Review Article

Mitochondrial oxidative stress regulatory approaches for diabetic encephalopathy

https://orcid.org/0000-0003-0960-1606, https://orcid.org/0000-0002-8637-8164, https://orcid.org/0000-0002-0425-0168, https://orcid.org/0000-0002-2762-2649
Hye Jih Kim1,2,https://orcid.org/0000-0003-0960-1606, Dae Hyun Kim1,2,https://orcid.org/0000-0002-8637-8164, Hang Hyo Jo1,2,https://orcid.org/0000-0002-0425-0168, Hyun Jik Lee1,2,*https://orcid.org/0000-0002-2762-2649
Author Information & Copyright
1College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Korea
2Institute for Stem Cell & Regenerative Medicine (ISCRM), Chungbuk National University, Cheongju 28644, Korea

†These authors contributed equally to this work.

*Corresponding author: Hyun Jik Lee, College of Veterinary Medicine and Veterinary Medicine Center, Chungbuk National University, Cheongju 28644, Korea, Tel: +82-43-261-2597, E-mail: leehyunjik@chungbuk.ac.kr

© Research Institute of Veterinary Medicine, Chungbuk National University.

Received: Nov 07, 2022; Revised: Nov 16, 2022; Accepted: Nov 19, 2022

Abstract

Diabetic encephalopathy is a major complication with cognitive impairment and neurodegeneration in patients with type 1 or type 2 diabetes mellitus (DM). DM-induced glucolipotoxicity is a risk factor for Alzheimer’s disease–like phenotype, including amyloidogenesis, tau hyperphosphorylation, and neuronal apoptosis. Although the detailed mechanism underlying the pathogenesis of diabetic encephalopathy remains unclear, mitochondrial oxidative stress is emerging as a key factor for diabetic complications and neurodegeneration. A deeper understanding of the regulatory mechanism of mitochondrial oxidative stress under hyperglycemic conditions will provide insights into the development of therapeutic strategies for diabetic encephalopathy. Here, we review the role of mitochondrial oxidative stress in diabetic encephalopathy and the regulatory mechanisms by which high glucose induces the generation of mitochondrial reactive species oxygen species in neuronal cells. This review also summarizes the mitochondrial-dependent and -independent pathways (O-linked-N-acetylglucosaminylation, calcium, and glycogen synthase kinase 3β signaling) that regulate mitochondrial oxidative stress in a DM model.

Keywords: diabetic encephalopathy; mitochondria; oxidative stress; calcium; glycogen synthase kinase 3 beta

General introduction of diabetic encephalopathy

Diabetic encephalopathy is a chronic complication with neurodegenerative disease in patients with type 1 or type 2 diabetes mellitus (DM) who have been receiving long-term treatment. In 2005, Eric steen et al. described that the dysregulation of insulin and insulin-like growth factor (IGF) causes dementia such as Alzheimer’s disease (AD) and first proposed diabetic encephalopathy as type 3 diabetes [1]. A previous follow-up study investigating the relationships between blood glucose levels and dementia risk confirmed that a higher blood glucose level is a potential risk factor for dementia. Furthermore, the risk of developing dementia was found to be closely associated with high blood glucose levels without DM [2]. An epidemiological study showed that the number of patients with type 2 DM or dementia is strongly increasing and suggested DM as a risk factor for vascular dementia and AD [3]. Several studies using human cell models and animal models demonstrated that both type 1 DM and type 2 DM accompanied neurodegenerative phenotypes and dementia with cognitive impairment [4]. There is no clear information on a precise physiological mechanism underlying the development of diabetic encephalopathy in patients with DM. Nevertheless, the chronic inflammation, oxidative stress, vascular damage, protein dysfunction, low acetylcholinesterase activity, low growth factor levels, and metabolic abnormality occurring during the development of type 2 DM are involved in the occurrence of diabetic encephalopathy [5, 6]. For diabetic encephalopathy treatment, correcting the low acetylcholinesterase activity and oxidative stress was found to be an effective strategy for treating memory loss impairment in a streptozotocin (STZ)-induced mice model. All abbreviations used in this article are listed in Table 1.

Table 1. A list of abbreviations used in this article
Abbreviation Meaning
AD Alzheimer’s disease
AMPK AMP-activated protein kinase
APP Amyloid beta precursor protein
BACE1 Beta-secretase 1
CALHM1 Calcium homeostasis modulator 1 channel
CaMKIV Calcium/calmodulin-dependent protein kinase IV
DM Diabetes mellitus
ER Endoplasmic reticulum
ETC Electron transfer chain
GLUT Glucose transporter
GSK3β Glycogen synthase 3 beta
IGF Insulin-like growth factor
IL Interleukin
LTCC L-type calcium channel
MAPK Mitogen-activated protein kinase
MCU Mitochondrial calcium uniporter
MT2 Melatonin receptor type 2
mTOR Mammalian target of rapamycin
NFTs Neurofibrillary tangles
NMDAR N-methyl-D-aspartate receptor
NO Nitric oxide
Nrf2 Nuclear factor erythroid 2–related factor 2
O-GlcNAcylation O-linked-N-acetylglucosaminylation
PI3K Phosphoinositide 3-kinases
PINK1 PTEN-induced kinase 1
ROS Reactive oxygen species
SIRT1 Sirtuin 1
SNO S-nitrosylation
SOCE Store-operated calcium entry
SOD Superoxide
STAT3 Signal transducer and activator of transcription 3
STZ Streptozotocin
TCA Tricarboxylic acid
TLR Toll-like receptor
VPS26a Vacuolar protein sorting-associated protein 26a
Download Excel Table

Glucose transporter (GLUT) in diabetic encephalopathy

Previous studies have demonstrated that alterations in GLUT and glucose metabolism in patients with DM are the primary factors for AD pathology [7, 8]. Among GLUTs, both GLUT1 and GLUT3 contribute to glucose uptake in the brain. GLUT1 is primarily expressed in glial and endothelial cells, whereas GLUT3 is primarily expressed in neuronal cells [9]. Neuronal GLUT3 expression was found to be decreased in an intracerebral glucose concentration–dependent manner, but astrocytic GLUT1 expression was independent of intracerebral glucose levels [8]. A noninvasive in vivo imaging analysis demonstrated decreased glucose uptake in the brain of rat exposed to an intracerebroventricular injection of amyloid beta [10]. In a Drosophila AD model, GLUT1 overexpression was found to restore glucose uptake, which prolonged life span with neuroprotection through the suppression of proteostasis [7]. Another study showed that GLUT1 deficiency in the blood–brain barrier accelerated neurodegeneration through microvascular degeneration, amyloid beta deposition, and cognitive impairment [11]. In astrocytes, a hyperglycemic condition was found to impair membrane localization of GLUT1, decreasing the glucose bioavailability of astrocytes in an AD model [12]. Considering that glucose uptake and protein glycosylation were increased in high glucose–exposed cells and DM models, further investigation of GLUT regulation response to hyperglycemic conditions is required to clarify the role of GLUTs in diabetic encephalopathy.

Effect of high glucose on neurodegeneration

High glucose–exposed neuronal and nonneuronal cells exhibited altered amyloid beta processing mechanisms through upregulated amyloid beta precursor protein (APP) gene transcription, beta-secretase 1 (BACE1)-mediated APP processing, and downregulated APP degradation [13, 14]. High glucose–activated Akt/glycogen synthase kinase 3β (GSK3β) pathway increased the expression levels of amyloid beta 1–42 and APP-cleaving enzymes, including BACE1 and presenilin 1, which was reversed by melatonin treatment [15]. APP processing under hyperglycemic conditions is controlled in a lipid raft–dependent manner. In neuronal cell line and primary hippocampal neuronal cells, it was observed that high glucose facilitated lipid raft reorganization and APP endocytosis of lipid raft with early endosomal enlargement [14, 16]. Furthermore, high glucose–induced DNA methyltransferase 1-dependent hypermethylation of vacuolar protein sorting-associated protein 26a (VPS26a), which interacts with the BACE1 enzyme. The induction of VPS26 was found to inhibit APP retention in the endosome, which subsequently suppressed amyloid beta and phosphorylated tau levels in neuronal cells [17].

Tau-regulated axonal transport contributes to GLUT4 trafficking, determining insulin resistance in the brain [18]. Moreover, high glucose conditions are closely associated with tau hyperphosphorylation and aggregation, forming neurofibrillary tangles (NFTs). It has been shown that high glucose decreased the expression of the lipid raft caveolae-forming protein Cav-1. Overexpression of Cav-1 inactivated the mammalian target of rapamycin (mTOR)/ribosomal protein S6 kinase beta-1 pathway, suppressing the high glucose–induced tau hyperphosphorylation [19]. Another study showed that toll-like receptor (TLR) 9-activated neuroinflammatory response under diabetic conditions increases tau hyperphosphorylation and neuronal apoptosis through p38 mitogen-activated protein kinase (MAPK) activation [20]. The dimethylbiguanide metformin, an antidiabetic drug, activated protein phosphatase 2a dephosphorylated hyperphosphorylated tau in direct and indirect mTOR-dependent manners [21]. Furthermore, protein glycosylation and S-nitrosylation (SNO) are involved in insoluble NFT formation and neurodegeneration. In a postmortem study, aberrant O-linked-N-acetylglucosaminylation (O-GlcNAcylation) signaling activation was detected in the brain of patients with AD [22]. O-glycosylation of tau at Ser400 under high glucose conditions was found to suppress the hyperphosphorylation of tau at Ser396 and Ser404 [23].

It has been reported that a hyperglycemic condition increases neuroinflammation in type 1 and type 2 DM. An astrocytic cell line exposed to high glucose exhibited increased inflammatory responses with an upregulated secretion of interleukin (IL)-6 and IL-8 [24]. Microglial activation with inflammatory gene upregulation was detected in animal models of both type 1 and type 2 DM [25]. In a BV2 microglial cell line, it was found that high glucose augmented lipopolysaccharide (LPS)-induced TLR4 expression and neuroinflammation [26]. In addition, p38 MAPK inhibition by sesamin treatment prevented from BV2 microglial cell activation and microglia-mediated neurotoxicity [27]. It was also observed that high glucose–stimulated triggering receptor expressed on myeloid cells 2 increased proinflammatory cytokine secretion and NLRP3 inflammasome activation, which play a key role in microglial activation in DM [28]. Collectively, high glucose exacerbates neurodegeneration through amyloidogenesis, hyperphosphorylation, retromer inactivation and neuroinflammation (Fig. 1).

jbtr-23-4-73-g1
Fig. 1. Effect of high glucose on neurodegeneration. High glucose condition mediates APP and APP processing enzymes, such as BACE1 and presenilin, for amyloidogenesis. High glucose induces hyper-phosphorylation of tau protein forming NFT regulated by mTOR/S6K1, p38 MAPK, and GSK3β pathways. High glucose inhibits VPS26a-mediated retromer activation, critical for APP processing. Moreover, high glucose-induced TLRs and ILs activates neuroinflammation of microglia and astrocyte facilitating neurodegeneration. APP, amyloid beta precursor protein; BACE1, beta-secretase 1; mTOR, mammalian target of rapamycin; GSK3β, glycogen synthase kinase 3β; VPS26a, vacuolar protein sorting-associated protein 26a; TLR, toll-like receptor; IL, interleukin; NFT, neurofibrillary tangle; MAPK, mitogen-activated protein kinase; GSK3β, glycogen synthase kinase 3β.
Download Original Figure

Mitochondrial oxidative stress in diabetic encephalopathy

The brain is a high-oxygen-demand organ and sensitive to lipid peroxidation induced by oxidative stress [29]. An imbalanced intracellular redox status and dysfunction of the reactive oxygen species (ROS) antioxidant system play a major role in the oxidative stress of neuronal cells. The endogenous source of ROS is classified into nonmitochondrial and mitochondrial. In particular, mitochondrial ROS is considered as a major endogenous source increasing the ROS of neuronal cells [29]. It has been proposed that there are multiple pathways of mitochondrial ROS generation, which are controlled by mitochondrial respiratory chain complexes such as complex 1 and complex 3 [29]. A recent study described about mitochondrial disease, indicating mitochondrial pathogenesis due to diabetic encephalopathy [30]. Mitochondrial diabetes in diabetic encephalopathy includes mitochondrial DNA mutation, mitochondrial dysfunction, alterations in the expression of axonal transport proteins, and decreased mitochondrial membrane potential [30]. Therefore, several studies have focused on mitochondrial ROS–based preventive and therapeutic strategies for neurodegeneration, which emphasizes the therapeutic potential of mitochondrial regulatory drug development for diabetic encephalopathy.

Cellular ROS is catalyzed and removed by intracellular antioxidant enzymes and small-molecule antioxidants. Mitochondrial superoxide (SOD) inactivates oxygen radicals, forming hydrogen peroxide, which is further catalyzed by glutathione peroxide, catalase, and peroxiredoxin [31]. It has been well documented that oxidative imbalance plays a key role in AD pathogenesis with neuronal cell damage [32]. During the early phase of AD pathogenesis, the intracellular accumulation of amyloid beta elicits mitochondrial dysfunction with failure of energy production metabolism [33]. Moreover, increased intracellular ROS generation induced by mitochondrial dysfunction in AD was found to mediate amyloid beta production and tau hyper-phosphorylation, forming NFTs [33]. Cellular amyloid was commonly detected in the extracellular region, but it was also detected in intracellular organelles, including the Golgi apparatus, endoplasmic reticulum, and mitochondria. In particular, amyloid accumulation in mitochondria affects mitochondrial respiratory function and mitochondrial ROS levels [34]. In addition to neurodegeneration, a recent study demonstrated that mitochondrial stress also activated neuroinflammation in neurons and astrocytes [35]. These findings implicate that mitochondrial oxidative stress is a critical factor for the vicious cycle in neurodegeneration. Based on the therapeutic potential of mitochondrial oxidative stress, several studies have reported natural mitochondrial antioxidant drugs, such as berberine and avocado oil, as potential drug candidates for treating diabetic encephalopathy through the restoration of mitochondrial function [36, 37].

Effect of high glucose on mitochondrial dynamics and nuclear factor erythroid 2–related factor 2 (Nrf2)-regulated antioxidative system

Mitochondrial dynamics change is a representative phenotype of cells exposed to high glucose conditions, associated with various diabetic complications, including diabetic encephalopathy, diabetic retinopathy, diabetic nephropathy, and diabetic heart. In a metabolomics study on an STZ-induced type 1 DM rat model, aberrant alterations of mitochondrial dynamics induced cognitive impairment in diabetic encephalopathy [38]. N-methyl-D-aspartate receptor (NMDAR) activation by high glucose was found to increase calcium and nitric oxide (NO) levels for the SNO of insulin-degrading enzyme and dynamin-related protein (DRP), which mediated mitochondrial dysfunction and neurodegeneration, including synaptic damage, dendritic loss, and cognitive impairment [39]. Other studies showed that in retinal cells, endothelial cells, and cardiomyocytes, high glucose–activated mitochondrial fission, inducing a fusion–fission imbalance and mitochondrial dysfunction through AMP-activated protein kinase (AMPK) inactivation and DRP1 activity regulation [4042]. In podocytes, high glucose inhibited PGC1α- and TFAM-dependent mitochondrial biogenesis and PTEN-induced kinase 1 (PINK1)-dependent mitophagy, as well as mitochondrial fission [43]. In neuronal cells, high glucose primarily upregulated the PINK1–parkin pathway for preventing mitophagy from excessive oxidative stress generation [44]. Melatonin-induced melatonin receptor type 2 (MT2) signaling was found to enhance PINK1–parkin pathway-dependent mitophagy, which is effective in preventing high glucose–induced neuronal cell death [44]. Mitophagy activation by parkin overexpression was found to activate DRP1-mediated mitochondrial fission, which alleviated the high glucose–induced apoptosis of cardiomyocytes [45]. This finding indicates a mutual positive interaction between mitophagy and mitochondrial fission. Under high glucose conditions, neuronal cells have high levels of mitochondrial calcium with endoplasmic reticulum (ER)–mitochondrial contact. High glucose–induced the expression of transglutaminase 2, mediating the interaction between voltage-dependent anion channel 1 and IP receptor for forming the ER–mitochondrial contact [46]. Urolithin A, a microbiome-derived metabolite, was reported as an ER–mitochondrial contact inhibitor, which reduced mitochondrial calcium levels, alleviating mitochondrial oxidative stress in neuronal cells under high glucose conditions [46].

The Nrf2-regulated antioxidant system has a potent capacity of mitochondrial ROS suppression in neuronal cells [47]. In the mouse hippocampal neuron cell line HT22, high glucose–induced ROS generation with Nrf2 and heme oxygenase 1 suppression. Treatment with Nrf2 activator suppressed the high glucose–induced neuronal cell death [47]. In rat retinal cells, it was found that high glucose decreased the expression of Nrf2 target genes, including glutathione and catalase with redox regulatory capacity [48]. In Nrf2 knock down and knock out assays, it was demonstrated that Nrf2 is a major protective factor for high glucose–induced mitochondrial oxidative stress and cellular apoptosis [49]. It has been well documented that sirtuin 1 (SIRT1) is a critical regulator of Nrf2 activity. It was found that activation of the salvianolic acid–induced SIRT1–Nrf2 pathway suppressed oxidative stress and neuronal degeneration [50]. High glucose–induced SIRT1 activation reduced neuronal apoptosis through p53 deacetylation in STZ-induced diabetic mice and SH-SY5Y neuroblastoma cell line [51]. Moreover, SIRT1 has been reported as a suppressive factor of insulin resistance, which is closely associated with SIRT1-regulated mitochondrial respiratory chain activity. In fact, SIRT1 overexpression was found to restore the activity of high glucose–reduced mitochondrial complex 1, which was reversed by SIRT3 knock down [52]. Nevertheless, there is a need for further investigation of the detailed mechanism by which high glucose regulates SIRT1 activity related to mitochondrial function and Nrf2 pathway to develop SIRT1-dependent therapeutic strategies for diabetic encephalopathy. Collectively, high glucose regulates mitochondrial dynamics and 2).

jbtr-23-4-73-g2
Fig. 2. Effect of high glucose on mitochondrial dynamics and Nrf2 activation in neuronal cells. High glucose activates DRP1-mediated mitochondrial fission but inactivates PINK1/parkin pathway-mediated mitophagy. High glucose suppresses PGC1α and TFAM-mediated mitochondrial biogenesis and reduces mitochondrial contents in neuronal cells. High glucose stimulates TGM2-mediated ER-mitochondrial interaction increasing mitochondrial calcium levels. In addition, high glucose-inhibited SIRT1/Nrf2 pathway stimulates mitochondrial ROS accumulation and dysfunction. DRP, dynamin-related protein; PINK1, PTEN-induced kinase 1; SIRT1, sirtuin 1; Nrf2, nuclear factor erythroid 2–related factor 2; ER, endoplasmic reticulum; ROS, reactive oxygen species.
Download Original Figure

Role of calcium in diabetic encephalopathy

Calcium homeostasis in brain tissue has been considered to be the major cause of diabetic encephalopathy [53]. Several studies have demonstrated that intracellular Ca2+ levels were increased in the brain of patients with diabetic encephalopathy, and large amounts of calcium inflow cause calcium homeostasis disorder in the brain [54]. Intracellular calcium is maintained by store-operated calcium entry (SOCE), the process by which emptying of ER calcium stores causes the influx of calcium across the plasma membrane for calcium signaling. Disorders related to SOCE cause the dysregulation of Ca2+ signaling in neuronal cells [55]. The L-type calcium channel (LTCC) is a plasma membrane–localized voltage-dependent calcium channel and is distributed in neuronal cell bodies [56]. LTCC has four isoforms, and CaV1.2 expression was increased in diabetic neurodegeneration by excessive calcium absorption [57]. Hence, the dysregulation of LTCC has been reported in various neurodegenerative diseases, including AD [57]. Nimodipine, an LTCC blocker, improves diabetic encephalopathy by blocking calcium channels, reducing the expression level of CaV1.2 and absorption of mitochondrial Ca2+ [57]. Calcium/calmodulin-dependent protein kinase IV (CaMKIV) activated by calmodulin is responsible for regulating the synthesis and release of neurotransmitters, and an increase in Ca2+ levels induces the activation of CaMKIV, inhibiting synaptic formation and resulting in neuronal loss and several neurodegenerative disorders, including AD and Parkinson’s disease [58, 59]. The calcium homeostasis modulator 1 channel (CALHM1) is a voltage-gated channel that increases cytosolic calcium influx in response to decreases in extracellular calcium levels. The role of CALHM1 in neurodegenerative diseases remains unclear, and a polymorphism has been detected in the genome of patients with AD, which was expected to disrupt calcium homeostasis [60]. Therefore, the impairment of calcium signaling or calcium channel regulation causes neurodegenerative pathogenesis under DM conditions, resulting in diabetic encephalopathy.

Calcium signals are involved in various functions of mitochondria and mtROS regulation. Mitochondrial Ca2+ absorption affects the tricarboxylic acid (TCA) cycle and electron transfer chain (ETC) activity to regulate ATP production and generate ROS signals [61]. Excessive calcium levels stimulate respiratory chain activity, leading to higher amounts of ROS [62]. Mitochondrial Ca2+ directly stimulates mtROS-producing enzymes such as glycerol phosphate and α-ketoglutaric acid dehydrogenase to form mtROS [62]. Ca2+ induces the generation of NO through a regulator of NO synthases, and the generated NO can block complex IV, causing mtROS generation [63]. Furthermore, the continuous calcium load opens the pathway in the inner mitochondrial membrane, ruptures the outer mitochondrial membrane, and reduces the mitochondrial membrane potential [64]. The reduced mitochondrial membrane potential also causes the generation of ROS and the generation of more Ca2+ levels, separating cytochrome c from the inner membrane cardiolipin. Eventually, mitochondrial permeable transition pores are opened, and the apoptosis factor Ca2+ is released into the cytoplasm. Calcium homeostasis in mitochondria is closely related to neurodegenerative diseases. Recent studies have reported mitochondrial calcium uniporter (MCU) as a candidate treatment target for neurodegenerative diseases through mitochondrial Ca2+ regulation [65]. It has been reported that TG-2112x, the MCU inhibitor, can partially inhibit mitochondrial calcium absorption and protect neurons [66]. However, ROS produced by excessive calcium increases the release of more calcium to the mitochondria through the calcium channel between ER and the mitochondria, and calcium is transferred to the mitochondrial substrate through the MCU. MCU oxidation by ROS induces the S-glutathionylation of the Cys-97 residue of the MCU protein and exhibits continuous MCU channel activity with higher Ca2+ absorption in the mitochondria, increased mitochondrial ROS generation, and improved mitochondrial Ca2+ overload–induced apoptosis [67]. Moreover, high glucose–induced ER–mitochondrial contact was found to increase mitochondrial Ca2+ accumulation, which is critical for the pathogenesis of diabetic encephalopathy. Urolithin A was found to disrupt the ER–mitochondrial contact and alleviate neurodegeneration [46]. Excess Ca2+ continuously activates the respiratory chain and calcium channels of mitochondria, resulting in mitochondrial ROS generation and neurodegeneration. Therefore, inhibition of mtROS production through the regulation of calcium homeostasis is a potential treatment for diabetic encephalopathy.

Role of O-linked-N-acetylglucosaminylation (O-GlcNAcylation) in diabetic encephalopathy

O-GlcNAcylation is a posttranslational modification process that plays a vital role in signaling and biogenesis metabolism [68]. Increased O-GlcNAcylation causes mitochondrial ROS induction and decreases ATP production and membrane potential, resulting in dysfunction and inflammatory reactions of mitochondria and apoptosis under hyperglycemia. Increased O-GlcNAcylation of TCA cycle enzymes and ETC complexes was found to increase ATP production but reduce ROS production in mouse mitochondria [69]. However, another study confirmed that the rate of O-GlcNAcylation in the human body was increased under hyperglycemia [70]. Excessive O-GlcNAcylation of mitochondrial proteins inactivated ETC complexes, leading to mitochondrial dysfunction in cardiomyocytes under hyperglycemia [71]. Increased O-GlcNAcylation of calcium/CaMKII in ventricular myocytes was found to increase calcium leakage and limit reabsorption, resulting in arrhythmia in diabetic hyperglycemia [72]. Moreover, increased O-GlcNAcylation of COXI and NDUFA9 protein was found to reduce ATP production and mitochondrial membrane potential, causing mitochondrial damage by generating ROS [73]. Increased O-GlcNAcylation in vascular endothelial cells under hyperglycemia was due to reduced expression of the microRNA 200a/200b family, which inhibited OGT expression. Increase of O-GlcNAcylation by OGT was found to elevate the expressions of intercellular adhesion molecular 1, vesicular cell adhesion molecular 1, and e-selectin, resulting in endothelial inflammation [74]. Conversely, an increase of O-GlcNAcylation in neuronal cells under high glucose conditions was also observed. It was found that increased O-GlcNAcylation of milton, the mitochondrial motor protein, caused a reduction of mitochondrial transport to the synapse in mouse neurons under hyperglycemia [75]. Moreover, upregulated O-GlcNAcylation of the AMPA receptor subunits GluA1 and GluA2 mediated the NMDAR-dependent long-term potentiation and long-term depression in the hippocampal synapse [76]. Nonetheless, the role of O-GlcNAcylation in diabetic encephalopathy and its relationship with mitochondrial ROS has not yet been clarified.

It has been reported that chronic hyperglycemia causes insulin resistance in the brain, resulting in reduced O-GlcNAcylation in patients with AD [77]. Reduced O-GlcNAcylation of milton was found to cause the mislocalization of mitochondria in neuronal cells. Subsequently, ATP and Ca2+ supply was impaired, which hindered synaptic formation [78]. Because the tau protein has more serine/threonine phosphorylation sites, where O-GlcNAcylation occurs, reduced O-GlcNAcylation in the AD brain with impaired glucose metabolism is closely associated with tau pathology, including the hyperphosphorylation of tau protein [79]. Therefore, the regulation of O-GlcNAcylation is an extremely important approach for AD treatment. O-GlcNAcase is a potential treatment for recovering the reduced O-GlcNAcylation in the brain of patients with AD. Treatment with the O-GlcNAcase inhibitor thiamet-G reduced the separation of proteins from O-GlcNAc by inhibiting the function of OGA. Treatment with thiamet-G also improved the membrane potential or ATP production function of mitochondria in the brain [78]. Moreover, increased O-GlcNAcylation of tau protein was found to inhibit hyperphosphorylation, suppressing the formation of NFTs [80].

Role of glycogen synthase kinase 3β (GSK3β) signaling in diabetic encephalopathy

In DM, GSK3β is a vital signaling molecule in glycogen synthesis and blood glucose control, and recent studies have demonstrated that the GSK3β signaling pathway is closely related to diabetes and neurodegeneration. When insulin and insulin receptors bind each other, phosphoinositide 3-kinases (PI3K)/Akt signaling inhibits the phosphorylation of GSK3β for tau dephosphorylation. In diabetes, insulin resistance was found to impair the activation of the PI3K/Akt pathway, causing GSK3β activation and oxidative stress generation [81]. Similarly, the dysfunction of insulin or IGF-1 signaling system eventually stimulated GSK3β by blocking the PI3K/Akt/Wnt signaling pathway [82]. It was found that Wnt3a-inhibited GSK3β induced β-catenin accumulation for TCF activation, which enhanced the production of NeuroD1-stimulated insulin [83].

GSK3β activation directly promotes neurodegeneration through tau hyperphosphorylation, neuroinflammation, and neuronal dysfunction. It has been reported that GSK3β is involved in synaptic plasticity and hippocampal long-term potentiation, a mechanism essential for memory formation [84]. Another study showed that patients with AD exhibited upregulated GSK3β activity, suggesting that hyperactivation of GSK3β is an early event that precedes and accompanies the formation of NFTs and other tau-positive inclusions [85]. There is also extensive research showing that hyperactivated GSK3β signaling is critical for amyloidogenesis and microglial activation in AD pathogenesis [84]. In fact, several GSK3β inhibitors, including lithium chloride, are currently being investigated in preclinical and clinical trials for AD due to their anti-neurodegeneration properties [86].

The GSK3β signaling pathway is essential for mitochondrial oxidative stress and mitochondrial regulation. GSK3β-inhibited β-catenin causes mitochondrial damage due to oxidative stress, which is critical for the pathogenesis of diabetic retinal degeneration. This finding indicates that active β-catenin signaling can be a diagnostic biomarker for the early onset of diabetic retinopathy [87]. IGF-1-mediated GSK3β inactivation promoted mitochondrial biogenesis through upregulation of the transcriptional coactivators peroxisome PGC1β and PGC1-related coactivator. Furthermore, the IGF-1/GSK3β pathway plays a key role in Bcl-2 interacting protein 3-dependent mitophagy and Nrf2 stabilization, involving in mitochondrial ROS homeostasis [88]. In addition, the Akt/GSK3β-regulated signal transducer and activator of transcription 3 (STAT3) plays an essential role in mitochondrial function and oxidative stress caused by DM. STAT3-Cyp-D interaction plays an essential role in regulating ROS generation and mitochondrial function by regulating mitochondrial permeability transition pores, ATP production, and ETC. This finding indicates that inhibition of the GSK3β-STAT3 pathway is a potential strategy for improving mitochondrial function and anti-apoptosis [82]. In the SH-SY5Y cell line, AMPK activation stimulated GSK3β signaling, inducing the activation of mitochondrial transcription factor A, which leads to the transcription/translation of mitochondrial DNA, promoting mitochondrial biosynthesis [89]. Considering that GSK3β acts as a key player linking between DM and neurodegeneration, GSK3β-based therapeutic strategies are promising for the prevention and treatment of diabetic encephalopathy.

Conclusion

Although the respective role of mitochondrial oxidative stress in DM and neurodegeneration have been well documented, there is a lack of a clear understanding of the detailed relationships between DM condition and diabetic encephalopathy pathogenesis. For developing preventive and therapeutic strategies of diabetic encephalopathy in DM patients, it is necessary to further investigate mitochondrial regulatory mechanism in neuronal cells under hyperglycemic condition. Finally, we suggest mitochondrial oxidative stress regulatory targets in diabetic encephalopathy, such as calcium, O-GlcNAcylation, and GSK3β signaling (Fig. 3).

jbtr-23-4-73-g3
Fig. 3. Role of calcium, O-GlcNAcylation, and GSK3β signaling in mitochondrial oxidative stress and diabetic encephalopathy pathogenesis. Hyperglycemic condition in diabetes mellitus regulates calcium, O-GlcNAcylation, and GSK3β signaling, which plays a critical role in mitochondrial reactive oxygen species accumulation through mitochondrial metabolism, dynamics, and antioxidant system. Impairment of mitochondrial ROS homeostasis causes diabetic encephalopathy with neuronal cell death, amyloidogenesis, and tau hyper-phosphorylation. CALHM1, calcium homeostasis modulator 1 channel; ER, endoplasmic reticulum; O-GlcNAcylation, O-linked-N-acetylglucosaminylation; GSK3β, glycogen synthase kinase 3β; ROS, reactive oxygen species.
Download Original Figure

Conflict of Interest

No potential conflict of interest relevant to this article was reported.

Acknowledgements

This work was supported by the research grant of the Chungbuk National University in 2020 and “Innovation Strategy (RIS)” through the National Research Foundation of Korea (NRF) funded by the Ministry of Education (MOE, 2021RIS-001).

Ethics Approval

Not applicable.

REFERENCES

1.

Steen E, Terry BM, Rivera EJ, Cannon JL, Neely TR, Tavares R, Xu XJ, Wands JR, de la Monte SM. Impaired insulin and insulin-like growth factor expression and signaling mechanisms in Alzheimer’s disease – is this type 3 diabetes? J Alzheimers Dis 2005;7:63-80.

2.

Crane PK, Walker R, Hubbard RA, Li G, Nathan DM, Zheng H, Haneuse S, Craft S, Montine TJ, Kahn SE, McCormick W, McCurry SM, Bowen JD, Larson EB. Glucose levels and risk of dementia. N Engl J Med 2013;369:540-548.

3.

Prince M, Bryce R, Albanese E, Wimo A, Ribeiro W, Ferri CP. The global prevalence of dementia: a systematic review and metaanalysis. Alzheimers Dement 2013;9:63-75.e2.

4.

Zilliox LA, Chadrasekaran K, Kwan JY, Russell JW. Diabetes and cognitive impairment. Curr Diab Rep 2016;16:87.

5.

Díaz-Gerevini GT, Repossi G, Dain A, Tarres MC, Das UN, Eynard AR. Cognitive and motor perturbations in elderly with longstanding diabetes mellitus. Nutrition 2014;30:628-635.

6.

Vieira LL, de Lima Gomes Soares R, da Silva Felipe SM, de Moura FC, de Castro Brito GA, Pacheco C, Soares PM. Physiological targets for the treatment of diabetic encephalopathy. Cent Nerv Syst Agents Med Chem 2017;17:78-86.

7.

Duran-Aniotz C, Hetz C. Glucose metabolism: a sweet relief of Alzheimer’s disease. Curr Biol 2016;26:R806-R809.

8.

An Y, Varma VR, Varma S, Casanova R, Dammer E, Pletnikova O, Chia CW, Egan JM, Ferrucci L, Troncoso J, Levey AI, Lah J, Seyfried NT, Legido-Quigley C, O’Brien R, Thambisetty M. Evidence for brain glucose dysregulation in Alzheimer’s disease. Alzheimers Dement 2018;14:318-329.

9.

Chen Z, Zhong C. Decoding Alzheimer’s disease from perturbed cerebral glucose metabolism: implications for diagnostic and therapeutic strategies. Prog Neurobiol 2013;108:21-43.

10.

Chen P, Shen Z, Wang Q, Zhang B, Zhuang Z, Lin J, Shen Y, Chen Y, Dai Z, Wu R. Reduced cerebral glucose uptake in an Alzheimer’s rat model with glucose-weighted chemical exchange saturation transfer imaging. Front Aging Neurosci 2021;13:618690.

11.

Winkler EA, Nishida Y, Sagare AP, Rege SV, Bell RD, Perlmutter D, Sengillo JD, Hillman S, Kong P, Nelson AR, Sullivan JS, Zhao Z, Meiselman HJ, Wendy RB, Soto J, Dale Abel E, Makshanoff J, Zuniga E, De Vivo DC, Zlokovic BV. GLUT1 reductions exacerbate Alzheimer’s disease vasculo-neuronal dysfunction and degeneration. Nat Neurosci 2015;18:521-530.

12.

Hendrix RD, Ou Y, Davis JE, Odle AK, Groves TR, Allen AR, Childs GV, Barger SW. Alzheimer amyloid-β-peptide disrupts membrane localization of glucose transporter 1 in astrocytes: implications for glucose levels in brain and blood. Neurobiol Aging 2021;97:73-88.

13.

Yang Y, Wu Y, Zhang S, Song W. High glucose promotes Aβ production by inhibiting APP degradation. PLOS ONE 2013;8:e69824.

14.

Lee HJ, Ryu JM, Jung YH, Lee SJ, Kim JY, Lee SH, Hwang IK, Seong JK, Han HJ. High glucose upregulates BACE1-mediated Aβ production through ROS-dependent HIF-1α and LXRα/ABCA1-regulated lipid raft reorganization in SK-N-MC cells. Sci Rep 2016;6:36746.

15.

Nopparat C, Chaopae W, Boontem P, Sopha P, Wongchitrat P, Govitrapong P. Melatonin attenuates high glucose-induced changes in beta amyloid precursor protein processing in human neuroblastoma cells. Neurochem Res 2022;47:2568-2579.

16.

Chae CW, Lee HJ, Choi GE, Jung YH, Kim JS, Lim JR, Kim SY, Hwang IK, Seong JK, Han HJ. High glucose-mediated PICALM and mTORC1 modulate processing of amyloid precursor protein via endosomal abnormalities. Br J Pharmacol 2020;177:3828-3847.

17.

Chae CW, Choi GE, Jung YH, Lim JR, Cho JH, Yoon JH, Han HJ. High glucose-mediated VPS26a down-regulation dysregulates neuronal amyloid precursor protein processing and tau phosphorylation. Br J Pharmacol 2022;179:3934-3950.

18.

Gonçalves RA, Wijesekara N, Fraser PE, De Felice FG. The link between tau and insulin signaling: implications for Alzheimer’s disease and other tauopathies. Front Cell Neurosci 2019; 13:17.

19.

Wu J, Zhou SL, Pi LH, Shi XJ, Ma LR, Chen Z, Qu ML, Li X, Nie SD, Liao DF, Pei JJ, Wang S. High glucose induces formation of tau hyperphosphorylation via Cav-1-mTOR pathway: a potential molecular mechanism for diabetes-induced cognitive dysfunction. Oncotarget 2017; 8:40843-40856.

20.

Sun Y, Xiao Q, Luo C, Zhao Y, Pu D, Zhao K, Chen J, Wang M, Liao Z. High-glucose induces tau hyperphosphorylation through activation of TLR9-P38MAPK pathway. Exp Cell Res 2017;359:312-318.

21.

Kickstein E, Krauss S, Thornhill P, Rutschow D, Zeller R, Sharkey J, Williamson R, Fuchs M, Köhler A, Glossmann H, Schneider R, Sutherland C, Schweiger S. Biguanide metformin acts on tau phosphorylation via mTOR/protein phosphatase 2A (PP2A) signaling. Proc Natl Acad Sci USA 2010;107:21830-21835.

22.

Zhu Y, Shan X, Yuzwa SA, Vocadlo DJ. The emerging link between O-GlcNAc and Alzheimer disease. J Biol Chem 2014;289:34472-34481.

23.

Smet-Nocca C, Broncel M, Wieruszeski JM, Tokarski C, Hanoulle X, Leroy A, Landrieu I, Rolando C, Lippens G, Hackenberger CPR. Identification of O-GlcNAc sites within peptides of the tau protein and their impact on phosphorylation. Mol Biosyst 2011;7:1420-1429.

24.

Bahniwal M, Little JP, Klegeris A. High glucose enhances neurotoxicity and inflammatory cytokine secretion by stimulated human astrocytes. Curr Alzheimer Res 2017;14:731-741.

25.

Rom S, Zuluaga-Ramirez V, Gajghate S, Seliga A, Winfield M, Heldt NA, Kolpakov MA, Bashkirova YV, Sabri AK, Persidsky Y. Hyperglycemia-driven neuroinflammation compromises BBB leading to memory loss in both diabetes mellitus (DM) type 1 and type 2 mouse models. Mol Neurobiol 2019;56:1883-1896.

26.

Hung HC, Tsai SF, Sie SR, Kuo YM. High glucose enhances lipopolysaccharide-induced inflammation in cultured BV2 microglial cell line. Immun Inflamm Dis 2022;10:e610.

27.

Kongtawelert P, Kaewmool C, Phitak T, Phimphilai M, Pothacharoen P, Shwe TH. Sesamin protects against neurotoxicity via inhibition of microglial activation under high glucose circumstances through modulating p38 and JNK signaling pathways. Sci Rep 2022;12:11296.

28.

Li Y, Long W, Gao M, Jiao F, Chen Z, Liu M, Yu L. TREM2 regulates high glucose-induced microglial inflammation via the NLRP3 signaling pathway. Brain Sci 2021;11:896.

29.

Kim GH, Kim JE, Rhie SJ, Yoon S. The role of oxidative stress in neurodegenerative diseases. Exp Neurobiol 2015;24:325-340.

30.

Eslami Gharaati M, Nahavandi A, Baluchnejad Mojarad T, Roghani M. Diabetic encephalopathy affecting mitochondria and axonal transport proteins. Basic Clin Neurosci 2020;11:781-793.

31.

Power JHT, Blumbergs PC. Cellular glutathione peroxidase in human brain: cellular distribution, and its potential role in the degradation of Lewy bodies in Parkinson’s disease and dementia with Lewy bodies. Acta Neuropathol 2009;117:63-73.

32.

Wang X, Wang W, Li L, Perry G, Lee H, Zhu X. Oxidative stress and mitochondrial dysfunction in Alzheimer’s disease. Biochim Biophys Acta 2014;1842:1240-1247.

33.

Zhao Y, Zhao B. Oxidative stress and the pathogenesis of Alzheimer’s disease. Oxid Med Cell Longev 2013;2013:316523.

34.

Picone P, Nuzzo D, Caruana L, Scafidi V, Di Carlo M. Mitochondrial dysfunction: different routes to Alzheimer’s disease therapy. Oxid Med Cell Longev 2014;2014:780179.

35.

Liyanagamage DSNK, Martinus RD. Role of mitochondrial stress protein HSP60 in diabetes-induced neuroinflammation. Mediators Inflamm 2020;2020:8073516.

36.

Tian L, Ri H, Qi J, Fu P. Berberine elevates mitochondrial membrane potential and decreases reactive oxygen species by inhibiting the Rho/ROCK pathway in rats with diabetic encephalopathy. Mol Pain 2021;17:1744806921996101.

37.

Ortiz-Avila O, Esquivel-Martínez M, Olmos-Orizaba BE, Saavedra-Molina A, Rodriguez- Orozco AR, Cortés-Rojo C. Avocado oil improves mitochondrial function and decreases oxidative stress in brain of diabetic rats. J Diabetes Res 2015;2015:485759.

38.

Zhou Y, Lian S, Zhang J, Lin D, Huang C, Liu L, Chen Z. Mitochondrial perturbation contributing to cognitive decline in streptozotocin-induced type 1 diabetic rats. Cell Physiol Biochem 2018;46:1668-1682.

39.

Akhtar MW, Sanz-Blasco S, Dolatabadi N, Parker J, Chon K, Lee MS, Soussou W, McKercher SR, Ambasudhan R, Nakamura T, Lipton SA. Elevated glucose and oligomeric β-amyloid disrupt synapses via a common pathway of aberrant protein S-nitrosylation. Nat Commun 2016;7: 10242.

40.

Trudeau K, Molina AJA, Roy S. High glucose induces mitochondrial morphology and metabolic changes in retinal pericytes. Invest Ophthalmol Vis Sci 2011;52:8657-8664.

41.

Zheng Y, Luo A, Liu X. The imbalance of mitochondrial fusion/fission drives high-glucose-induced vascular injury. Biomolecules 2021;11:1779.

42.

Wu QR, Zheng DL, Liu PM, Yang H, Li LA, Kuang SJ, Lai YY, Rao F, Xue YM, Lin JJ, Liu SX, Chen CB, Deng CY. High glucose induces Drp1-mediated mitochondrial fission via the Orai1 calcium channel to participate in diabetic cardiomyocyte hypertrophy. Cell Death Dis 2021;12:216.

43.

Audzeyenka I, Rachubik P, Typiak M, Kulesza T, Topolewska A, Rogacka D, Angielski S, Saleem MA, Piwkowska A. Hyperglycemia alters mitochondrial respiration efficiency and mitophagy in human podocytes. Exp Cell Res 2021;407:112758.

44.

Onphachanh X, Lee HJ, Lim JR, Jung YH, Kim JS, Chae CW, Lee SJ, Gabr AA, Han HJ. Enhancement of high glucose-induced PINK1 expression by melatonin stimulates neuronal cell survival: involvement of MT2/Akt/NF-κB pathway. J Pineal Res 2017;63:e12427.

45.

Kobayashi S, Zhao F, Zhang Z, Kobayashi T, Huang Y, Shi B, Wu W, Liang Q. Mitochondrial fission and mitophagy coordinately restrict high glucose toxicity in cardiomyocytes. Front Physiol 2020;11:604069.

46.

Lee HJ, Jung YH, Choi GE, Kim JS, Chae CW, Lim JR, Kim SY, Yoon JH, Cho JH, Lee SJ, Han HJ. Urolithin A suppresses high glucose-induced neuronal amyloidogenesis by modulating TGM2-dependent ER-mitochondria contacts and calcium homeostasis. Cell Death Differ 2021;28:184-202.

47.

Zhao J, Liu L, Li X, Zhang L, Lv J, Guo X, Chen H, Zhao T. Neuroprotective effects of an Nrf2 agonist on high glucose-induced damage in HT22 cells. Biol Res 2019;52:53.

48.

Albert-Garay JS, Riesgo-Escovar JR, Salceda R. High glucose concentrations induce oxidative stress by inhibiting Nrf2 expression in rat Muller retinal cells in vitro. Sci Rep 2022;12:1261.

49.

He X, Kan H, Cai L, Ma Q. Nrf2 is critical in defense against high glucose-induced oxidative damage in cardiomyocytes. J Mol Cell Cardiol 2009;46:47-58.

50.

Zhang X, Wu Q, Lu Y, Wan J, Dai H, Zhou X, Lv S, Chen X, Zhang X, Hang C, Wang J. Cerebroprotection by salvianolic acid B after experimental subarachnoid hemorrhage occurs via Nrf2- and SIRT1-dependent pathways. Free Radic Biol Med 2018;124:504-516.

51.

Shi X, Pi L, Zhou S, Li X, Min F, Wang S, Liu Z, Wu J. Activation of sirtuin 1 attenuates high glucose-induced neuronal apoptosis by deacetylating p53. Front Endocrinol 2018;9:274.

52.

Zhang HH, Ma XJ, Wu LN, Zhao YY, Zhang PY, Zhang YH, Shao MW, Liu F, Li F, Qin GJ. SIRT1 attenuates high glucose-induced insulin resistance via reducing mitochondrial dysfunction in skeletal muscle cells. Exp Biol Med 2015;240:557-565.

53.

Marambaud P, Dreses-Werringloer U, Vingtdeux V. Calcium signaling in neurodegeneration. Mol Neurodegener 2009;4:20.

54.

Ge M, Zhang J, Chen S, Huang Y, Chen W, He L, Zhang Y. Role of calcium homeostasis in Alzheimer’s disease. Neuropsychiatr Dis Treat 2022;18:487-498.

55.

Secondo A, Bagetta G, Amantea D. On the role of store-operated calcium entry in acute and chronic neurodegenerative diseases. Front Mol Neurosci 2018;11:87.

56.

Yagami T, Kohma H, Yamamoto Y. L-type voltage-dependent calcium channels as therapeutic targets for neurodegenerative diseases. Curr Med Chem 2012;19:4816-4827.

57.

Singhal K, Sandhir R. L-type calcium channel blocker ameliorates diabetic encephalopathy by modulating dysregulated calcium homeostasis. J Neurosci Res 2015;93:296-308.

58.

Trombetta-Lima M, Sabogal-Guáqueta AM, Dolga AM. Mitochondrial dysfunction in neurodegenerative diseases: a focus on iPSC-derived neuronal models. Cell Calcium 2021;94:102362.

59.

Sałaciak K, Koszałka A, Żmudzka E, Pytka K. The calcium/calmodulin-dependent kinases II and IV as therapeutic targets in neurodegenerative and neuropsychiatric disorders. Int J Mol Sci 2021;22:4307.

60.

Schrank S, Barrington N, Stutzmann GE. Calcium-handling defects and neurodegenerative disease. Cold Spring Harb Perspect Biol 2020;12:a035212.

61.

Sena LA, Chandel NS. Physiological roles of mitochondrial reactive oxygen species. Mol Cell 2012;48:158-167.

62.

Görlach A, Bertram K, Hudecova S, Krizanova O. Calcium and ROS: a mutual interplay. Redox Biol 2015;6:260-271.

63.

Brookes PS, Yoon Y, Robotham JL, Anders MW, Sheu SS. Calcium, ATP, and ROS: a mitochondrial love-hate triangle. Am J Physiol Cell Physiol 2004;287:C817-C833.

64.

Bachurin SO, Shevtsova EF, Makhaeva GF, Grigoriev VV, Boltneva NP, Kovaleva NV, Lushchekina SV, Shevtsov PN, Neganova ME, Redkozubova OM, Bovina EV, Gabrelyan AV, Fisenko VP, Sokolov VB, Aksinenko AY, Echeverria V, Barreto GE, Aliev G. Novel conjugates of aminoadamantanes with carbazole derivatives as potential multitarget agents for AD treatment. Sci Rep 2017;7:45627.

65.

Calvo-Rodriguez M, Bacskai BJ. Mitochondria and calcium in Alzheimer’s disease: from cell signaling to neuronal cell death. Trends Neurosci 2021;44:136-151.

66.

Esteras N, Abramov AY. Mitochondrial calcium deregulation in the mechanism of beta-amyloid and tau pathology. Cells 2020;9:2135.

67.

Dong Z, Shanmughapriya S, Tomar D, Siddiqui N, Lynch S, Nemani N, Breves SL, Zhang X, Tripathi A, Palaniappan P, Riitano MF, Worth AM, Seelam A, Carvalho E, Subbiah R, Jaña F, Soboloff J, Peng Y, Cheung JY, Joseph SK, Caplan J, Rajan S, Stathopulos PB, Madesh M. Mitochondrial Ca2+ uniporter is a mitochondrial luminal redox sensor that augments MCU channel activity. Mol Cell 2017;65:1014-1028.E7.

68.

Akella NM, Ciraku L, Reginato MJ. Fueling the fire: emerging role of the hexosamine biosynthetic pathway in cancer. BMC Biol 2019;17:52.

69.

Dontaine J, Bouali A, Daussin F, Bultot L, Vertommen D, Martin M, Rathagirishnan R, Cuillerier A, Horman S, Beauloye C, Gatto L, Lauzier B, Bertrand L, Burelle Y. The intra-mitochondrial O-GlcNAcylation system rapidly modulates OXPHOS function and ROS release in the heart. Commun Biol 2022;5:349.

70.

Copeland RJ, Bullen JW, Hart GW. Cross-talk between GlcNAcylation and phosphorylation: roles in insulin resistance and glucose toxicity. Am J Physiol Endocrinol Metab 2008;295:E17- E28.

71.

Akinbiyi EO, Abramowitz LK, Bauer BL, Stoll MSK, Hoppel CL, Hsiao CP, Hanover JA, Mears JA. Blocked O-GlcNAc cycling alters mitochondrial morphology, function, and mass. Sci Rep 2021;11:22106.

72.

Hegyi B, Fasoli A, Ko CY, Van BW, Alim CC, Shen EY, Ciccozzi MM, Tapa S, Ripplinger CM, Erickson JR, Bossuyt J, Bers DM. CaMKII serine 280 O-GlcNAcylation links diabetic hyperglycemia to proarrhythmia. Circ Res 2021;129:98-113.

73.

Hu Y, Suarez J, Fricovsky E, Wang H, Scott BT, Trauger SA, Han W, Hu Y, Oyeleye MO, Dillmann WH. Increased enzymatic O-GlcNAcylation of mitochondrial proteins impairs mitochondrial function in cardiac myocytes exposed to high glucose. J Biol Chem 2009;284:547-555.

74.

Lo WY, Yang WK, Peng CT, Pai WY, Wang HJ. MicroRNA-200a/200b modulate high glucose-induced endothelial inflammation by targeting O-linked N-acetylglucosamine transferase expression. Front Physiol 2018;9:355.

75.

Pekkurnaz G, Trinidad JC, Wang X, Kong D, Schwarz TL. Glucose regulates mitochondrial motility via Milton modification by O-GlcNAc transferase. Cell 2014;158:54-68.

76.

Taylor EW, Wang K, Nelson AR, Bredemann TM, Fraser KB, Clinton SM, Puckett R, Marchase RB, Chatham JC, McMahon LL. O-GlcNAcylation of AMPA receptor GluA2 is associated with a novel form of long-term depression at hippocampal synapses. J Neurosci 2014;34:10-21.

77.

Barone E, Di Domenico F, Cassano T, Arena A, Tramutola A, Lavecchia MA, Coccia R, Allen Butterfield D, Perluigi M. Impairment of biliverdin reductase-A promotes brain insulin resistance in Alzheimer disease: a new paradigm. Free Radic Biol Med 2016;91:127-142.

78.

Pinho TS, Correia SC, Perry G, Ambrósio AF, Moreira PI. Diminished O-GlcNAcylation in Alzheimer’s disease is strongly correlated with mitochondrial anomalies. Biochim Biophys Acta Mol Basis Dis 2019;1865:2048-2059.

79.

Liu F, Shi J, Tanimukai H, Gu J, Gu J, Grundke-Iqbal I, Iqbal K, Gong CX. Reduced O-GlcNAcylation links lower brain glucose metabolism and tau pathology in Alzheimer’s disease. Brain 2009;132:1820-1832.

80.

Li X, Han J, Bujaranipalli S, He J, Kim EY, Kim H, Im JH, Cho WJ. Structure-based discovery and development of novel O-GlcNAcase inhibitors for the treatment of Alzheimer’s disease. Eur J Med Chem 2022;238:114444.

81.

Medina M, Avila J. Glycogen synthase kinase-3 (GSK-3) inhibitors for the treatment of Alzheimer’s disease. Curr Pharm Des 2010;16:2790-2798.

82.

Wang H, Ai J, Shopit A, Niu M, Ahmed N, Tesfaldet T, Tang Z, Li X, Jamalat Y, Chu P, Peng J, Ma X, Qaed E, Han G, Zhang W, Wang J, Tang Z. Protection of pancreatic β-cell by phosphocreatine through mitochondrial improvement via the regulation of dual AKT/IRS-1/GSK-3β and STAT3/Cyp-D signaling pathways. Cell Biol Toxicol 2022;38:531-551.

83.

Lee J, Kim K, Yu SW, Kim EK. Wnt3a upregulates brain-derived insulin by increasing NeuroD1 via Wnt/β-catenin signaling in the hypothalamus. Mol Brain 2016;9:24.

84.

Lauretti E, Dincer O, Praticò D. Glycogen synthase kinase-3 signaling in Alzheimer’s disease. Biochim Biophys Acta Mol Cell Res 2020;1867:118664.

85.

Leroy K, Yilmaz Z, Brion JP. Increased level of active GSK-3β in Alzheimer’s disease and accumulation in argyrophilic grains and in neurones at different stages of neurofibrillary degeneration. Neuropathol Appl Neurobiol 2007;33:43-55.

86.

Arciniegas Ruiz SM, Eldar-Finkelman H. Glycogen synthase kinase-3 inhibitors: preclinical and clinical focus on CNS-A decade onward. Front Mol Neurosci 2021;14:792364.

87.

Shu XS, Zhu H, Huang X, Yang Y, Wang D, Zhang Y, Zhang W, Ying Y. Loss of β-catenin via activated GSK3β causes diabetic retinal neurodegeneration by instigating a vicious cycle of oxidative stress-driven mitochondrial impairment. Aging 2020;12:13437-13462.

88.

Riis S, Murray JB, O’Connor R. IGF-1 signalling regulates mitochondria dynamics and turnover through a conserved GSK-3β–Nrf2–BNIP3 pathway. Cells 2020;9:147.

89.

Ngamsiri P, Watcharasit P, Satayavivad J. Glycogen synthase kinase-3 (GSK3) controls deoxyglucose-induced mitochondrial biogenesis in human neuroblastoma SH-SY5Y cells. Mitochondrion 2014;14:54-63.