S, Grupillo M, Masini M, Lupi R, Bugliani M, Torri S, et al. Gliclazide protects human islet beta-cells from apoptosis induced by intermittent high glucose. Diabetes/metabolism research and reviews. 2007;23:234-8. [24] Kaneto H, Matsuoka TA, Nakatani Y, Kawamori D, Matsuhisa M, Yamasaki Y. Oxidative stress and the JNK pathway in diabetes. Current diabetes reviews. 2005;1:6572. [25] Kaneto H, Matsuoka TA, Nakatani Y, Kawamori D, Miyatsuka T, Matsuhisa M, et al. Oxidative stress, ER stress, and the JNK pathway in type 2 diabetes. Journal of molecular medicine (Berlin, Germany). 2005;83:429-39. [26] Grimes CA, Jope RS. CREB DNA binding activity is inhibited by glycogen synthase kinase-3 beta and facilitated by lithium. J Neurochem. 2001;78:1219-32. [27] Pugazhenthi S, Nesterova A, Sable C, Heidenreich KA, Boxer LM, Heasley LE, et al. Akt/protein kinase B up-regulates Bcl-2 expression through cAMP-response element-binding protein. J Biol Chem. 2000;275:10761-6. [28] Tanioka T, Tamura Y, Fukaya M, Shinozaki S, Mao J, Kim M, et al. Inducible nitric-oxide synthase and nitric oxide donor decrease insulin receptor substrate-2 protein expression by promoting proteasome-dependent degradation in pancreatic betacells: involvement of glycogen synthase kinase-3beta. J Biol Chem. 2011;286:2938896. [29] Gagliardino JJ, Del Zotto H, Massa L, Flores LE, Borelli MI. Pancreatic duodenal homeobox-1 and islet neogenesis-associated protein: a possible combined marker of activateable pancreatic cell precursors. The Journal of endocrinology. 2003;177:24959. [30] Brownlee M. A radical explanation for glucose-induced beta cell dysfunction. The

A

CC

EP

TE D

M

A

N

U

SC R

IP T

Journal of clinical investigation. 2003;112:1788-90. [31] Liu Y, Tanabe K, Baronnier D, Patel S, Woodgett J, Cras-Meneur C, et al. Conditional ablation of Gsk-3beta in islet beta cells results in expanded mass and resistance to fat feeding-induced diabetes in mice. Diabetologia. 2010;53:2600-10. [32] Sims-Robinson C, Kim B, Rosko A, Feldman EL. How does diabetes accelerate Alzheimer disease pathology? Nature reviews Neurology. 2010;6:551-9. [33] Liolitsa D, Powell J, Lovestone S. Genetic variability in the insulin signalling pathway may contribute to the risk of late onset Alzheimer's disease. Journal of neurology, neurosurgery, and psychiatry. 2002;73:261-6. [34] Yokoo H, Nemoto T, Yanagita T, Satoh S, Yoshikawa N, Maruta T, et al. Glycogen synthase kinase-3beta: homologous regulation of cell surface insulin receptor level via controlling insulin receptor mRNA stability in adrenal chromaffin cells. J Neurochem. 2007;103:1883-96. [35] Moh A, Zhang W, Yu S, Wang J, Xu X, Li J, et al. STAT3 sensitizes insulin signaling by negatively regulating glycogen synthase kinase-3 beta. Diabetes. 2008;57:1227-35. [36] Das GC, Hollinger FB. Molecular pathways for glucose homeostasis, insulin signaling and autophagy in hepatitis C virus induced insulin resistance in a cellular model. Virology. 2012;434:5-17. [37] Hsieh MJ, Lan KP, Liu HY, Zhang XZ, Lin YF, Chen TY, et al. Hepatitis C virus E2 protein involve in insulin resistance through an impairment of Akt/PKB and GSK3beta signaling in hepatocytes. BMC Gastroenterol. 2012;12:74. [38] Wang Y, Feng W, Xue W, Tan Y, Hein DW, Li XK, et al. Inactivation of GSK3beta by metallothionein prevents diabetes-related changes in cardiac energy metabolism, inflammation, nitrosative damage, and remodeling. Diabetes. 2009;58:1391-402. [39] Lappas M. GSK3beta is increased in adipose tissue and skeletal muscle from women with gestational diabetes where it regulates the inflammatory response. PLoS One. 2014;9:e115854. [40] Heilbronn LK, Campbell LV. Adipose tissue macrophages, low grade inflammation and insulin resistance in human obesity. Curr Pharm Des. 2008;14:122530. [41] Benjamin WB, Pentyala SN, Woodgett JR, Hod Y, Marshak D. ATP citrate-lyase and glycogen synthase kinase-3 beta in 3T3-L1 cells during differentiation into adipocytes. The Biochemical journal. 1994;300 ( Pt 2):477-82. [42] Chen G, Mou LP, Yao J, You TT, Shen XY, Zhu XQ, et al. [Construction of GSK3beta-targeting RNAi adenovirus vector and the influence of Wnt/beta-catenin pathway in proliferation of human thyrocytes]. Zhonghua yi xue za zhi. 2008;88:2821-5. [43] Cao J, Feng XX, Yao L, Ning B, Yang ZX, Fang DL, et al. Saturated free fatty acid sodium palmitate-induced lipoapoptosis by targeting glycogen synthase kinase-3beta activation in human liver cells. Digestive diseases and sciences. 2014;59:346-57. [44] Boden G. Free fatty acids (FFA), a link between obesity and insulin resistance. Frontiers in bioscience : a journal and virtual library. 1998;3:d169-75. [45] Storlien LH, Jenkins AB, Chisholm DJ, Pascoe WS, Khouri S, Kraegen EW. Influence of dietary fat composition on development of insulin resistance in rats. Relationship to muscle triglyceride and omega-3 fatty acids in muscle phospholipid. Diabetes. 1991;40:280-9. [46] Reaven GM. Banting lecture 1988. Role of insulin resistance in human disease. Diabetes. 1988;37:1595-607. [47] Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin

A

CC

EP

TE D

M

A

N

U

SC R

IP T

resistance and type 2 diabetes. Nature. 2006;444:840-6. [48] Niu L, Han DW, Xu RL, Han B, Zhou X, Wu HW, et al. A High-sugar High-fat Diet Induced Metabolic Syndrome Shows some Symptoms of Alzheimer's Disease in Rats. The journal of nutrition, health & aging. 2016;20:509-13. [49] Shieh JM, Wu HT, Cheng KC, Cheng JT. Melatonin ameliorates high fat dietinduced diabetes and stimulates glycogen synthesis via a PKCzeta-Akt-GSK3beta pathway in hepatic cells. Journal of pineal research. 2009;47:339-44. [50] Wang ZB, Zeng HC, Wei HS, Yi GH, Yu J, Wang YT, et al. NO-1886 ameliorates glycogen metabolism in insulin-resistant HepG2 cells by GSK-3beta signalling. The Journal of pharmacy and pharmacology. 2012;64:293-301. [51] Hanger DP, Hughes K, Woodgett JR, Brion JP, Anderton BH. 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. 1992;147:58-62. [52] Paudel HK, Lew J, Ali Z, Wang JH. Brain proline-directed protein kinase phosphorylates tau on sites that are abnormally phosphorylated in tau associated with Alzheimer's paired helical filaments. The Journal of biological chemistry. 1993;268:23512-8. [53] Hernandez F, Lucas JJ, Avila J. GSK3 and tau: two convergence points in Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2013;33 Suppl 1:S141-4. [54] Pei JJ, Braak E, Braak H, Grundke-Iqbal I, Iqbal K, Winblad B, et al. Distribution of active glycogen synthase kinase 3beta (GSK-3beta) in brains staged for Alzheimer disease neurofibrillary changes. Journal of neuropathology and experimental neurology. 1999;58:1010-9. [55] Terwel D, Muyllaert D, Dewachter I, Borghgraef P, Croes S, Devijver H, et al. Amyloid activates GSK-3beta to aggravate neuronal tauopathy in bigenic mice. The American journal of pathology. 2008;172:786-98. [56] Spittaels K, Van den Haute C, Van Dorpe J, Terwel D, Vandezande K, Lasrado R, et al. Neonatal neuronal overexpression of glycogen synthase kinase-3 beta reduces brain size in transgenic mice. Neuroscience. 2002;113:797-808. [57] Llorens-Martin M, Lopez-Domenech G, Soriano E, Avila J. GSK3beta is involved in the relief of mitochondria pausing in a Tau-dependent manner. PloS one. 2011;6:e27686. [58] Povellato G, Tuxworth RI, Hanger DP, Tear G. Modification of the Drosophila model of in vivo Tau toxicity reveals protective phosphorylation by GSK3beta. Biology open. 2014;3:1-11. [59] Yeh PA, Chang CJ, Tu PH, Wilson HI, Chien JY, Tang CY, et al. Phosphorylation alters tau distribution and elongates life span in Drosophila. Journal of Alzheimer's disease : JAD. 2010;21:543-56. [60] Ittner A, Chua SW, Bertz J, Volkerling A, van der Hoven J, Gladbach A, et al. Sitespecific phosphorylation of tau inhibits amyloid-beta toxicity in Alzheimer's mice. Science (New York, NY). 2016;354:904-8. [61] Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW. A protein factor essential for microtubule assembly. Proc Natl Acad Sci U S A. 1975;72:1858-62. [62] Cleveland DW, Hwo SY, Kirschner MW. Purification of tau, a microtubuleassociated protein that induces assembly of microtubules from purified tubulin. J Mol Biol. 1977;116:207-25. [63] Cleveland DW, Hwo SY, Kirschner MW. Physical and chemical properties of purified tau factor and the role of tau in microtubule assembly. Journal of molecular biology. 1977;116:227-47.

A

CC

EP

TE D

M

A

N

U

SC R

IP T

[64] Dixit R, Ross JL, Goldman YE, Holzbaur EL. Differential regulation of dynein and kinesin motor proteins by tau. Science (New York, NY). 2008;319:1086-9. [65] Billingsley ML, Kincaid RL. Regulated phosphorylation and dephosphorylation of tau protein: effects on microtubule interaction, intracellular trafficking and neurodegeneration. The Biochemical journal. 1997;323 ( Pt 3):577-91. [66] Lesort M, Blanchard C, Yardin C, Esclaire F, Hugon J. Cultured neurons expressing phosphorylated tau are more resistant to apoptosis induced by NMDA or serum deprivation. Brain research Molecular brain research. 1997;45:127-32. [67] Cho JH, Johnson GV. Primed phosphorylation of tau at Thr231 by glycogen synthase kinase 3beta (GSK3beta) plays a critical role in regulating tau's ability to bind and stabilize microtubules. J Neurochem. 2004;88:349-58. [68] Woodgett JR. Molecular cloning and expression of glycogen synthase kinase3/factor A. The EMBO journal. 1990;9:2431-8. [69] Leroy K, Brion JP. Developmental expression and localization of glycogen synthase kinase-3beta in rat brain. J Chem Neuroanat. 1999;16:279-93. [70] Takahashi M, Tomizawa K, Kato R, Sato K, Uchida T, Fujita SC, et al. Localization and developmental changes of tau protein kinase I/glycogen synthase kinase-3 beta in rat brain. J Neurochem. 1994;63:245-55. [71] Sun W, Qureshi HY, Cafferty PW, Sobue K, Agarwal-Mawal A, Neufield KD, et al. Glycogen synthase kinase-3beta is complexed with tau protein in brain microtubules. J Biol Chem. 2002;277:11933-40. [72] Rogaeva EA, Fafel KC, Song YQ, Medeiros H, Sato C, Liang Y, et al. Screening for PS1 mutations in a referral-based series of AD cases: 21 novel mutations. Neurology. 2001;57:621-5. [73] Duff K, Eckman C, Zehr C, Yu X, Prada CM, Perez-tur J, et al. Increased amyloidbeta42(43) in brains of mice expressing mutant presenilin 1. Nature. 1996;383:710-3. [74] Murayama M, Tanaka S, Palacino J, Murayama O, Honda T, Sun X, et al. Direct association of presenilin-1 with beta-catenin. FEBS letters. 1998;433:73-7. [75] Kang DE, Soriano S, Frosch MP, Collins T, Naruse S, Sisodia SS, et al. Presenilin 1 facilitates the constitutive turnover of beta-catenin: differential activity of Alzheimer's disease-linked PS1 mutants in the beta-catenin-signaling pathway. The Journal of neuroscience : the official journal of the Society for Neuroscience. 1999;19:4229-37. [76] Takashima A, Murayama M, Murayama O, Kohno T, Honda T, Yasutake K, et al. Presenilin 1 associates with glycogen synthase kinase-3beta and its substrate tau. Proceedings of the National Academy of Sciences of the United States of America. 1998;95:9637-41. [77] Tanemura K, Chui DH, Fukuda T, Murayama M, Park JM, Akagi T, et al. Formation of tau inclusions in knock-in mice with familial Alzheimer disease (FAD) mutation of presenilin 1 (PS1). The Journal of biological chemistry. 2006;281:5037-41. [78] Prager K, Wang-Eckhardt L, Fluhrer R, Killick R, Barth E, Hampel H, et al. A structural switch of presenilin 1 by glycogen synthase kinase 3beta-mediated phosphorylation regulates the interaction with beta-catenin and its nuclear signaling. The Journal of biological chemistry. 2007;282:14083-93. [79] Twomey C, McCarthy JV. Presenilin-1 is an unprimed glycogen synthase kinase3beta substrate. FEBS letters. 2006;580:4015-20. [80] Lucas JJ, Hernandez F, Gomez-Ramos P, Moran MA, Hen R, Avila J. Decreased nuclear beta-catenin, tau hyperphosphorylation and neurodegeneration in GSK-3beta conditional transgenic mice. The EMBO journal. 2001;20:27-39. [81] Zhang Y, Zhang Z, Wang H, Cai N, Zhou S, Zhao Y, et al. Neuroprotective effect of ginsenoside Rg1 prevents cognitive impairment induced by isoflurane anesthesia in

A

CC

EP

TE D

M

A

N

U

SC R

IP T

aged rats via antioxidant, anti-inflammatory and anti-apoptotic effects mediated by the PI3K/AKT/GSK-3beta pathway. Mol Med Rep. 2016;14:2778-84. [82] Biessels GJ, Reagan LP. Hippocampal insulin resistance and cognitive dysfunction. Nature reviews Neuroscience. 2015;16:660-71. [83] Wang Y, Liu W, He X, Zhou F. Parkinson's disease-associated DJ-1 mutations increase abnormal phosphorylation of tau protein through Akt/GSK-3beta pathways. Journal of molecular neuroscience : MN. 2013;51:911-8. [84] Wang Y, Wu C, Han B, Xu F, Mao M, Guo X, et al. Dexmedetomidine attenuates repeated propofol exposure-induced hippocampal apoptosis, PI3K/Akt/Gsk-3beta signaling disruption, and juvenile cognitive deficits in neonatal rats. Mol Med Rep. 2016;14:769-75. [85] Qi Y, Dou DQ, Jiang H, Zhang BB, Qin WY, Kang K, et al. Arctigenin Attenuates Learning and Memory Deficits through PI3k/Akt/GSK-3beta Pathway Reducing Tau Hyperphosphorylation in Abeta-Induced AD Mice. Planta medica. 2017;83:51-6. [86] Yao ES, Tang Y, Liu XH, Wang MH. TPPU protects tau from H2O2-induced hyperphosphorylation in HEK293/tau cells by regulating PI3K/AKT/GSK-3beta pathway. Journal of Huazhong University of Science and Technology Medical sciences = Hua zhong ke ji da xue xue bao Yi xue Ying De wen ban = Huazhong keji daxue xuebao Yixue Yingdewen ban. 2016;36:785-90. [87] Chen Y, Wang C, Hu M, Pan J, Chen J, Duan P, et al. Effects of ginkgolide A on okadaic acid-induced tau hyperphosphorylation and the PI3K-Akt signaling pathway in N2a cells. Planta Med. 2012;78:1337-41. [88] Haan MN. Therapy Insight: type 2 diabetes mellitus and the risk of late-onset Alzheimer's disease. Nature clinical practice Neurology. 2006;2:159-66. [89] Verdile G, Keane KN, Cruzat VF, Medic S, Sabale M, Rowles J, et al. Inflammation and Oxidative Stress: The Molecular Connectivity between Insulin Resistance, Obesity, and Alzheimer's Disease. Mediators of inflammation. 2015;2015:105828. [90] Willette AA, Bendlin BB, Starks EJ, Birdsill AC, Johnson SC, Christian BT, et al. Association of Insulin Resistance With Cerebral Glucose Uptake in Late Middle-Aged Adults at Risk for Alzheimer Disease. JAMA neurology. 2015;72:1013-20. [91] Kumar DK, Choi SH, Washicosky KJ, Eimer WA, Tucker S, Ghofrani J, et al. Amyloid-beta peptide protects against microbial infection in mouse and worm models of Alzheimer's disease. Sci Transl Med. 2016;8:340ra72. [92] Lin M, Yiu WH, Li RX, Wu HJ, Wong DW, Chan LY, et al. The TLR4 antagonist CRX-526 protects against advanced diabetic nephropathy. Kidney Int. 2013;83:887900. [93] Wang YL, Wang K, Yu SJ, Li Q, Li N, Lin PY, et al. Association of the TLR4 signaling pathway in the retina of streptozotocin-induced diabetic rats. Graefe's archive for clinical and experimental ophthalmology = Albrecht von Graefes Archiv fur klinische und experimentelle Ophthalmologie. 2015;253:389-98. [94] Kiasalari Z, Rahmani T, Mahmoudi N, Baluchnejadmojarad T, Roghani M. Diosgenin ameliorates development of neuropathic pain in diabetic rats: Involvement of oxidative stress and inflammation. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie. 2017;86:654-61. [95] Zhao M, Zhou A, Xu L, Zhang X. The role of TLR4-mediated PTEN/PI3K/AKT/NF-kappaB signaling pathway in neuroinflammation in hippocampal neurons. Neuroscience. 2014;269:93-101. [96] Herber DL, Roth LM, Wilson D, Wilson N, Mason JE, Morgan D, et al. Timedependent reduction in Abeta levels after intracranial LPS administration in APP

A

CC

EP

TE D

M

A

N

U

SC R

IP T

transgenic mice. Experimental neurology. 2004;190:245-53. [97] Herber DL, Mercer M, Roth LM, Symmonds K, Maloney J, Wilson N, et al. Microglial activation is required for Abeta clearance after intracranial injection of lipopolysaccharide in APP transgenic mice. Journal of neuroimmune pharmacology : the official journal of the Society on NeuroImmune Pharmacology. 2007;2:222-31. [98] Huang NQ, Jin H, Zhou SY, Shi JS, Jin F. TLR4 is a link between diabetes and Alzheimer's disease. Behav Brain Res. 2017;316:234-44. [99] Lee HG, Perry G, Moreira PI, Garrett MR, Liu Q, Zhu X, et al. Tau phosphorylation in Alzheimer's disease: pathogen or protector? Trends Mol Med. 2005;11:164-9. [100] Smith MA, Casadesus G, Joseph JA, Perry G. Amyloid-beta and tau serve antioxidant functions in the aging and Alzheimer brain. Free Radic Biol Med. 2002;33:1194-9. [101] Papon MA, El Khoury NB, Marcouiller F, Julien C, Morin F, Bretteville A, et al. Deregulation of protein phosphatase 2A and hyperphosphorylation of tau protein following onset of diabetes in NOD mice. Diabetes. 2013;62:609-17. [102] Jolivalt CG, Lee CA, Beiswenger KK, Smith JL, Orlov M, Torrance MA, et al. Defective insulin signaling pathway and increased glycogen synthase kinase-3 activity in the brain of diabetic mice: parallels with Alzheimer's disease and correction by insulin. J Neurosci Res. 2008;86:3265-74. [103] Kim DY, Jung SY, Kim TW, Lee KS, Kim K. Treadmill exercise decreases incidence of Alzheimer's disease by suppressing glycogen synthase kinase-3beta expression in streptozotocin-induced diabetic rats. Journal of exercise rehabilitation. 2015;11:87-94. [104] Wang X, Zhao L. Calycosin ameliorates diabetes-induced cognitive impairments in rats by reducing oxidative stress via the PI3K/Akt/GSK-3beta signaling pathway. Biochemical and biophysical research communications. 2016;473:428-34. [105] Liu Y, Liu F, Grundke-Iqbal I, Iqbal K, Gong CX. Deficient brain insulin signalling pathway in Alzheimer's disease and diabetes. The Journal of pathology. 2011;225:54-62. [106] Fei M, Yan Ping Z, Ru Juan M, Ning Ning L, Lin G. Risk factors for dementia with type 2 diabetes mellitus among elderly people in China. Age and ageing. 2013;42:398-400. [107] Tolosa E, Litvan I, Hoglinger GU, Burn D, Lees A, Andres MV, et al. A phase 2 trial of the GSK-3 inhibitor tideglusib in progressive supranuclear palsy. Movement disorders : official journal of the Movement Disorder Society. 2014;29:470-8. [108] Lovestone S, Boada M, Dubois B, Hull M, Rinne JO, Huppertz HJ, et al. A phase II trial of tideglusib in Alzheimer's disease. Journal of Alzheimer's disease : JAD. 2015;45:75-88. [109] De Simone A, Fiori J, Naldi M, D'Urzo A, Tumiatti V, Milelli A, et al. Application of an ESI-QTOF method for the detailed characterization of GSK-3beta inhibitors. Journal of pharmaceutical and biomedical analysis. 2017;144:159-66. [110] Takadera T, Ohyashiki T. Caspase-dependent apoptosis induced by calcineurin inhibitors was prevented by glycogen synthase kinase-3 inhibitors in cultured rat cortical cells. Brain Res. 2007;1133:20-6. [111] Feng ZC, Donnelly L, Li J, Krishnamurthy M, Riopel M, Wang R. Inhibition of Gsk3beta activity improves beta-cell function in c-KitWv/+ male mice. Laboratory investigation; a journal of technical methods and pathology. 2012;92:543-55. [112] Mudher A, Shepherd D, Newman TA, Mildren P, Jukes JP, Squire A, et al. GSK3beta inhibition reverses axonal transport defects and behavioural phenotypes in

A

CC

EP

TE D

M

A

N

U

SC R

IP T

Drosophila. Molecular psychiatry. 2004;9:522-30. [113] Fuster-Matanzo A, Jurado-Arjona J, Benvegnu S, Garcia E, Martin-Maestro P, Gomez-Sintes R, et al. Glycogen synthase kinase-3beta regulates fractalkine production by altering its trafficking from Golgi to plasma membrane: implications for Alzheimer's disease. Cellular and molecular life sciences : CMLS. 2017;74:1153-63. [114] Schmole AC, Brennfuhrer A, Karapetyan G, Jaster R, Pews-Davtyan A, Hubner R, et al. Novel indolylmaleimide acts as GSK-3beta inhibitor in human neural progenitor cells. Bioorganic & medicinal chemistry. 2010;18:6785-95. [115] Ding Y, Qiao A, Fan GH. Indirubin-3'-monoxime rescues spatial memory deficits and attenuates beta-amyloid-associated neuropathology in a mouse model of Alzheimer's disease. Neurobiology of disease. 2010;39:156-68. [116] Sharma S, Taliyan R. Neuroprotective role of Indirubin-3'-monoxime, a GSKbeta inhibitor in high fat diet induced cognitive impairment in mice. Biochemical and biophysical research communications. 2014;452:1009-15. [117] Zhang SG, Wang XS, Zhang YD, Di Q, Shi JP, Qian M, et al. Indirubin-3'monoxime suppresses amyloid-beta-induced apoptosis by inhibiting tau hyperphosphorylation. Neural regeneration research. 2016;11:988-93. [118] Dey A, Hao S, Wosiski-Kuhn M, Stranahan AM. Glucocorticoid-mediated activation of GSK3beta promotes tau phosphorylation and impairs memory in type 2 diabetes. Neurobiology of aging. 2017;57:75-83. [119] del Ser T, Steinwachs KC, Gertz HJ, Andres MV, Gomez-Carrillo B, Medina M, et al. Treatment of Alzheimer's disease with the GSK-3 inhibitor tideglusib: a pilot study. Journal of Alzheimer's disease : JAD. 2013;33:205-15. [120] Hoglinger GU, Huppertz HJ, Wagenpfeil S, Andres MV, Belloch V, Leon T, et al. Tideglusib reduces progression of brain atrophy in progressive supranuclear palsy in a randomized trial. Movement disorders : official journal of the Movement Disorder Society. 2014;29:479-87. [121] Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027-31. [122] Schwartz MW, Porte D, Jr. Diabetes, obesity, and the brain. Science (New York, NY). 2005;307:375-9. [123] van Bussel FC, Backes WH, van Veenendaal TM, Hofman PA, van Boxtel MP, Schram MT, et al. Functional Brain Networks Are Altered in Type 2 Diabetes and Prediabetes: Signs for Compensation of Cognitive Decrements? The Maastricht Study. Diabetes. 2016;65:2404-13. [124] Ransohoff RM. How neuroinflammation contributes to neurodegeneration. Science (New York, NY). 2016;353:777. [125] Moreira PI, Santos MS, Seica R, Oliveira CR. Brain mitochondrial dysfunction as a link between Alzheimer's disease and diabetes. Journal of the neurological sciences. 2007;257:206-14. [126] Vignini A, Giulietti A, Nanetti L, Raffaelli F, Giusti L, Mazzanti L, et al. Alzheimer's disease and diabetes: new insights and unifying therapies. Current diabetes reviews. 2013;9:218-27.

SC R

IP T

Figures

U

Fig.1. A simplified schematic diagram representing that GSK-3β involved in blood

CC

EP

TE D

M

A

N

glucose regulation.

A

Fig.2. A simplified schematic diagram representing the relationship between GSK-3β and insulin resistance.

IP T SC R

Fig.3. A simplified schematic diagram representing the role of GSK-3β in insulin

A

CC

EP

TE D

M

A

N

U

resistance-related AD.

Table Table 1 GSK-3β Inhibitors in AD. GSK-3β Inhibitors Inhibitor

Reference(s) [110, 111]

AR-A014418

[112, 113]

IM-12

[114]

Indirubin

[115-117]

lithium

[112, 113]

TDZD-8

[118]

Tideglusib

[107, 108, 119, 120]

A

CC

EP

TE D

M

A

N

U

SC R

IP T

1-Azakenpaullone