Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

Free Radical Biology and Medicine 193 (2022) 134–157

Contents lists available at ScienceDirect

Free Radical Biology and Medicine


journal homepage: www.elsevier.com/locate/freeradbiomed

Altered glucose metabolism in Alzheimer’s disease: Role of mitochondrial


dysfunction and oxidative stress
Saikat Dewanjee a, Pratik Chakraborty a, Hiranmoy Bhattacharya a, Leena Chacko b,
Birbal Singh c, Anupama Chaudhary d, Kalpana Javvaji e, Saumya Ranjan Pradhan f,
Jayalakshmi Vallamkondu f, Abhijit Dey g, Rajkumar Singh Kalra h, Niraj Kumar Jha i, j, k,
Saurabh Kumar Jha i, j, k, P. Hemachandra Reddy l, m, n, o, p, **, Ramesh Kandimalla e, q, *
a
Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700 032, West Bengal, India
b
BioAnalytical Lab, Meso Scale Discovery, 1601 Research Blvd, Rockville, MD, USA
c
ICAR-Indian Veterinary Research Institute (IVRI), Regional Station, Palampur, 176061, Himachal Pradesh, India
d
Orinin-BioSystems, LE-52, Lotus Road 4, CHD City, Karnal, 132001, Haryana, India
e
CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, India
f
Department of Physics, NIT Warangal, Warangal, India
g
Department of Life Sciences, Presidency University, Kolkata, 700073, India
h
Immune Signal Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 9040495, Japan
i
Department of Biotechnology, School of Engineering & Technology (SET), Sharda University, UP, 201310, India
j
Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India
k
Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, 248007, India
l
Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
m
Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA
n
Neurology Departments School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
o
Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
p
Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA
q
Department of Biochemistry, Kakatiya Medical College, Warangal, India

A R T I C L E I N F O A B S T R A C T

Keywords: Increasing evidence suggests that abnormal cerebral glucose metabolism is largely present in Alzheimer’s disease
Alzheimer’s disease (AD) (AD). The brain utilizes glucose as its main energy source and a decline in its metabolism directly reflects on
Dementia brain function. Weighing on recent evidence, here we systematically assessed the aberrant glucose metabolism
Diabetes
associated with amyloid beta and phosphorylated tau accumulation in AD brain. Interlink between insulin
Glucose metabolism
Insulin signaling
signaling and AD highlighted the involvement of the IRS/PI3K/Akt/AMPK signaling, and GLUTs in the disease
Mitochondrial dysfunction progression. While shedding light on the mitochondrial dysfunction in the defective glucose metabolism, we
Neurodegenerative diseases further assessed functional consequences of AGEs (advanced glycation end products) accumulation, polyol
activation, and other contributing factors including terminal respiration, ROS (reactive oxygen species), mito­
chondrial permeability, PINK1/parkin defects, lysosome-mitochondrial crosstalk, and autophagy/mitophagy.
Combined with the classic plaque and tangle pathologies, glucose hypometabolism with acquired insulin resis­
tance and mitochondrial dysfunction potentiate these factors to exacerbate AD pathology. To this end, we further
reviewed AD and DM (diabetes mellitus) crosstalk in disease progression. Taken together, the present work
discusses the emerging role of altered glucose metabolism, contributing impact of insulin signaling, and mito­
chondrial dysfunction in the defective cerebral glucose utilization in AD.

Abbreviations: AGEs, advanced glycation end products; Akt, protein kinase B; AMPK, adenosine monophosphate-activated protein kinase; ApoE4, apolipoprotein
E4; APP, amyloid precursor protein; Aβ, amyloid beta; BB, blood-brain barrier; FOX, forkhead box; GLUT, glucose transporter; GSK3, glycogen synthase kinase 3; IDE,
insulin-degrading enzyme; IGF, insulin growth factor; IRS, insulin receptor substrate; mTORC2, mammalian target for rapamycin complex 2; PDK1, phosphoinositide-
dependent protein kinase 1; PGC1-α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha; PI3K, phosphatidylinositol 3-kinase; PIP2, phosphati­
dylinositol (3,4)-bisphosphate; PIP3, phosphatidylinositol (3,4,5)-trisphosphate; RAGE, receptor for advanced glycation end products; RTK, receptor tyrosine kinase.
* Corresponding author. CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, India.
** Corresponding author. Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA.
E-mail address: ramesh.kandimalla@gmail.com (R. Kandimalla).

https://doi.org/10.1016/j.freeradbiomed.2022.09.032
Received 5 August 2022; Received in revised form 16 September 2022; Accepted 29 September 2022
Available online 4 October 2022
0891-5849/© 2022 Elsevier Inc. All rights reserved.
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

1. Introduction disorders in an age-dependent manner. Altered mitochondrial dynamics,


including increased mitochondrial fragmentation and defective
The occurrence of age-related degenerative disorders has markedly biogenesis through modifications in expressions of mitochondria
increased in the last few decades. Progressive dementia and related biogenesis genes typically indicate AD. Biopsy studies have revealed
neurodegenerative diseases have become a matter of serious concern presence of more mitochondria with broken cristae in AD neurons
among geriatric populations all over the world [1]. Clinical reports of compared to age-matched controls [6]. Maintenance of the delicate
progressive neurodegeneration together with a gradual loss of cognition balance between mitochondrial fission and fusion is an important factor
and memory are all symptoms of Alzheimer’s disease (AD), which ac­ for proper bioenergetic regulations. It is important to note here that Aβ
counts for almost 80% of dementia cases worldwide [2]. Extracellular and phosphorylated TAU are critically important for mitochondrial ab­
amyloid plaques and intracellular neurofibrillary tangles are the two normalities and oxidative stress in AD. AD is often characterized by
major players behind AD onset. However, AD can best be explained as a abnormalities in glucose metabolism and respiratory enzymes. The
multifactorial disorder rather than merely a downstream consequence of primary cause of amyloid buildup, mainly Aβ1-42 in AD arises from
amyloid beta (Aβ) deposition and the formation of neurofibrillary tan­ misprocessing of amyloid precursor protein (APP), which sets up a chain
gles. The progressive decline in glucose utilization in the frontotemporal of subsequent events that results in the death of neuronal cells [7].
cortex during the early stages of dementia indicates the onset of Altered homoeostatic balance resulting from oxidant insult gives rise to
neurodegenerative diseases [3]. Since glucose is the sole source of en­ oxidative stress that plays an important role in the progress of AD, or
ergy in the brain, reduced glucose metabolism affects the brain early and other neurodegenerative complications [7]. Oxidative stress, in the long
to a high extent [4]. During AD, mitochondrial and bioenergetic alter­ run, induces cellular damage, DNA impairment, and mitochondrial
ations are remindful of those observed with advancing age but are of a dysfunction, contributing to age-related degenerative diseases [8–10]. A
greater extent [5]. The hallmark pathomechanisms of the AD brain growing body of evidence showed the relevance of dysfunctional
include senile plaques, neurofibrillary tangles, the malfunctioning glucose metabolism and subsequent oxidative stress plays a key role in
cholinergic system, impaired synaptic transmission and plasticity, AD progression [11]. Impaired glucose catabolism results in the down­
oxidative stress, and neuroinflammation [2]. regulation of ATP synthesis that is involved in this process. On the
Oxidative stress and genetic mutations contribute to mitochondrial mechanistic side, these efforts shed light on the roles of reactive oxygen
damage and dysfunction, which in turn, leads to neurodegenerative species (ROS) and reactive nitrogen species (RNS) in oxidative damage,

Fig. 1. Schematic diagram showing factors involved in the susceptibility to AD. Besides the age, family history, gender, lifestyle, environmental hazards, and
anthropological factors, glucose levels and mitochondrial utilization of glucose play important role in the progression of AD pathology. Aberrant glucose metabolism
and low glucose uptake through the BBB are the significant markers of dementia triggered at a later stages of clinical AD. Aβ, amyloid β; APP, amyloid precursor
protein; BBB, blood-brain barrier; COX, cytochrome oxidase; GLUT1, glucose transporter 1; RCC; regulator of chromosome condensation; ROS, reactive oxy­
gen species.

135
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

insulin signaling, mTOR activation, and proteostasis abnormalities in and neurotrophic and neuroendocrine functions via GSK-3β signaling
AD brains and underline the need for early detection and antioxidant [32]. Resistance to insulin and insulin-like growth factor 1 (IGF1) are
interventions in AD [11,12]. However, a synergistic understanding of key features of AD that cause a reduction in utilization/catabolism of
these processes with mitochondrial dysfunction/bioenergetic alterations glucose and generation of oxidative stress [30].
in glucose metabolism and AD - diabetes mellitus (DM) crosstalk is A high requirement of energy in the brain indicates a small energy
elusive. The present work attempts to sum up current knowledge on reserve, thereby making the brain highly prone to malfunction on a
these associations among insulin signaling, mitochondrial dysfunction, compromised supply of glucose and/or oxygen. During compromised
and subsequent improper cerebral glucose utilization with AD pathol­ conditions, neuronal firing suffers to a high extent as synaptic functions
ogies (Fig. 1). require high energy, leading to impaired impulse transduction. To sus­
tain cell viability, the brain tends to shift metabolism towards the con­
2. Cerebral glucose metabolism: normal versus AD servation of energy in the form of ATP. It is, however, beneficial in a
short period, but it may lead to a reduction in brain function and neu­
Reduced brain glucose metabolism has been proposed to be biolog­ rodegeneration in the long run. For example, acute reduction in the
ically linked to AD [13]. In healthy brains, glucose is an essential sub­ activity of α-ketoglutarate dehydrogenase complex aids to minimize
strate for neuronal cells to gain energy. Brain utilizes more than 20% of external oxidative stress, while long-term suppression of the activity of
body glucose [14]. The respiratory quotient of the brain is close to 1 the same results in the AD-like changes in Ca2+ metabolism and impairs
indicating carbohydrates as the principal substrate for aerobic meta­ the ability of neurons to sustain the impact of oxidative stress [33].
bolism. Transport of glucose across the blood-brain barrier (BBB) and Deficient glucose uptake in the brain, insulin resistance, and impaired
intracellular catabolism of glucose are two main pillars of cerebral glycosylation together contribute to compromised glucose metabolism
glucose metabolism, wherein the former requires the help of several during AD [34].
glucose transporters including glucose transporter (GLUT) isoforms Reduced metabolism of glucose in the hippocampal region gradually
[15]. Thus, insulin, insulin receptors, and insulin-sensitive GLUT iso­ spreading to the posterior cingulate cortex, temporal-parietal cortex,
forms provide a platform for insulin-stimulated glucose uptake in certain frontal cortex, and occipital cortex indicates the onset and development
brain areas including the hippocampus, a memory-supporting structure of mild cognitive impairment and reduction of synaptic density and
in the brain [16]. Of note, during the stage-wise progression of AD, activity. ROS, peroxides, and other free radicals are formed as a result of
cerebral blood flow is compromised by 20–65% and cerebral glucose the premature leak of electrons to oxygen at electron transport chain
utilization is reduced by nearly 45% [17]. Low glucose uptake through complexes I and III [35]. The imbalance between ROS formation and
the BBB is a significant marker of dementia that often triggers at a later scavenging leads to oxidative damage of intracellular lipids, proteins,
stage of AD [18]. and DNA. Subsequently, reduced glucose metabolism in synaptic regions
Cognitive decline can be directly correlated with reduced glucose and reduced production of ATP in mitochondria impairs cytochrome
metabolism [19]. Deficiency of glucose transporters i.e., GLUT1 in the oxidase (COX) activities [36].
BBB endothelial cells, astrocytes, oligodendrocytes, and neuroglia and Neuronal glucose utilization decline with age making old age the
GLUT3 in the neuronal membrane has been evinced to accelerate most common factor behind AD onset [37]. Impaired glucose uptake and
cognitive decline through neurodegeneration and Aβ deposition [15]. utilization leads to the lesser formation of acetyl-CoA that ultimately
The Michaelis-Menten constant of GLUT3 is on the lower side which hampers acetylcholine production and in turn, cholinergic signaling.
allows a steady supply of glucose to neurons even at reduced glucose Deficient acetyl-CoA also causes neuronal cell membrane abnormality
concentrations towards sustaining brain functions [20]. Microinjection owing to limited production of membrane lipid components like
of glucose into the hippocampus, striatum, amygdala, and medial cholesterol and other neurosteroids [38]. Impaired glucose metabolism
septum has been observed to improve memory performance [19]. In a leads to deficiency of UDP-GlcNAc as well. This, in turn, results in
mouse model of AD, GLUT1 deficiency has been found to accelerate Aβ downregulation of O-GlcNAcylation of microtubule-associated TAU
deposition leading to neurodegeneration [21]. In a post-mortem brain protein, consequently TAU gets hyperphosphorylated, initiating a
study, researchers found higher glucose concentration in early-onset AD cascade of events for AD onset as observed in the preclinical study [39].
brains along with reduced glycolytic flux and lesser GLUT3 levels [22]. Hyperglycemia due to hypometabolism of glucose compels
The apolipoprotein E4 (ApoE4) carriers are more prone to hypo­ auto-oxidation of glucose to form advanced glycation end products
metabolism of glucose contributing to subsequent cognitive impair­ (AGEs) that induce Aβ and TAU, promoting the formation of senile
ments [23]. Additionally, it has been discovered that suppression of plaques and neurofibrillary tangles respectively [40].
triggering receptors expressed on myeloid cells 2 (TREM2) is linked to
decreased levels of glucose transporters, glycolytic enzymes, and tran­ 2.1. Aberrant glucose metabolism: role in amyloid accumulation
scription factors that regulate the glycolytic pathway [24]. Reduced
cerebral blood flow and downregulation of glycolytic enzymes, such as Amyloidogenesis refers to a situation where soluble Aβ protein turns
pyruvate dehydrogenase, isocitrate dehydrogenase, and α-ketoglutarate into insoluble fibrillar aggregates. According to the amyloid cascade
dehydrogenase also play vital roles in AD [25]. AD patients seem to rely hypothesis, proteolytic cleavage of APP by β-secretase and γ-secretase
more on anaerobic glycolysis for the generation of energy evidenced by results in the formation of senile plaques consisting of Aβ1-40 and Aβ1-
elevated lactate and reduced pyruvate [26]. 42 [41]. The abnormal protein processing leads to the accumulation of
2-[18F]fluoro-2-deoxy-D-glucose-positron emission tomography insoluble Aβ aggregates, as a result, downstream AD-related neuropa­
(FDG-PET) analysis demonstrated that adult offspring of AD-affected thologies are triggered. Aβ1-42 is more prone to aggregation than
mothers are more likely to exhibit AD-like brain abnormalities than Aβ1-40 [42]. The β-sheet fibrillar conformation of Aβ1-42 is responsible
adult children of AD-affected fathers [27]. Since mitochondrial DNA is for the cytotoxic effect. It is to be noted that soluble Aβ oligomers act as
acquired from the mother, it established an association between cerebral primary neurotoxins impairing glucose metabolism, promoting synaptic
glucose metabolism and mitochondria. A correlation between brain dysfunction by virtue of oxidative damage to the synaptic membrane,
aerobic glycolysis and Aβ deposition has also been established [28]. cognitive decline, and affecting learning and memory processes [43].
Evidence-based estimation suggests that nearly 65% of diabetics pose Glycation, enhances the lifespan of oligomeric forms of Aβ, thereby
with increased risk of AD [29]. Within the central nervous system (CNS), facilitating faster cognitive decline, and neuronal death [44]. The
Aβ competes with insulin to interact with insulin-degrading enzyme increased presence of C99, a metabolic product of APP, in
(IDE), suggesting that both peptides play an important role to maintain mitochondria-associated endoplasmic reticulum membrane (MAM) in­
homeostasis [30,31]. Insulin is involved in maintaining neuroplasticity, duces apposition between endoplasmic reticulum (ER) and

136
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

mitochondria. It also elevates sphingolipid turnover causing aberrations suggest that the abnormal glucose metabolism in the brain triggers TAU
in membrane lipid homeostasis in the mitochondria-associated ER phosphorylation that is implicated in the pathogenesis of AD.
membrane [45].
During the late stages of AD, APP is arrested in mitochondrial protein 3. Interlink between insulin signaling and AD
transport channels [46]. The accumulation of APP across the mito­
chondrial protein transport channels subsequently impairs membrane Insulin signaling plays a major role in the proper utilization of
integration leading to an abnormal metabolism in mitochondria and glucose, thus, diabetic patients often experience memory loss and
elevation in H2O2 levels. Increased presence of H2O2 leads to a decreased cognitive impairment [62]. Cerebral glucose metabolism is a mecha­
activity of pyruvate dehydrogenase and increased lipid peroxidation nistic link between DM and AD. Type 3 diabetes or “brain diabetes”
even before Aβ deposition, suggesting peroxide-mediated mitochondrial shares the common molecular and cellular characteristics of type 1 DM,
dysfunction in AD [47]. Accumulation of Aβ via translocase of the outer type 2 DM, and insulin resistance associated with the development of
membrane (TOM) within mitochondria promotes opening of mito­ neurodegeneration [63]. Insulin resistance has been considered as one
chondrial permeability transition pores (MPTPs), which reduces activity of the key characters of both AD and type 2 DM. Brain has higher
of respiratory chain enzymes, especially complex III and complex IV. number of insulin receptors compared to the rest of the body that are
Free radicals produced by these synaptic mitochondria activate β-sec­ majorly distributed among the hypothalamus, hippocampus, olfactory
retase in late-onset AD, which results in the elevated generation of Aβ bulb, and cerebral cortex [64]. Insulin receptor substrate (IRS) plays a
[48]. Compromised production of ATP, in turn, affects neurotransmis­ major role in learning, memory functions, and synaptic plasticity.
sion that further aggravates cognitive decline. In the transgenic mouse Despite the fact that DM is a peripheral syndrome; while, AD is a CNS
models, Aβ has been observed to cause stepwise degradation of synapses disease, reduced insulin levels are seen in AD brains [65]. Under normal
aiding the progress of AD [49,50]. Injection of Aβ in the isolated synaptic physiological condition, the brain contains 10–100 times more insulin
terminal of a neuron has been found to increase ROS levels and leads to than the rest of the body, which is regulated independently [66]. Even
apoptosis in a rat model of early AD [51]. In addition to inactivating though the exact underlying mechanism is still elusive, and no conclu­
mTORC1 in lysosomes, Aβ can also interact with dynamin-related pro­ sive theory is yet established. However, studies proposed impaired in­
tein 1 (Drp1), a mitochondrial fission protein, to impair the normal sulin signaling as a risk factor in AD [67,68].
mitochondrial dynamics [50]. Insulin is transported through the BBB into the CNS by a specific
In AD, functional astrocytes generated from induced pluripotent transporter system coupled to insulin receptors, abundantly present in
stem cells (iPSCs) have been observed to increase oxidative stress in vitro brain microvessels [69]; insulin is also locally produced in the hippo­
by increasing the Aβ production [52]. Ca2+ homeostasis and Aβ accu­ campus, cerebral cortex etc. [70,71]. In the brain, insulin signaling is
mulation are mutually related risk factors behind AD. Aβ1-42 oligomers activated by binding insulin with insulin receptors (IRs). Insulin, on
increase neuronal Ca2+ levels leading to stress-dependent death of binding with α subunits of the transmembrane IR, promotes autophos­
neuronal cells [53]. A growing body of evidence shows that mitochon­ phorylation of tyrosine kinases on the β subunits of the receptor. IR
drial Na+/Ca2+ exchanger (NCLX) is impaired in AD resulting in an activation endorses phosphorylation and acetylation of IRS1 to accom­
increased accumulation of Ca2+ in mitochondria, which endorses modate phosphatidylinositol 3-kinase (PI3K). While PI3K, in turn,
memory loss and Aβ and TAU accumulation [54]. The genetic rescue of phosphorylates phosphatidylinositol (3,4)-bisphosphate (PIP2) into
neuronal NCLX restores the pathological events and cognitive decline phosphatidylinositol (3,4,5)-trisphosphate (PIP3). PIP3, in conjunction
[55]. Age-associated reduction in IDE further aids Aβ accumulation due with the phosphoinositide-dependent protein kinase (PDK) activates
to reduced degradation of amyloid aggregates. Increasing ATP produc­ protein kinase B (Akt) by phosphorylating the same. In the hippocam­
tion by anaerobic pathways is suggested to improve impulse conduction pus, Akt1 and Akt3 are scattered throughout the somatic cells, whereas
and accelerate Aβ clearance, thereby could delay the progression of AD Akt2 are mostly present in the astrocytes instead of neurons [72].
[56]. Phosphorylated Akt, in turn, is responsible for the translocation of
GLUT4 to the plasma membrane to facilitate the intracellular uptake of
2.2. Aberrant glucose metabolism: role in TAU pathology glucose. The activation of Akt also suppresses the expression of genes
responsible for phosphoenolpyruvate carboxykinase and
Hyperphosphorylation of TAU proteins produces neurofibrillary glucose-6-phosphatase aided by forkhead box protein O1 (FOXO1) [73].
tangles, which is a significant hallmark of AD pathogenesis [57]. Akt simultaneously suppresses glycogen synthase kinase 3β (GSK-3β) so
Elevated Aβ and ApoE4 levels are responsible for reduced glucose uti­ that glycogen synthase can convert transported glucose into glycogen
lization in the AD brain. Cerebral glucose metabolism is affected more without any interruption for storage [74]. Consequently, glycogenesis is
by ApoE4 than ApoE2 [58]. O-GlcNAcylation of TAU is decreased in AD, increased while glycogenolysis is downregulated [75]. It also increases
as a consequence, TAU phosphorylation is increased. Again, O-GlcNA­ cell survival due to the inhibition of the proapoptotic Bcl2-associated
cylation of APP encourages secretion of soluble amyloid precursor agonist of cell death (BAD) gene [76]. Interestingly, neoglucogenesis
protein alpha (sAPPα) that is rather beneficial for neuronal excitability also takes place within certain brain cells, such as astrocytes, though less
and synaptic plasticity [59]. TAU hyperphosphorylation promotes the efficient than in the liver [77,78]. Astrocytes are equipped to allow
self-assembly of paired filaments into tangles that are both helical and enzymatic pathways for neoglucogenesis possibly to serve as an alter­
straight in shape, disrupting the stabilized microtubules that are seen native source of energy for neurons during stressed conditions. Astro­
under normal conditions. Suppression of protein phosphatase 2A (PP2A) cytes display sufficient 6-phosphofructo-2-kinase/fructose-2,
in AD brain further supports neurofibrillary degeneration [60]. Cleavage 6-bisphosphatase activity, which is supported by expressions of the
of inhibitor 2 (I2, also known as SET) of PP2A into I2CTF and I2NTF, and glucose-6-phosphatase 3 gene in the brain to demonstrate neogluco­
subsequent translocation from nucleus to neuroplasm initiate AD genesis from multiple precursors [77,79,80].
development. Introduction of I2CTF into rat brain has resulted in inhi­ A population-based cohort study found that impaired insulin
bition of PP2A, hyperphosphorylation of TAU, neurodegeneration, and signaling causes type 2 DM, which doubles the risk of AD [81]. Both AD
cognitive decline [61]. Synaptic dysfunctions and progression of TAU and type 2 DM are amyloid-forming diseases sharing the common fea­
pathology are both facilitated by compromised glucose availability in tures of insoluble protein deposition i.e. Aβ in the brain and amylin in
the CNS. Deficient glucose metabolism demonstrated memory impair­ the pancreas respectively [82]. The metabolism of glucose in mito­
ments, long-term reduction of synaptic potentiation, and p38 chondria is the primary source of energy in brain cells, thus proper
mitogen-activated protein kinase (MAPK)-mediated increase in TAU functioning of cognition, memory, learning, neuronal development etc.
phosphorylation in tauopathy mouse model [13]. Clearly, these reports are dependent on glucose metabolism. Hypometabolism of glucose in

137
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

brain cells acquires the depletion of cognitive abilities gradually leading IGF1R improves the state of neuronal degeneration in AD, reducing
to pathological symptoms of AD [23]. It has been reported that geriatric behavioral deficits and Aβ buildup [113,114]. The impaired IRS2
patients with type 2 DM experience an elevated risk of dementia during signaling in brain has been shown to prolong the lifespan, improve
intermittent hypoglycemic states [83]. cognitive functions, and decrease Aβ accumulation in preclinical models
Insulin facilitates glucose metabolism, defective insulin signaling of AD under normoglycemic milieu [111,115,116]. The deletion or
leads to decreased glucose metabolism that causes AD symptoms to rise suppression of IGF1R does not alter Akt and GSK-3β activities in the
[36]. AD patients tend to lead a life with up to 80% decreased insulin brain of AD mice [117]. Thus, it could be presumed that reduction in
compared to normal healthy people [84]. Also, peripheral insulin IGF1R/IRS2 signaling exerts neuroprotective action in AD [116].
resistance has been a very common phenomenon in AD [68]. Insulin
resistance and impaired insulin signaling affect the processing of both 3.2. PI3K/Akt in AD
TAU and Aβ, which play important roles in the pathogenesis of AD.
Compromised glucose metabolism cause alterations in APP processing PI3K/Akt pathway regulates numerous downstream factors
and produces amyloidogenic depositions [85]. Studies have also estab­ including GSK3, mTOR complex 1 (mTORC1), and the forkhead box
lished a direct connection between Aβ metabolism and insulin signaling (FOX) transcription factors that control several essential activities in the
pathways in neuronal cells [57,86]. In a study, insulin-resistant mice brain and peripheral system [118]. Impaired insulin signaling decreases
have shown elevated Aβ levels in the brain and an increased amount of P13K activity, which thereby reduces downstream Akt activity. It
Aβ synthesizing enzymes [87]. Insulin and Aβ are both substrates for cumulatively affects neuronal survival and metabolism [119]. The
IDE, thereby allowing hyperinsulinemia to competitively inhibit the drastic decrease in PI3K/Akt activities was observed in the brain cells of
degradation of Aβ [88]. Aβ has been experimentally proven to have AD patients even in absence of diabetes [120]. In AD brain, the alter­
detrimental effects on insulin signaling by inhibiting autophosphor­ ations in the insulin/PI3K/Akt signaling seemed to be associated with
ylation of the receptors [89,90]. Impaired insulin signaling can also decreased phosphorylation or total level of these transcription factors
trigger neurodegeneration by increasing the phosphorylation of TAU [65]. A growing body of evidence has shown that Aβ oligomers inhibit
proteins [91]. Of note, insulin and IGF1 regulate TAU phosphorylation PI3K/Akt pathway and promotes neuronal death [65,121]. Postmortem
through inhibition of GSK-3β in brain cells [92]. Therefore, impairments AD brains also revealed decreased quantities of IGF1R, IGF1, IRs, insu­
of insulin signaling in the brain results in an increase in GSK-3β activity lin, p110, and p85 subunits and reduced phosphorylation of Akt [65,
by depletion of Akt that leads to hyperphosphorylation of TAU, which in 110]. These alterations are associated with several pathological impli­
turn, results in the production of neurofibrillary tangles [57]. Peripheral cations of AD including the neurofibrillary tangles, microglial and
hyperinsulinemia has been demonstrated to elevate TAU phosphoryla­ astroglial markers [119]. PI3K/Akt pathway inhibits activity of GSK-3β,
tion [93]. Thus, it is clear that insulin signaling and TAU protein and are a TAU-phosphorylating kinase by phosphorylating serine 9 residue
intricately associated, and malfunction in either leads to neuro­ [122] (Fig. 2).
degeneration in AD [94]. Disruptions of autophagy play a key role in neurodegenerative dis­
orders, which is marked by the accumulation of harmful intracellular
3.1. IRS signaling in AD protein aggregates [123]. The mTOR is a key regulator of autophagy,
which forms two complexes, mTORC1, and mTOR complex 2 (mTORC2)
Insulin upon binding with IR attracts the IRS1 protein. IRS1, being that are interestingly the downstream factors of PI3K/Akt pathway [75,
phosphorylated and acetylated by the activated IR, undergoes confor­ 124]. Phosphorylation of Akt mediated by mTORC2, in turn, triggers
mational changes to accommodate PI3K. PI3K is a key factor in several mTORC1 that inhibits autophagy and promotes neuronal protein syn­
subsequent intracellular signaling pathways. Insulin and IGF1 bind with thesis [125]. In N2a cells, Aβ expression reportedly seen to acquire in­
the IR and IGF1R, respectively to promote tyrosine kinase activities. This sulin resistance, as evidenced by the inhibition of Akt phosphorylation
induces tyrosine phosphorylation in cellular substrates including IRS1-4 and increased activity of phosphorylated adenosine
[95]. Insulin signaling regulates mitochondrial glucose metabolism by monophosphate-activated protein kinase (AMPK) i.e., negative
IRS1 and IRS2 [96]. The IRS proteins are constituted with mTORC1 regulator [126]. Interestingly in the same study, mTORC2 in
amino-terminal pleckstrin homology and a phosphotyrosine binding rat primary cortical neurons exhibited the resistance to the effects of
domain that precedes a tail of tyrosine and serine/threonine binding site intracellular amyloids.
[97]. The structural changes induced in IRS by IR/IGF1R increase the Under normal conditions, insulin promotes the PI3K/Akt/mTORC1
flexibility of IRS to bind with PI3K [98]. PIP3, a downstream substrate of pathway to suppress autophagy, but when this route is impaired, auto­
PI3K involves the serine/threonine kinase, PDK1 by moving into the cell phagy increases and the accompanying biomarkers for the development
membrane, where Akt is activated [99]. The adverse effects of impaired of AD shift significantly [119]. Chronic low-grade inflammation is a
insulin signaling in the brain are mediated by alterations of protein major pathological feature of both type 2 DM and neurodegenerative
functions in IRS by serine/threonine phosphorylation [100]. disorders including AD [127,128]. Of note, inflammatory markers, such
Insulin triggers tyrosine phosphorylation of IRS1 while serine as TNF-α are found in blood and the brain samples of AD patients [129].
phosphorylation of IRS1 is decreased. Both Aβ and insulin resistance Hyperinsulinemia can induce neuroinflammation based on increased
endorses serine phosphorylation of IRS1, implicating a mechanistic as­ levels of TNF-α, IL-1β, and IL-6 in CSF [130,131]. Proinflammatory M1
sociation of negative and positive regulatory sites of IRS1 in both AD and activation of microglia occurs when toll-like receptor (TLR) 4 is acti­
DM pathophysiology [100–103]. Other murine models of AD showed vated by ligands like lipopolysaccharides. Production and secretion of
increased serine phosphorylation of IRS1 [105–108]. A growing body of proinflammatory cytokines viz. TNF-α, IL-1β, and IL-6 are distinctly
evidence revealed that abnormal IRS1 serine phosphorylation is linked enhanced under activated TLR4 [132]. TLRs also regulate NF-κB via
to the tauopathy and TAU pathology in AD [103–109]. In the line with PI3K/Akt/mTORC1 signaling to influence transcription, cytokines pro­
the findings in preclinical AD models and postmortem analyses, human duction, and survival of immune cells [133].
brain cells also exhibited abnormal serine phosphorylation of IRS1 with
concomitant diminishing total IRS1 and IRS2 protein levels [110,111]. 3.3. GLUTs in AD
These findings indicate a strong correlation between AD and serine
phosphorylation on IRS1. However, the specific molecular mechanism Given the fact that glucose is the sole source of energy in the brain,
of the relationship between IRS1 serine phosphorylation and AD path­ the defective passage of glucose into brain cells may lead to neurode­
ophysiology remains to be unveiled [111,112]. generative pathologies [134]. Insulin stimulates insulin-dependent
Accumulating evidence reveals that reduction and/or deletion of GLUTs, GLUT4 and GLUT8 that are abundantly present in the

138
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

Fig. 2. Schematic diagram showing PI3K-Akt intracellular signaling. Insulin binding to IR causes tyrosine phosphorylation of IRS and thereby activates PI3K
(composed of regulatory p85 and catalytic p110 subunits). PI3K converts PIP2 into PIP3 that recruits Akt-kinase to cell membrane. PIP3 activates PDK1 to phos­
phorylate Thr308 of Akt1. PI3K/Akt via their downstream factor, viz. Akt substrate 160 kDa (AS160) regulates GLUT4 translocation to the cell membrane and
cellular glucose uptake into the cell, herein mTORC1 regulates several functions including protein synthesis, autophagy, and cell growth/axon growth, GSK3
signaling, glycogen synthesis, and TAU phosphorylation; while, FOX transcription factors regulate functions including cell survival. TREM2, a surface receptor,
signaling in the microglia regulates the phagocytosis, survival, and proliferation. TREM2 is usually activated by lipoproteins, phospholipids, and oligomeric Aβ that
promotes its interaction with DAP12 i.e., activating adaptor protein, which subsequently activates PI3K/Akt pathway. Akt, protein kinase B; AS160, Akt substrate of
160 kDa; DAP12, DNAX activating protein of 12 kDa; FOXO, forkhead box protein O; GLUT, glucose transporter; GRB, growth factor receptor-bound protein 2; GSK-
3β, glycogen synthase kinase 3 beta; IR, insulin receptor; IRS, insulin receptor substrate; MAPK, mitogen activated protein kinase; mTORC, mammalian target for
rapamycin complex, PDK1, phosphoinositide-dependent protein kinase 1; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; PIP2, phosphatidylinositol (3,4)-
bisphosphate; PIP3, phosphatidylinositol (3,4,5)-trisphosphate; PTEN, phosphatase and tensin homolog; RAF, rapidly accelerated fibrosarcoma; RTK, receptor
tyrosine kinase; Shc, SHC-transforming protein; SHIP, SH2-containing inositol 5′ -phosphatase; SOS, son of sevenless; SYK, spleen tyrosine kinase; TREM2, triggering
receptor expressed on myeloid cells 2.

hippocampus, amygdala, cerebral cortex, and cerebellum. Upon acti­ Accumulating evidence suggests that the involvement of GLUTs in
vation by insulin, GLUT4 translocates from cytosol to cell membrane to the pathogenesis of AD [140]. A study back in 1989 reported reduced
facilitate glucose entry into the cell, whereas GLUT8 translocates to GLUTs in the neocortex and hippocampal region of the postmortem AD
rough ER within cytosol to retrieve dispensable glucose after the protein brain [140]. In the rat model of sporadic streptozotocin-induced AD,
glycosylation. These activities are compromised by downregulated in­ decreased levels of GLUT1 were observed along with impaired insulin
sulin signaling giving rise to neurodegeneration [135]. GLUT1, an signaling, increased phosphorylation of TAU protein and neurofila­
insulin-independent GLUT is present at the BBB, epithelial cells, and ments, and decreased microtubule binding capacity of TAU protein
astrocytes. GLUT3 transports glucose into neurons, these transporters [138]. Reduced levels of GLUT1 in different parts of the brain in
are also present at low levels in endothelial and astroglial cells. different stages of AD have also been reported in preclinical studies
GLUT2 is present at low levels in astrocytes. Some insulin-sensitive [141–143]. Reduction of GLUT1 level has been found to hamper glucose
transporters e.g., GLUT4 (hypothalamic neurons, hippocampal neu­ transport leading to TAU phosphorylation, Aβ builds up, and formation
rons, cerebellar neurons, sensorimotor cortex, pituitary), GLUT8 (hy­ of neurofibrillary tangles [137,144]. Reduction of GLUT3 levels has also
pothalamic neurons, hippocampal neurons), and GLUT12 (cortical been seen in the AD brains of both humans and rodents [22,142,145,
astrocytes) are also present in the brain at low levels. Disruption in the 146]. It has also been seen that GLUT3 inhibition is connected to
proper functioning of these transporters leads to deficient glucose up­ reduced insulin signaling, elevated phosphorylation of TAU and neu­
take by the brain cells, thereby severely affecting the cerebral glucose rofilaments, and reduced TAU protein’s capacity to bind to microtubules
metabolism [136]. In AD, deficiency of transport and metabolism of [138]. Significant upregulation of GLUT4 and GLUT12 was seen in the
glucose may be a result of impaired insulin signaling that might occur AD brains of both humans and rodents [137,143,147,148]. Increased
due to disturbances in the functioning of insulin-sensitive GLUTs [134]. level of GLUT2 was observed in the brain of AD rats [139]. Studies have
In a study to understand the correlation between insulin signaling and demonstrated AD-related aggravation as a result of GLUT1 deficiency
GLUTs in AD pathology, it has been found that GLUT1 has a positive that includes a suppression of low density lipoprotein receptor-related
correlation with the insulin signaling proteins, such as IRS1, whereas protein 1 (LRP1) level in brain capillary, reduction of Aβ clearing
GLUT4 has a negative correlation with the same [137]. Insulin signaling transporter at the BBB, reduction of the BBB integrity, decrease in ce­
has been found be to positively associated with GLUT1 and GLUT3 ac­ rebral blood flow, enhancement of Aβ accumulation in hippocampus
tivities, while an inverse association is seen between insulin signaling and cerebral cortex and neuronal impairment and cognitive dysfunction
and GLUT2 activity [138,139]. [21,149].

139
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

3.4. AMPK signaling in AD Glucose metabolism is mainly dependent on mitochondria to


generate energy. During inefficient oxidation, the ratio of ATP produc­
AMPK acts as a connecting link between DM and AD. AMPK plays a tion and/or oxygen consumption becomes very low, leading to increased
key role in the etiology of AD through its influence on glucose and lipid production of superoxides [172]. Mitochondrial alterations serve as the
metabolism, and energy balance. AMPK aids in energy production primary driving force to malfunction behind insulin resistance, thus
through elevated glucose and lipid metabolism and by apprehending abnormalities in insulin signaling arising from mitochondrial malfunc­
energy-consuming procedures that includes synthesis of cholesterol and tion are often termed as ‘mitochondrial diabetes’ [173]. On the other
proteins [150]. In insulin signaling, AMPK is closely associated with hand, compromised actions of insulin can also dysregulate mitochon­
neoglucogenesis through both upstream and downstream kinases in the drial functions [174]. During insulin-resistant conditions, IRSs fail to
favor of amelioration of DM [151,152]. The effect of AMPK on insulin inhibit FOXO1, thus hyperactivated FOXO1 induces heme oxygenase 1
resistance is yet to be unanimously approved by the scientific commu­ (HMOX1) to consume ‘heme’ and disrupt the electron transport chain
nity. Though it is generally considered that AMPK reciprocates insulin leading to increased production of ROS [175]. Downregulation of
resistance by regulating GLUT4 and free fatty acids, while some re­ NEUROD6 in the AD brain further assists in this cascade [176]. AD brain
searchers have claimed the involvement of AMPK in developing insulin witnesses abnormal Kreb’s cycle that causes alterations in oxidative
resistance [153,154]. AMPK is thought to be activated by its upstream phosphorylation, resulting in glucose hypometabolism, elevated ROS
kinases through phosphorylation at Thr172 [155]. AMP: ATP ratio also generation and apoptosis. Interestingly, mitochondrion itself is highly
plays a vital role in the activation of AMPK-associated metabolic path­ susceptible to oxidative stress which allows room to initiate a vicious
ways. Proteomic analyses have discovered that many of the proteins cycle [177]. Deposition of amyloids later follows with other manifes­
dysregulated during AD are closely associated with AMPK signaling tations of clinical AD.
[156,157]. AMPK acts as a metabolic sensor that restores normal brain
energy metabolism and reduces Aβ deposition by modulating β-secretase 4.1. AGEs accumulation and activation of the polyol
[158,159]. It also affects hyperphosphorylation of TAU negatively by
acting upon sirtuin 1 (SIRT1) and PP2A [153]. In addition, AMPK aids in AGEs are a group of molecules produced by non-enzymatic in­
the alleviation of AD by inducing autophagy to resolve amyloid depo­ teractions between AGEs are a group of molecules produced by non-
sition [160]. enzymatic interactions with carbonyl compounds (glyoxal, methyl­
In contrast, a non-human primate (female vervet monkeys) model of glyoxal, glyceraldehyde, 3-deoxyglucosone, sugars, etc.) and proteins,
AD showed enhanced AMPKα2 activity in the AD hippocampus, while phospholipoprotein, or nucleic acids [178]. The polyol pathway con­
the activity α1 subunit remained unaltered [157]. However, in a mouse verts glucose to fructose and promotes glycation; fructose is then
model of AD, it was observed that genetic reduction of AMPKα1 rescues transformed to 3-deoxyglucose and fructose-3-phosphate, the two very
cognitive decline better than AMPKα2 [156]. Surprisingly, a similar powerful non-enzymatic glycation agents [179,180]. Both exogenous
APP/presenilin 1 (PS1) mouse model demonstrated metformin to (diet-derived) and endogenous (polyol-pathway-derived) fructose have
improve cognitive deficits, presumably through AMPK activation [161]. been identified as a novel pathogenic factor for a variety of metabolic,
Sphingosine-1-phosphate and miR (micro-RNA)-590-3 have been re­ inflammatory, neurodegenerative diseases [181,182]. Hexokinase 1
ported to boost neuronal apoptosis in AD through AMPK signaling accumulates in the cytoplasm of murine brain when protein deglycase
[162]. The contraindicative observations apprehend a definitive (DJ-1) is silenced rather than its usual position in mitochondria [183].
conclusion on the relationship between AMPK and AD. Activation of Among the various types of AGEs, acetaldehyde-derived AGEs
AMPK has been evidenced to delay the aging processes up to a certain (AA-AGEs) are neurotoxic [184]. In cortical neuronal cells,
limit and thus it acquires potentially beneficial effects against glyceraldehyde-derived AGE (AGE-2) has powerful neurotoxic effects
age-related neurodegenerative disorders [163]. On the contrary, with nephropathy undergo hemodialysis were entirely decreased by the
the continuous increase in neuronal stress, AMPK works to trigger and addition of an anti-AGE-2 antibody, but not by antibodies against other
augment age-related neuropathologies [150]. Due to diminished mito­ forms of AGEs, indicating that AGE-2 could be implicated in neuro­
chondrial biogenesis and functioning, lower AMPK activity compro­ degeneration [185]. The presence of AGE-2 antigen has been found to be
mises energy balance in the AD brain. Additionally, due to its effect on linked with AD as demonstrated by histological and serum analyses
APP distribution, cholesterol levels, and sphingomyelin levels, AMPK [185]. AGE-2 is found largely in the neuroplasm of the hippocampus and
can control the production of Aβ [164]. Further, AMPK decreases mTOR parahippocampal gyrus neurons, but not in the senile plaques found in
activity to aid in the lysosomal destruction of Aβ by promoting auto­ the brains of AD patients [186]. It has been suggested that AA-AGEs may
phagy [165]. Again, AMPK can act as a physiological TAU kinase to play a role in neurodegeneration [187]. When AGEs interact with AGE
phosphorylate TAU at Ser262, Thr231, and Ser396/404 [166]. How­ receptors, it results in neurodegeneration, which is linked to the etiology
ever, to conclude the precise mechanism of AMPK in AD pathophysi­ of AD [40]. Increased expression of cathepsin B and asparagine endo­
ology, more research is needed. peptidase (AEP) in the AGE-RAGE axis altered proteome, resulted in an
increase in Aβ production and activation of TAU phosphorylation. These
4. Defective glucose metabolism mediated by mitochondrial consequences were seen in AD brain, which coincided with increased
dysfunction in AD levels of AGEs, establishing a new mechanistic link between AGEs and
AD pathogenesis [40]. Through the generation of AGEs, non-enzymatic
Neuronal abnormalities mediated by mitochondria are far more glycation of myelin, or oxidative damage, the role of polyol pathway in
significant in AD pathogenesis than neuroinflammation-induced mito­ brain abnormalities and cognitive impairment has been hypothesized
chondrial dysfunction. Defective glucose metabolism and impaired in­ [188,189]. Under hyperglycemic circumstances, the polyol pathway
sulin signaling in the brain cause mitochondrial dysfunction [167,168] turns glucose into sorbitol by aldose reductase (AR), and subsequently
(Fig. 3). Mitochondrial dysfunction coupled with compromised oxida­ sorbitol to fructose via sorbitol dehydrogenase (SDH). Increments in
tive activities in mitochondria results in an elevation of ROS accumu­ intracellular glucose levels in the hyperglycemic milieu have been
lation and enhancement of lipid and protein oxidation in the in the demonstrated to activate the polyol pathway that produces fructose in
nucleus accumbens and striatum [169]. FDG-PET studies have estab­ insulin-independent organs including the brain [190]. Polyol activation
lished reduced uptake of glucose into the brain, especially the and concomitant consumption of cofactors NADPH and NAD+ set off a
temporal-parietal cortex region that shows early symptom of AD prior to chain reaction of metabolic abnormalities. AR, nitric oxide synthase
the appearance of other pathologies including cognitive dysfunction (NOS), and glutathione reductase (GR) all use NADPH as a cofactor and
[170,171]. its depletion inhibits the other enzymes [191]. Reduced NOS activity

140
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

Fig. 3. Schematic diagram showing factors involved in the impaired glucose metabolism in the AD pathology. The most prevalent causes of AD are reduced
glucose uptake and utilization, ApoE4 and PS1 mutations, which are characterized by defective glucose utilization in the brains of AD patients. GLUT1 and GLUT3 are
the key glucose transporters that regulate the BBB function and mitochondrial processes including respiration and the Krebs cycle. Alterations in glucose levels
perturb AMPK that subsequently affects Akt-mediated insulin signaling. Mitochondrial dysfunction inhibits PGC-1α that diminishes α-secretase through FOXO3α to
exacerbate Aβ and AD pathology; whereas, AGE interaction with its receptor RAGE promotes Aβ translocation and Tau hyperphosporylation via Akt inhibition, and
activation of GSK-3β. Next, impaired insulin signaling lowers PI3K activity, resulting in reduced Akt activity and increased GSK-β activity that phosphorylates Tau,
promoting Aβ development and deposition. IGF induces insulin resistance and thereby affects Aβ levels and degradation by reducing the levels of IDE. In the
glycosylation defects, O- GlcNAcelytaion and phosphorylation inversely affect the modification of TAU protein, where N-glycosylation stabilizes and modifies TAU
structure. AGEs, advances glycation end products; Akt, protein kinase B; AMPK, adenosine monophosphate-activated protein kinase; ApoE4, apolipoprotein E4
genotype; Aβ, amyloid beta; BBB, blood-brain barrier; GLUT, glucose transporter; GSK-3β, glycogen synthase kinase 3 beta; IDE, insulin-degrading enzyme; IGF,
insulin growth factor; N-GlcNAc, N-acetylglucosamine; PGC1-α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha; PI3K, phosphatidylinositol 3-
kinase; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; PS1, presenilin 1; RAGE, receptor for advanced glycation end products.

and NO content can lead to decreased neuronal blood flow, whereas GR with a lower density of neurofibrillary tangles and less neuronal loss
inhibition causes a reduction in reduced glutathione (GSH), which leads showed normal SOD activity in contrast to the severely afflicted regions,
to an increase in free radical generation and oxidative stress [192]. indicating that SOD may be able to inhibit TAU aggregation and
NAD+ is transformed to the reduced form NADH during the second step neurodegenerative processes [198]. Multiple subunits of different
of the SDH-catalyzed process. Because NADH is a substrate for NADH complexes of electron transport chain have been recognized in the
oxidase, NADH transformation causes an increase in NADH oxidase extracellular vesicles recovered from the medium of cultured mouse
activity and the creation of superoxide anions, which further contribute hippocampus neurons [199]. A growing body of evidence has proposed
to oxidative stress [193]. structural and functional mitochondrial abnormalities in AD brain cells
[194,199,200]. The patterns of low levels of complexes I, III, IV, and
ATP synthase were also previously recorded in a proteomic investigation
4.2. Defective glucose metabolism in the brain on mitochondrial electron of frozen brain tissue samples of AD patients, but the lower level of
transport chain and ROS generation intracellular vesicles was only observed in later stages [201]. Partial
rather than total reduction of oxygen leads to ROS generation, which is
Generation of energy in mitochondria is accomplished through favoured by an imbalance between greater complex IV levels and lower
oxidative phosphorylation in conjunction with electron transfer across complex V levels [194]. A study back in 2001 found that neurons with
the mitochondrial electron transport chain (terminal respiration) that greater oxidative damage in AD exhibit a remarkable increase in COX
includes complexes I through IV and ATP synthase. In the terminal activities [202]. Moreover, the reduced synaptic connection has been
respiration, final recipient of electrons is oxygen; however, partial correlated to the mitochondrial level of complex IV, which is increas­
reduction of oxygen may also occur, resulting in potentially cytotoxic ingly being highlighted as a key aspect of AD pathogenesis [203,204].
ROS [194]. Because of the high oxygen consumption and low quantities ROS interacts with aggregating proteins involved in AD and other
of endogenous antioxidants, the brain is notably vulnerable to oxidative neurodegenerative disorders [12,205]. Neurotoxic Aβ peptides reduce
damage [195]. Low level of superoxide dismutase (SOD), a mitochon­ the levels and activity of complexes I, II, and IV, while it increases ROS
drial enzyme that neutralizes superoxide radical, is involved in the formation that promotes the production of Aβ peptides [200,206]. Aβ
pathogenesis of AD by developing oxidative stress in the brain [196]. In co-localizes with the ATP synthase in the hippocampus and cortex of the
AD patients, a reduction in SOD-positive neurons in the hippocampus AD brain. This association has also been seen in the plasma membrane
and frontotemporal cortex has been observed [197]. The brain regions

141
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

that results in lower extracellular ATP levels leading to decreased rodent model of AD, given the inhibitory effect of CypD depletion and
long-term potentiation (LTP) and increased long-term depression (LTD) oligomycin-sensitivity-conferring protein (OSCP) overexpression on
at synapses [207]. Moreover, limiting oxidative metabolism enhances MPTP opening in Aβ-rich settings [228,229]. Indeed, in brain mito­
the amyloidogenic breakdown of APP [194]. Aβ and ROS also syner­ chondria from animal models with AD-like brain amyloidosis as well as
gistically damage synapses, activate microglia and neuroinflammation, in Aβ-insulted cells, enhanced sensitivity of MPTP opening is noticeable
and impair cerebral microvasculature and the BBB functioning [208, [230,231]. Excess parenchymal Aβ build-up results from disruptions in
209]. Cytoplasmic SOD1 binds to intracellular Aβ in the brain cells, Aβ metabolism, including increased synthesis and impaired breakdown
generating perinuclear complexes that decrease SOD1 activity [210]. [232]. The interaction between Aβ and CypD activates MPTP in the
SOD1 deficiency promotes Aβ oligomerization and cognitive/behavioral brain mitochondria, resulting in mitochondrial malfunction and neural
impairments; while, SOD1 treatment slows down AD progression and stress [228,233]. F1F0 ATP synthase uncoupling and MPTP opening are
improves cognition [211]. mediated by the development of an Aβ/oligomycin sensitivity conferral
protein (OSCP) complex, as observed in the AD brains, hence OSCP is
4.3. Defective glucose metabolism increases mitochondrial permeability in also a binding partner of Aβ [229,234]. Surprisingly, Aβ strengthens
the brain cells CypD and OSCP-related MPTP formation in Aβ-rich environments by
enhancing CypD regulation of OSCP by boosting their interaction [226,
Enhanced mitochondrial permeability precedes apoptosis. During 229]. A specific role for Aβ in promoting MPTP formation is shown by
oxidative stress, ROS alters mitochondrial membrane potential (MMP) the interaction of Aβ with MPTP-forming/regulating proteins, as well as
leading to its depolarization [212]. Resultant pores allow entry of BAD by the effects of Aβ on neuronal Ca2+ dysregulation and oxidative stress
and cause cytochrome c release to the cytosol. Defective glucose meta­ in AD-related pathological conditions [235]. Apart from Aβ, hyper­
bolism in combination with greater glucose accumulation leads to phosphorylated TAU aggregation is another hallmark of AD brains
increased ROS generation and activation of NOX and MAPK pathways, [236]. TAU also influences MPTP through promoting mitochondrial
which eventually increase mitochondrial permeability [213]. Mito­ dysfunction and enhancing mitochondrial ROS production [237]. In
chondrial entry of BAD dislocates B-cell lymphoma 2 (Bcl-2) and en­ human brain tissues, neurotoxic amino-terminal (NH2)-derived TAU
courages the release of cytochrome C, which together induce apoptosis interacts with ANT and the ANT/CypD complex, promoting the
[214]. Additionally, hyperglycemia directly causes mitochondrial Aβ/ANT/CypD supercomplex [238]. Importantly, TAU silencing reduces
fission and fragmentation, which increases the production of ROS and MPTP opening and suppresses CypD expression [239]. These findings
triggers the programmed cell death process. establish a link between TAU and MPTP in AD.
Excess of Ca2+ along with other cellular sensors, such as oxidative
stress, depletion of adenine nucleotides, and excess of inorganic phos­ 4.4. Defective glucose metabolism in the brain cause defect in PINK1/
phate level could endorse the mitochondrial permeability transition parkin
pore (MPTP) to open [215]. MPTP is a non-selective channel that im­
proves the inner mitochondrial membrane permeability to low molec­ Phosphatase and tensin homolog (PTEN)-induced kinase 1 (PINK1)
ular weight solutes and water. Irreversible MPTP activation, owing to its functions as a metabolic sensor coupling biogenetics of cells and stress
non-selective nature, results in severe mitochondrial dysfunction [216]. signals to mitochondrial dynamics. Level of PINK1 in healthy mito­
The prolonged opening of MPTPs results in mitochondrial depolariza­ chondria is very low due to proteases and ubiquitin ligases. Under stress
tion, oxidative phosphorylation uncoupling, and excessive formation of conditions e.g. elevated ROS levels, depolarized mitochondrial mem­
ROS [217]. In addition, unregulated entry of tiny osmolytes and water brane, increase in misfolded proteins, etc. PINK1 accumulates in the
into mitochondria via MPTPs results in mitochondrial swelling, which outer mitochondrial membrane [240]. Parkin is selectively recruited
disrupts the outer mitochondrial membrane [216]. Increased mito­ into dysfunctional mitochondria with low MMP to execute mitophagy.
chondrial permeability also promotes cell death pathways by depleting PINK1-dependent mitochondrial recruitment of parkin in response to
energy currency of cells and allowing cytochrome C to leak from the mitochondrial damage requires active glucose metabolism [241]. De­
inter-membrane space [218,219]. It has been found that abnormal fects in parkin recruitment can be correlated with suppression of PINK1
glucose phosphorylation products and ROS are responsible for mito­ upregulation in response to mitochondrial depolarization. Parkin
chondrial dysfunction in AD. ROS is also a key regulator of MPTP. One mitochondrial translocation and maintaining high PINK1 levels during
possible mechanism for ROS-mediated MPTP formation is the estab­ mitophagy are crucially regulated by ATP. Low levels of ATP arising
lishment of aqueous channels by oxidatively altered transmembrane mainly from improper utilization of glucose, appear to prevent PINK1
proteins with the aid of cyclophilin D (CypD) to promote mitochondrial translation [242]. Moreover, respiratory chain defects lead to loss of
membrane permeabilization [220,221]. PINK1 causing a decrease in MMP and enhanced sensitivity to oxidative
ROS-induced ROS release is an intriguing notion in the context of stress [243].
ROS-mediated opening of MPTPs on reaching ‘the mitochondrial PINK1 and parkin play their roles in Ca2+ homeostasis [244]. Cyto­
permeability transition (MPT)-ROS threshold. MPTPs trigger the release solic PINK1 initiates transduction cascades that result in the suppression
of mitochondrial ROS into the cytosol, where they subsequently interact of autophagy/mitophagy and the promotion of neuronal branching by
with the adjacent mitochondria [222]. The mechanism of MPT signal activating the mTORC2/Akt and protein kinase A signaling pathways.
amplification still exists even after the ROS-inducing cell stress are Mitochondrial respiration defects in the cerebral cortex and aconitase
removed, leading to an MPT propagation across all mitochondria [223]. inhibition in the striatum have been found to be associated with PINK1
Through this mechanism, MPTP propagation also causes mitochondrial silencing [245]. Interference with PINK1 has been found to increase
Ca2+ dysregulation, and Ca2+ leakage from damaged mitochondria in­ sensitivity to oxidative stress owing to mitochondrial dysregulations.
creases the role of ROS in driving MPTP activation in nearby mito­ PINK1 is also involved in the impairment of the electron transport chain
chondria. It results in severe mitochondrial stress and, in the worst leading to reduced oxygen consumption. At a metabolic level, loss of
scenario, the induction of an intrinsic apoptotic pathway [224]. PINK1 significantly inhibits glucose uptake by pancreatic primary islets
CypD expression always remains higher in the AD brains of both including β-cells that is accompanied by increased insulin secretion and
humans and animals [225,226]. Though the role of adenine nucleotide enhanced glucose tolerance [246].
translocase (ANT) in the formation of MPTP is still debated, mass Drop in MMP initiates the translocation of parkin. On reaching to
spectrometry examination of brain samples from patients with mild damaged mitochondria, parkin ubiquitinates membrane fusion protein,
cognitive impairment and AD revealed overexpression of ANT [227]. mitofusin-2 leading to proteasomal degradation of the same to prevent
These findings imply a propensity for MPTP activation as seen in the the fusion of damaged mitochondria with the healthy mitochondria

142
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

[247]. Depolarized mitochondria cannot fuse, thus excessive fission extracellular Aβ [266–268]. Knock-in of a point mutation F121A of
leads to small fragments that easily fall prey to autophagosomes and/or beclin-1 endorses autophagy by preventing the association of beclin-1
lysosomes. Mitochondrial degradation follows, aided by p62 protein and with its inhibitor Bcl-2, which endorses amyloid oligomer sequestra­
protein 1 light chain 3 (LC3) signaling. MUL1 also acts simultaneously to tion resulting in reciprocation of cognitive impairment [269]. In a pre­
complement the removal of dead mitochondria along with parkin/­ ceding experiment, beclin-1 silencing has been found to endorse the
PINK1. Accumulating evidence demonstrate the protective role of accumulation of both intraneuronal and extracellular Aβ aggregation
PINK1 against neurodegeneration mediated by oxidative stress, pro­ [270]. Autophagy appears to influence Aβ clearance at various stages.
teasomal activities, ER stress, and mitochondrial dysfunction. Interest­ Cathepsin B is a lysosomal protease that is essential for breakdown of
ingly, translocation of parkin from cytosol causes decreased proteasomal autophagic substrates. Ablation of cathepsin B gene has been found to
A
activities in cytosol leading to accumulation of TAU, Aβ, etc. PINK1 exacerbate AD pathology and increases β42 accumulation [271]. These
plays a crucial role in balancing mitochondrial fusion and fission. observations suggest that autophagic flux could be beneficial for miti­
Overexpression of PINK1 promotes mitochondrial fission, while its in­ gating AD progression.
hibition is found to trigger mitochondrial fusion [248]. Under normal Selective autophagy, in addition to bulk autophagy, has been
conditions, a low level of PINK1 is maintained, however, stresses like discovered to play a role in Aβ metabolism. Normally, Aβ binds to
elevated ROS level and depolarized mitochondrial membrane tends to ubiquitin to create a complex that prevents formation of insoluble fibrils
cumulate PINK1 at the outer mitochondrial membrane. In contrast, a [272]. Apart from Aβ metabolism, autophagy plays an important func­
study proposed that the recruitment of PINK1 by hexokinase 1 and 2 tion in TAU homeostasis. Autophagy is likely to affect tauopathies as
under stressed conditions stabilizes the outer mitochondrial membrane, well. Interruption of autophagic flow has been shown to impair TAU
while depolarizing the inner one [249]. Mitochondria-associated ER clearance and endorse insoluble TAU clump accumulation [273]. In the
membrane is upregulated in AD. Interestingly, they are considered to brains of familial AD patients, hyperphosphorylated TAU has been found
initiate autophagy as the isolation membrane forms at this site [250]. to be colocalized with the autophagosome marker LC3 and the auto­
This site also produces necessary proteins for the formation of Aβ from phagy receptor p62/SQSTM1 (sequestosome 1), but not in healthy
APP. Parkin and parkin interacting substrates suppress the biogenesis of controls [274]. In a cell line-based study, immunoreactivity to LC3 and
mitochondria through suppressing gene expression within the cell nuclei p62/SQSTM1 was consistently linked to TAU aggregates [275]. Indeed,
[251]. Mitochondria-specific autophagy is closely linked to glucose TAU phosphorylation was found to be significantly increased in condi­
metabolism through AMPK and mTOR signaling [252]. Under hypoxic tional ATG7 knockout mice, presumably due to the build-up of GSK3 i.e.,
conditions, FUN14 domain-containing 1 interacts with micro­ one of the major TAU kinases in the brains of ATG7 knockout mice
tubule-associated LC3 via specific motifs present on it to form auto­ [276].
phagosomes to incite mitophagic activity. Aβ clearance is also modulated by autophagy. The mTOR inhibitor
rapamycin drastically reduced intracellular and extracellular amyloid
4.5. Defective glucose metabolism in brain lysosome-mitochondrial buildup in the brains of AD mice, which resulted in a notable
crosstalk and autophagy improvement in cognition by restoring autophagy [277,278]. Auto­
phagy induction also ameliorates tauopathies, in line with its role in Aβ
Lysosome-mitochondria crosstalk is impaired in neurodegenerative metabolism [279]. Rapamycin treatment to TAU-mutant mice exhibited
disorders. Mitochondria are associated with the production of energy, considerably less extent of TAU phosphorylation [277,278]. Tuberous
while lysosomes process cellular wastes. Autophagy is a catabolic pro­ sclerosis complex 2 (TSC2) is a negative regulator of mTOR, thus TSC2
cess mediated by autophagosomes, whereby non-functional components silencing significantly activate mTOR signaling, resulting in a higher
are transferred to lysosomes for digestion/destruction. Stress and hyp­ accumulation of phosphorylated TAU in the brain of experimental ani­
oxia are among the chief activators of autophagy which is controlled by mals [280]. In addition to the aforementioned findings, other research
a number of signaling pathways, including mTOR, FOXO, and insulin showed that autophagy activation could aid in the prevention of tauo­
signaling [75,253]. Lysosome autophagy dysfunctions affect the proper pathies in AD [281–283]. The autophagy receptor NDP52 (nuclear dot
functioning of other organelles including the mitochondria, leading to protein 52 kDa), in addition to p62/SQSTM1, has been preclinically
an excess of ROS resembling pathological features associated with aging, found to recognize phosphorylated TAU in the AD brains [284].
chronic inflammation, and neurological diseases [254]. Immature Increased production of NDP52 by its upstream transcription factor
autophagosome accumulation and dystrophic neurites have been found nuclear factor erythroid 2-related factor 2 (Nrf2) endorses the break­
in the brains of AD patients [255]. Autophagosomes are predominantly down of phosphorylated TAU [285]. In addition to mTOR and auto­
accumulated in the dendrites rather than in the soma [256,257]. Amy­ phagy receptors, transcription factor EB (TFEB), one of the key
loid pathology of AD is preceded with autophagosome accumulation; autophagy regulators is thought to play a role in TAU-mediated disor­
however, their clearance is compromised by downregulation of various ders. TFEB upregulation has been shown to reduce soluble phosphory­
autophagy-related proteins as observed in the brains of AD mice [258, lated TAU and insoluble TAU clumps and improved cognitive
259]. These findings suggest that defective or compromised autophagy functioning [286]. According to a recent study, TFEB regulates TAU
contributes in AD pathogenesis. exocytosis in lysosomes, which promotes TAU clearance [287]. In TAU
Autophagy influences Aβ metabolism by regulating the production P301S transgenic PS19 mice, conditional silencing of TFEB resulted in
and clearance of Aβ [260]. Aβ is formed by the cleavage of the precursor lower TAU quantity in the interstitial fluid but higher intraneuronal TAU
protein APP by the enzymes β-secretase and γ-secretase [261]. Auto­ phosphorylation, and increased TAU spreading [288].
phagy related 7 protein (ATG7) functions as an E1-like enzyme in C-terminus of TAU has two motifs that can be recognized as sub­
autophagosome formation for ubiquitin-like conjugation. Extracellular strates for chaperon-mediated autophagy (CMA). TAU mutants, on the
Aβ synthesis is significantly reduced by ATG7 silencing and autophagy other hand, are still targeted to lysosomes via lysosome-associated
deficiency [262]. Additionally, Aβ secretion is hindered by autophagy membrane protein 2 (LAMP2A) oligomerization. However, TAU mu­
disruption [263,264]. A recent clinical study found that human subjects tants cannot reach all the way into the lysosomal lumen, instead mutant
with deleterious ATG7 variations exhibit a variety of neuro­ TAU proteins can enter the lysosomal lumen partially [289]. The lumen
developmental problems [265]. Surprisingly, two variant carriers under sections are then destroyed and the smaller fragments on the lysosome
the age of 30 years were found to have poor learning abilities, suggesting membrane oligomerize and impede CMA [289,290]. TAU acetylation is
a potential correlation between dysfunctional autophagy and impaired resistant to CMA-mediated degradation and tauopathies are exacerbated
cognition [265]. when CMA is compromised [291]. The same research group experi­
Microglial endocytosis is thought to be involved in the clearance of mentally revealed that loss of CMA accelerates TAU aggregation and

143
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

increases disease development, whereas increasing CMA activity by glucose in the brain, like other regions is largely mediated by
drugs dramatically mitigates AD pathology [292]. In the etiology of AD, insulin-dependent GLUTs, which makes insulin an important player in
the accumulation of Aβ produces an abundance of ROS and damages brain energy metabolism. In the hypothalamus, DM-associated insulin
mitochondria; wherein damaged mitochondria are usually eliminated resistance triggers autophagy to ultimately boost the progression of
by mitophagy [293–295]. When the MMP collapses due to too much neurodegenerative disorders. Amyloidogenesis, impaired brain insulin
ROS, mitophagy is activated in mammals. The PINK1-Parkin route is a signaling, abnormal brain glucose metabolism, mitochondrial dysfunc­
well-studied MMP-dependent mitophagy pathway. In normal func­ tion, and oxidative stress are common denominators in both AD and DM
tioning mitochondria, PINK1 is translocated into the mitochondrial that potentiate brain dysfunction [82].
matrix. However, in the injured mitochondria, the weakened MMP Reduced levels of Nrf2 and compromised synaptic integrity are
prevents PINK1 from being imported and keeps it on the outer mito­ common factors in cases of DM and AD. Impaired mitochondrial respi­
chondrial membrane, where it is autophosphorylated and activated ration and altered MMP have been revealed to be characteristic etiol­
[296,297]. Parkin and ubiquitin are phosphorylated and activated by ogies of AD in type 2 DM [311]. High cerebral superoxide levels
activated PINK1 [298,299]. Parkin detects the phospho-ubiquitin on contribute to the initiation of AD [312]. Overexpression of ApoE4 has
outer mitochondrial membrane as an E3 ligase, causing increased parkin been demonstrated to hinder insulin-stimulated mitochondrial respira­
recruitment [300]. Typically, autophagy receptors p62/SQSTM1 and tion by interacting with IRs in an age-dependent manner [313]. The
optineurin identify ubiquitinated mitochondria, which subsequently resultant decrease in the ATP generation and increment in protein
initiate autophagic destruction [301]. Parkin is proven to be unessential, phosphorylation leads to the exacerbation of pre-existing AD conditions.
as phosphorylated ubiquitin produced by PINK1 is sufficient for auto­ Mitochondrial dysfunction aided by insulin resistance generates high
phagy receptor NDP52 and optineurin recognition [302]. PTEN-L, a new oxidative stress leading to improper glucose metabolism, and the for­
isoform of PTEN, was identified as a critical negative regulator of mation of amyloids in the CNS. On the other hand, generated Aβ and
PINK1/parkin-mediated mitophagy [303]. PTEN-L is discovered to oxidative stress further exacerbate mitochondrial dysfunction,
inhibit mitophagy by dephosphorylating parkin and ubiquitin through hampering glucose homeostasis to a greater extent. Insulin activates
its intrinsic phosphatase activity [304]. Multiple different kinds of PI3K/Akt pathway to restore insulin-signaling and promotes mito­
mitophagy have been reported in addition to PINK1-parkin mediated chondrial biogenesis to improve cognition in DM-associated neuronal
mitophagy, demonstrating the complexity of mitochondrial homeostasis disorders [314]. Interestingly, since insulin resistance is a common
maintenance. Consequently, mitochondrial dysfunction is identified as a causative factor in both AD and type 2 DM, the antidiabetic drugs have
common sign of AD [295,301,305]. shown positive effects in the treatment of AD [315].
In post-mortem study of AD patients showed suppression of hippo­ Abnormal cerebral glucose metabolism and mitochondrial dysfunc­
campal mitophagy [34,306]. Since mitochondrial failure is an early tion can be blamed upon decreased activities of mitochondrial thiamine-
pathological aspect of AD and entorrhinal cortex is the first region to be dependent enzymes e.g. pyruvate dehydrogenase complex and α-keto­
impacted by AD, it has been proposed that dysfunctional mitophagy in glutarate dehydrogenase complex modulating crucial aspects of brain
the entorhinal cortex could be an early hallmark of AD pathology [306]. glucose metabolism that are critical to the normal functioning of the
Similar phenotypes were found in preclinical model of AD and neurons brain [250]. A study reported depletion of pyruvate dehydrogenase
produced from induced pluripotent stem cells of AD patients, implying complex, α-ketoglutarate dehydrogenase complex and isocitrate dehy­
that mitophagy activation could help to slow the progression of AD drogenase, and increased activities of succinate dehydrogenase and
[209,307]. In this line, pharmaceutical mitophagy agonists have been malate dehydrogenase during AD, while other enzymes of Kreb’s cycle
proven to be useful to treat AD [307]. The findings showed that remained unaltered [316]. Lessened or incomplete aerobic metabolism
increased mitophagy improved the cognitive capabilities of AD by of glucose might lead to oxidative stress and consequent mitochondrial
alleviating both Aβ and TAU abnormalities. Thus, mitophagy-mediated dysfunction in the brain neurons [317].
clearance of defective mitochondria would have therapeutic potential
for AD intervention. 6. Mitochondrial involvements and oxidative stress in AD

5. AD and DM crosstalk Mitochondrial dysfunction is deeply connected to the generation of


ROS and mitochondria-driven apoptosis, which appear to be aggravated
AD and type 2 DM both share the common pathology of insoluble in the brain of AD patients [318]. ROS concentration beyond the
protein depositions i.e. Aβ in the brain and amylin in the pancreas clearance capacity of cells leads to oxidative stress, mitochondrial
respectively [308]. Combined with the classic plaque and tangle pa­ dysfunction, cellular damage, and cell death. Thus, it suggests oxidative
thology, glucose hypometabolism and associated pathophysiological stress as a possible common mechanism underlying age-related diseases
alterations initiate a chain of factors contributing to the exacerbation of like AD. Since neurons contain a lot of polyunsaturated fatty acids and
AD. Progressive decline of cerebral glucose metabolism rate precedes AD have a high oxygen need for energy, they are often quite vulnerable to
onset [309]. The correlation between reduced glucose utilization and oxidative stress. In addition, neurons also have a weak antioxidant de­
the pathogenesis of AD is known to the scientific community since the fense. Thus, neurons are critically sensitive to mitochondrial conditions
1980s, associating the reduced rate of cerebral glucose metabolism with and alterations that underlines role of mitochondria as the master
worsened disease pathology [310]. DM, mainly type 2 DM enhances the coordinator of energy metabolism and a major producers of ROS [319].
risk of AD significantly, wherein oxidative stress plays a key role [309]. Situation becomes further exacerbated as mitochondria themselves are
In the brain, the active involvement of oxidative stress in insulin resis­ highly susceptible to ROS [224]. Oxidative stress upregulates β-secretase
tance and AMPK dysfunctions was highlighted earlier to acquire neu­ expression through c-jun N-terminal kinase (JNK) and p38 MAPK
rodegeneration by promoting neurotoxicity [164]. A detrimental effect signaling to promote TAU phosphorylation via GSK-3β [320]. Mito­
of that on the proteins regulating insulin signaling leading to metabolic chondria are intricately involved in the regulation of redox signaling,
dysfunctions by AMPK dysregulation was suggested to be critically Ca2+ homeostasis, and synaptic plasticity thereby purposefully influ­
involved in AD-associated metabolic pathways [164]. Progressive loss of encing cell survival [321]. Activated MAPKs critically contribute to
glucose metabolism in the frontotemporal dementia Fpi3indicates the synaptic deficits in AD cybrid cells via further augmentation of mito­
onset of neurodegenerative diseases. Thus, glucose metabolism largely chondrial oxidative stress and consequent mitochondrial dysfunction
serves a mechanistic link between DM and AD. AMPK connects DM and [322]. Oxidative stress tends to shift the balance of apoptotic regulations
AD by playing a key role in the development of AD through modulating towards neuronal death and thus promoting neurodegeneration [323].
glucose and lipid metabolism, and energy balance. Intracellular entry of Impairments of mitochondrial gene expressions include genes

144
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

responsible for complex IV of the electron transport chain, p53 protein, spreading throughout the brain, neurofibrillary tangles impair memory,
and NADPH oxidase that regulates the severity of AD [324]. Mutations judgement, and speech [345]. Aβ aggregations originating from APP in
in the presenilin 1 cause an imbalance in Ca2+ metabolism in ER leading extraneuronal space trigger the immune system to initiate neuro­
to mitochondrial dysfunction and implicated in familial AD. Crosstalk inflammation. Pathogen recognition receptors like CD14, CD36, α6β1,
between mitochondria and ER deleteriously affects neurodegeneration and TLRs are employed to regulate the activities of ROS, NO, and cy­
as both produce ROS and compromise antioxidant defense [325]. tokines [346]. Moreover, AD cybrids have demonstrated mitochondrial
Extracellular accumulation of Aβ alters glutaminergic neurotransmis­ dysfunction as evidenced by reduced activities of COX and elevated
sion via (N-methyl-D-aspartate) NMDA receptors resulting in a production of free radicals [347,348]. Post-mortem analysis of AD brain
concomitant hike in cytosolic Ca2+, and impairment of synaptic activity. cells reported decreasing amounts of COX subunits with the disease
Increased Ca2+ is rapidly taken up by the mitochondria and ER and this progression [349]. It has also been noted that elderly AD patients
overload encourages ROS formation. possess lower levels of COX in comparison to younger AD patients [350].
Higher than normal oxidative stress is a common observation in the These findings imply that COX decreases with ageing and may eventu­
AD brain. The effect of oxidative stress on the progression of AD was first ally pass a threshold, which may be associated with and contribute to
noted back in 1965 [27,326]. ROS-controlled lipid peroxidation pro­ the pathogenesis of AD [351]. By blocking COX with N-methyl­
duces toxic malonaldehyde in the brain that causes DNA damage [327, protoporphyrin, control fibroblasts were induced to undergo AD-like
328]. Post-mortem examinations have revealed an abundance of alterations, demonstrating that oxidative damage in AD substantially
oxidized cellular macromolecules in AD brain [329,330]. Simultaneous coincides with mitochondrial dysfunction [347]. AD brain with
accumulation of 8-oxoG responsible for initiating cell death pathways increased numbers of COX-deficient neurons further supports this hy­
and Aβ in the hippocampus and temporal cortex of AD brain further pothesis [352].
supports the interrelation between oxidative stress and AD [331,332]. With aging, intracellular oxidative stress tends to increase owing to
Mitochondrial Aβ deposition starts well ahead of extracellular plaques to the bioenergetic imbalance and mitochondrial dysfunction. Activity of
disrupt the respiratory chain by disturbing oxidative phosphorylation oxoguanine (oxoG) DNA glycosylase declines in aging brain that com­
[225]. While dysfunction in the electron transport chain of mitochon­ promises repairing capacity of nucleic acid damage due to modification
dria characterized by a selective reduction in COX activities has been of the enzyme by 4-hydroxynonenal, i.e., a bi-product of ROS-mediated
observed in platelets, lymphocytes, and post-mortem brain tissue of AD lipid peroxidation that promotes the formation of 8-oxoG [327,353].
patients [333,334]. Impaired COX of the mitochondrial electron trans­ Lipid peroxidation and production of 4-hydroxynonenal in neuronal and
port chain produces more H2O2 than usual when exposed to oxidative mitochondrial membranes are influenced by Aβ1-42 oligomers, which
stress, making it itself a powerful tool of ROS generation [335]. subsequently alters protein structures and functions. Smaller oligos can
Aβ is primarily responsible for the beginning of mitochondrial more easily penetrate lipid bilayers than bigger ones, which increases
dysfunction. Aβ induces mitochondrial dysfunction either by interacting the risk of oxidative damage [11]. The loss of LTP and subsequent
with Aβ binding alcohol dehydrogenase (ABAD) or by localizing into oxidative damage to synaptic membranes caused by Aβ oligomers ulti­
cristae by virtue of translocase present in the outer mitochondrial mately impact learning and memory processes. Oxidative damage to
membrane [336]. In an AD mouse model, reduced activity of ABAD was synapses causes malfunctioning in interneuron communications, cyto­
observed due to the mitochondrial localization of ABAD-Aβ conjugate skeletal dynamics, and neuronal plasticity [354]. Aβ-mediated in­
resulting in elevated ROS levels and consequent mitochondrial impair­ crements in ROS and RNS trigger a series of molecular events through
ment [337]. Aβ alters mitochondrial membrane permeability and re­ TGF-β, NOX, and NF-κB signaling favoring the onset of AD [355]. Free
duces enzyme activity involved in mitochondrial respiration to initiate radical-mediated TAU phosphorylation destabilizes neuronal microtu­
mitochondrial dysfunction leading to an oxidative stress and subsequent bules that in turn, hampers synaptic conductivity. The regular func­
neuronal damage [8]. Normal mitochondrial activity is also hampered tioning of the cells is hampered by peptides that are prone to
by the inhibition of APP in the mitochondria. A study has demonstrated aggregation as a consequence of oxidative damage to mitochondrial
that inhibition of the respiratory chain complexes causes an elevation of nucleic acids [351]. Elevated oxidative stress also hampers cholinergic
Aβ formation predominantly dependent on augmented ROS levels transmission by the reduction in cholinergic transporters and receptors
[338]. Early pathogenic characteristics of AD include synaptic mainly in the hippocampus and neocortex [356]. Increased expression
dysfunction and synaptic loss, which are likely caused by abnormalities of matrix metalloproteinase-9 controlled by elevated ROS level, in turn,
in synaptic mitochondria [339]. In this context, post-mortem examina­ favors AD development by allowing entry of inflammatory factors and
tion of AD patients has revealed an increase in oxidative biomarkers more ROS through the BBB into CNS [357].
localized in the synapses indicating the important role of oxidative stress Elevated oxidative stress oxidizes the bases in the mitochondrial
in AD-associated synaptic decay [340]. The presequence protease (PreP) nucleic acids that could serve as markers for AD, wherein 8-oxoG is found
suppression decreases the cleavage of Aβ in the temporal lobe of AD to be the most prominent among all [66]. Immunohistochemical evalua­
patients, which could be attributed to the high levels of lipid peroxi­ tion of postmortem AD brains revealed neuroplasmic accumulation of
dation [341]. In the AD brain, nerve impulse transduction and synaptic 8-oxoG in hippocampal Cornu Ammonis (CA)1 and CA3 pyramidal and
plasticity are compromised as a result of an imbalance in Ca2+ homeo­ temporal cortex neurons where Aβ is also highly accumulated [168].
stasis. Aβ and TAU play their parts by compromising the capacity of 2-deoxyguanosine is oxidized to 8-hydroxy-2-deoxyguanosine and subse­
mitochondria to handle Ca2+ metabolism properly leading to enhanced quently to 8-oxo-dGTP (8-Oxo-2′ -deoxyguanosine-5′ -triphosphate) that is
oxidative stress, altered MMP, decreased MPTP formation, and lowered mismatched to an adenine nucleotide converting guanine (G)-cytosine (C)
ATP production [342]. Upstream of these events, an imbalance in base pair to thymine (T)-adenine (A) base pair [358]. Interestingly, C can
intraneuronal Ca2+ activities promotes Aβ aggregation, which again aids be converted into uracil (U) by oxidative deamination and pair with A
in releasing Ca2+ from ER to further increase cytosolic Ca2+ [342]. instead of G, thus replication leads to the formation of T-A instead of C-G
Dementia and progressive cognitive decline are the two main fea­ base pair [359,360].
tures of AD. Most experts approve that neurodegeneration plays a sig­ Glucose is the sole source of energy for neurons and ROS play a role
nificant role in the gradual loss of memory and other cognitive in this aspect as well. Lower expressions of GLUT1 in the BBB and GLUT3
impairments [343,344]. The concomitant mitochondrial degeneration, in neurons mediated by ROS allow lesser transport of glucose into the
which is frequently brought on by numerous neuro-immune reactions, CNS and inside nerve cells, giving rise to glucose hypometabolism in the
makes the situation even more severe. The interaction of ROS, TAU brain [361]. AD neurons reportedly display elevated oxidative stress and
protein, and APP potentiates the formation of neurofibrillary tangles mitochondrial autophagy that leads to decreased mitochondrial mass
and Aβ depositions [41]. Beginning in the hippocampus and gradually and increased mitochondrial DNA in the cytosol. Increased COX level in

145
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

lipofuscin vacuoles has also been observed [36]. Although oxidative late-onset AD patients reportedly evinced multiple inter-related bio­
stress and inflammation are not exclusive to AD, the pathologies overlap energetic alterations, such as alterations in glycolysis, alterations in
extensively to contribute to the progression of AD. Amyloids are pro­ bioenergetic substrate processing and transfer of reducing agents,
posed to limit mitochondrial function at later stages to increase oxida­ reduced levels of NAD/NADH, downregulated glucose uptake and
tive stress and neuroinflammation, despite the fact that oxidative response to INS/IGF-1 signaling, decreased population of INS receptor
damage caused by mitochondria and the related inflammatory response and GLUT1, and changes in the metabolic transcriptome [380].
are implicated in the development of AD [362]. The ATN (Aβ deposition, TAU phosphorylation, and neuro­
degeneration) standard is cementing its place in the diagnosis of AD
7. Hypometabolism-mediated bioenergetic alterations in AD clustering neuroimaging techniques with standardized indicators [381].
Proteomic data in the animal model have suggested dysregulation of
The pathophysiology of AD has been widely associated with glucose metabolism in AD liver [382]. AD lymphocytes exhibit increased
disturbed bioenergetics, including elevated mitochondrial oxidative apoptosis [383–385]. In peripheral mononuclear blood cells of AD pa­
stress and impaired glucose metabolism. Energy metabolisms become tients, basal mitochondrial oxygen consumption was reduced, while
dysfunctional with age, especially in neurodegenerative diseases like dATP levels were increased [386].
AD. Reduction in brain metabolism occurs before the depositions of Aβ
and neurofibrillary tangles in AD. AD patients with glucose hypo­ 8. Interconnection between impaired glucose metabolism and
metabolism exhibits its onset from the hippocampus that gradually mitochondrial dysfunction in AD
spread to parieto-temporal lobe, posterior cingulate cortex, and the
frontal areas of the brain [363,364]. This also justifies the initial tran­ Mitochondrial dysfunction associated with impaired glucose meta­
sient dementia during AD as the hippocampus is responsible for memory bolism is viewed as significant contributing factor in the AD pathology.
functions. Disruption of the pyramidal neurons in the perforantal path Patients with AD have fewer mitochondria per neuron, even the existing
aids to disconnect the hippocampus from the rest of the cortex, strongly mitochondria differs in shapes and morphologies [243]. Glucose hypo­
contributing to the memory decline as observed during the early stages metabolism and mitochondrial dysfunction collectively give rise to
of AD [365]. During AD, glycolysis and Krebs cycle, both are compro­ several other pathophysiological factors namely oxidative stress, Aβ
mised mainly due to the poor performance of the pyruvate dehydroge­ build-up, TAU hyperphosphorylation, apoptosis, AGEs accumulation,
nase complex [366]. AD also involves the reduction in synaptic density and excitotoxicity [170]. Insulin can directly affect mitochondrial
and activity impairing impulse transduction, thinner axons with reduced glucose metabolism by activating pyruvate dehydrogenase that is vitally
myelin sheath also add to deficient impulse transduction [367]. These involved in bridging glycolysis and Kreb’s cycle.
neurons require higher energy thus hypometabolism associated with AD It has been observed that impaired insulin signaling in the AD brain
could be correlated with this pathophysiology. FDG-PET scanning can gives rise to mitochondrial dysfunction due to severe impairment in
differentiate between the initial stage of AD and non-AD related de­ glucose metabolism [387]. AD and type 2 DM share common features
mentia [368]. ApoE4 and presenilin 1 genes are suggested to be the like inflammation, oxidative stress, and impaired glucose and insulin
important players in the development of AD and glucose hypo­ metabolism. Evidence from FDG-PET imaging has revealed that glucose
metabolism at the temporoparietal and posterior cingulate cortex acts as uptake in the temporal-parietal cortex gets hampered in AD patients
a key contributor [369]. It is also claimed that cerebral hypometabolism even before the appearance of cognitive and behavioral symptoms
of glucose may be associated with grey matter atrophy [370]. However, [388]. Abnormal basal phosphorylation levels of proteins in the IRS/Akt
during cognition disorders, hypometabolism of glucose and Aβ are pathway were also seen in different studies, involving post-mortem AD
regulated independently from each other [351]. Moreover, elevated brains. These abnormalities contribute positively to Aβ and TAU lesions
production of ketone bodies has been observed during AD [371]. and negatively to memory and cognition. Insulin resistance was revealed
Hypometabolism of glucose during AD enforces metabolic shift in the to be associated with Aβ and TAU lesions independent of cognitive and
brain from glucose to lipid for survival, which again correlates with memory functions in the hippocampal region [103,120]. Contemporary
amyloid and TAU pathology [372]. research also supports the notion that mitochondria are the primary
Neurodegenerative disorders are highly susceptible to mitochondri­ metabolic platforms of cell’s malfunction during insulin resistance [389,
opathy owing to the dependency of nerve cells on oxidative phosphor­ 390]. In the IRS/P13K/Akt cascade, Akt phosphorylates FOXO tran­
ylation to meet their high energy demands [373]. Aβ can act directly on scription factors responsible for gene expressions regarding lipid meta­
mitochondria and interact with ABAD which may cause mitochondrial bolism, neoglucogenesis, and stress resistance [391]. IRSs are strong
failure by increasing the MMP and reducing the activities of enzymes inhibitors of FOXO1 through Akt-mediated phosphorylation in the liver.
involved in mitochondrial respiration [336]. Again, the interaction of Aβ Insulin resistance causes hyperactivation of FOXO1 that in turn, induces
with CypD and COX enhances ROS production by disturbing the electron HMOX1 to consume heme to disturb the integrity of the electron
transport chain. These interactions also decrease the generation of ATP transport system within mitochondria leading to excessive ROS pro­
and thus affect energy production [374]. Aβ also interferes with Krebs duction [390,392]. ROS induces oxidative stress giving rise to several
cycle enzymes e.g., pyruvate dehydrogenase, malate dehydrogenase, neurodegenerative factors including the mutations of mitochondrial
and aconitase [375]. Antioxidant activities are also disrupted in AD DNA, apoptosis, and build-up of oxidative lesions [393]. Two essential
since Aβ interferes with SOD and catalase. However, the mitochondrial metabolic actions of mitochondria i.e., Kreb’s cycle and oxidative
cascade hypothesis proposes that impairment of the respiratory chain phosphorylation are found to be severely affected in AD brains [168].
and bioenergetic mechanisms are more important players in the AD This gives rise to not only the hypometabolism of glucose but also an
development than Aβ pathologies [200]. increase in the production of ROS, oxidative damage, and apoptosis.
Although neurons depend on oxidative phosphorylation for long- Studies found that the aging-related hypometabolism in mitochondria is
term survival, impairment of mitochondrial functions enforce them to different from the hypometabolism due to AD as age-related hypo­
switch more to glycolysis for energy production [376]. Inverse Warburg metabolism occurs in the medial-lateral cortex, while AD majorly in­
hypothesis postulates AD as a downstream consequence of mitochon­ duces hypometabolism in the lateral frontal cortex [170,394]. The AD
drial dysregulation [377,378]. Thus, a malfunction in the respiratory pattern of glucose hypometabolism that precedes the clinical manifes­
chain and bioenergetics of mitochondria leads to the cell stress response, tation elevates, expands, and worsens through the clinical deterioration
changes in neurotransmitters, excessive ROS production, disrupted Ca2+ ensues.
homeostasis, Aβ buildup, activation of NF-κB, inflammation and Impaired glucose metabolism in brain cells, specifically modified
neuronal cell death [379]. Astrocytes and neural progenitor cells from thiamine metabolism and insulin resistance might be responsible for

146
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

promoting Aβ build-up and TAU hyperphosphorylation [395]. These central insulin resistance, impaired cerebral glucose metabolism, and
cascades are directly involved in AD pathophysiology and cognitive mitochondria failure may represent a dangerous trio in AD
dysfunction. During AD, mitochondria cannot consume pyruvate as fast pathophysiology.
as it is produced by glycolysis, as a result, lactate accumulation leads to
toxic acidosis [304]. Insulin resistance leads to excess activation of 9. Discussion
GSK-3β, this may activate β-secretase activity to result in increased
cellular accumulation of Aβ [396]. Alterations in thiamine metabolism The prevalence of neurological diseases is rising in tandem with the
led to a build-up of Aβ plaques and hyperphosphorylation of TAU. TAU increase in the aged population globally. Progressive dementia along
hyperphosphorylation and the formation of neurofibrillary tangles can with cognitive deficiencies characterize AD. Extracellular amyloid de­
also be induced by glucose transporter abnormalities. Insulin resistance posits, intracellular neurofibrillary tangles, cholinergic deficits, synaptic
promotes TAU hyperphosphorylation through the P13K/Akt pathway abnormalities, oxidative stress, and inflammation contribute to the gross
[397] while reducing E2K exaggerates plaque formation and memory cerebral bioenergetic malfunction in AD brain. During AD, signaling
deficits in animal models of AD [316]. The interruption of metabolism pathways including AMPK, IRS, mTOR, NF-κB, and SIRT1 in combina­
leads to hyperphosphorylated TAU and the production of Aβ1–42 [398]. tion mediate amyloid aggregation, inflammation, and mitochondrial
The reduction of glucose metabolism leads to activation of β-secretase, energy dynamics. Table 1 enlists biomarker-specific alterations during
which is critical to aggregation of amyloids [399]. Glycolysis in astro­ AD. A few studies proposed amyloids and TAU toxicity as upstream
cytes may also have a role in amyloid accumulation and cytotoxicity. events of mitochondrial dysfunction, while others claimed them to be
Inhibiting astrocytic glycolysis and associated lactate shuttle results in downstream events of abnormally functioning mitochondria [411,412].
an accumulation of amyloid protein and vulnerability to The proper functioning of both neurons and glial cells is critical in
amyloid-related cytotoxicity [400]. Insulin is also considered to be a neurodegenerative disorders. DM has been associated with neuronal
factor in the degradation of Aβ outside cells by altering the activity of dysfunction and cognitive decline, increasing the risk of developing AD.
IDE [401]. Amyloidogenesis, impaired brain insulin signaling, abnormal brain
The requirement and production of energy are much higher in the glucose metabolism, mitochondrial dysfunction, and oxidative stress are
brain than other organs in the body. This requires great share of mito­ common denominators in AD and DM that potentiate brain dysfunction.
chondrial activity, and any disruption to it can produce mitochondrial Volume losses in the hippocampus, entorhinal cortex, and para­
dysfunction as well as oxidative stress. Oxidative markers such as 8- hippocampal gyrus are repeatedly displayed during the development of
hydroxyguanosine is usually observed before hallmark features of AD- AD from mild cognitive impairment.
like Aβ precipitates and neurofibrillary tangles [402]. In AD, iron Gradual loss of cholinergic neurons, excessive oxidative stress, and
deposition has been reported to building up oxidative stress that causes
protein and DNA oxidation, and inactivation of brain muscarinic re­
ceptors required for memory [403]. Table 1
Apoptosis can be mediated by AD-related pathogenesis, such as AD-associated modulations in the brain.
oxidative stress, altered phosphorylation, mitochondrial DNA muta­ Sl Biomarkers Associated pathways Type of modulations
tions, etc. The neurons with AD-specific mutations are sensitive towards No.
mitochondrial toxin-induced apoptosis [310]. It has also been observed 1 Aldolase Glycolysis Downregulation
that Aβ encourages the release of cytochrome c from mitochondria in AD 2 ATP synthase Oxidative Downregulation
neurons, which induces neuronal apoptosis [404]. Glucose hypo­ phosphorylation
metabolism also leads to inflammation in brain tissues and enhances the (electron transport
chain)
pathological expression of AD. The inflammatory factors are the results
3 Aβ plaques Synaptic impulse Upregulation
of critical insults that include Aβ build-up, oxidative stress, and mito­ transduction
chondrial dysfunctions [405,406]. Suppression of autophagy in the 4 Caspases Neuronal apoptosis Upregulation
brain due to hypometabolism of glucose leads to CNS degeneration and 5 Complex I Oxidative Downregulation
phosphorylation
exacerbates the pathology of AD [407]. Another important stimulus for
(electron transport
autophagy is mTOR which can sense metabolites as well as hormonal chain)
signaling. mTOR is also involved in AD pathology as enhanced mTOR 6 Complex IV Oxidative Downregulation
signaling increases Aβ deposits and neurofibrillary tangles in the brain phosphorylation
[150]. Impaired glucose metabolism disrupts the uptake of glutamates (electron transport
chain)
via astrocytes giving rise to excitotoxicity, an early event in AD brains
7 Enolase Glycolysis Upregulation
[408]. 8 Lactate Glycolysis Upregulation
Serum profiling study has identified 12 deregulated proteins in AD dehydrogenase
patients including phosphoenolpyruvate carboxykinase 2, adenylate 9 Malate Kreb’s cycle Upregulation
kinase 2, heat shock protein family A member 9, cytochrome C, somatic, dehydrogenase
10 Phosphoglycerate Glycolysis Downregulation
dihydrolipoamide dehydrogenase, glycine amidinotransferase, etc. mutase
[409]. To add, AD pathology involves dysregulation of oxidative phos­ 11 Pro-apoptotic Neuronal apoptosis Upregulation
phorylation, inducible NOS (iNOS), and NF-κB signaling. proteins
Ridge and colleagues earlier have prepared a database of mito­ 12 Pyruvate Glycolysis Downregulation
dehydrogenase
chondrial genomes to aid future research on mitochondrial association
13 Pyruvate kinase Glycolysis Upregulation
with neurodegenerative disorders [410]. Since mitochondria are the 14 RNS Oxidative stress Upregulation
central coordinators of energy metabolism and are sources and targets of 15 ROS Oxidative stress Upregulation
ROS, their impairment may constitute a downstream event of DM 16 Succinate Kreb’s cycle Upregulation
and/or AD-associated abnormal brain insulin and glucose metabolism. dehydrogenase
17 TAU Synaptic impulse Hyperphosphorylation
Defective insulin signaling makes neurons energy-deficient and more transduction
vulnerable to oxidizing insults, which could cause structural and func­ 18 Trios phosphate Glycolysis Upregulation
tional alterations in mitochondria. Aβ and hyperphosphorylated TAU isomerase
synergistically impair mitochondrial bioenergetics and potentiate 19 α-ketogluterate Kreb’s cycle Downregulation
dehydrogenase
oxidative stress, accelerating neurodegenerative mechanisms. Overall,

147
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

formation of misfolded proteins form the base of AD. Research results [6] A. John, P.H. Reddy, Synaptic basis of Alzheimer’s disease: focus on synaptic
amyloid beta, P-tau and mitochondria, Ageing Res. Rev. 65 (2021), 101208,
from a rigorous mouse model of AD suggest that AMPK activators could
https://doi.org/10.1016/j.arr.2020.101208.
be viable drug candidates for the treatment of AD [413]. Multi-target [7] E. Tonnies, E. Trushina, Oxidative stress, synaptic dysfunction, and alzheimer’s
therapy collectively comprising of anti-aging pathways mentioned disease, J Alzheimers Dis 57 (4) (2017) 1105–1121, https://doi.org/10.3233/
earlier holds strong promise for impaired cellular metabolic processes in JAD-161088.
[8] H.A. Elfawy, B. Das, Crosstalk between mitochondrial dysfunction, oxidative
AD. Future might belong to double-impact therapeutic avenues meant stress, and age related neurodegenerative disease: etiologies and therapeutic
for both DM and AD. With the translation of renewed insight into the strategies, Life Sci. 218 (2019) 165–184, https://doi.org/10.1016/j.
mechanisms regarding cellular and mitochondrial abnormalities into lfs.2018.12.029.
[9] A. Chaudhary, R.S. Kalra, C. Huang, J. Prakash, S.C. Kaul, R. Wadhwa, 2, 3-
novel therapeutic alternatives against neuronal degeneration, it would Dihydro-3β-methoxy Withaferin-A protects normal cells against stress: molecular
minimize the gap between basic science and clinical research for AD. evidence of its potent cytoprotective activity, J Nat Prod 80 (10) (2017)
2756–2760, https://doi.org/10.1021/acs.jnatprod.7b00573.
[10] R. Wadhwa, R.S. Kalra, S.C. Kaul, CARF is a multi-module regulator of cell
10. Inference proliferation and a molecular bridge between cellular senescence and
carcinogenesis, Mech. Ageing Dev. 166 (2017) 64–68, https://doi.org/10.1016/j.
mad.2017.07.008.
Malfunctions in cerebral glucose metabolism kickstart a series of
[11] D.A. Butterfield, B. Halliwell, Oxidative stress, dysfunctional glucose metabolism
pathological events gradually leading to neurodegenerative disorders and Alzheimer disease, Nat. Rev. Neurosci. 20 (3) (2019) 148–160, https://doi.
including AD. Mitochondria, via multiple signaling pathways, play vital org/10.1038/s41583-019-0132-6.
roles in glucose metabolism. Clearly, mitochondrial dysfunction is an [12] D.A. Butterfield, Perspectives on oxidative stress in alzheimer’s disease and
predictions of future research emphases, J Alzheimers Dis 64 (s1) (2018)
important factor in the reduced utilization of glucose. Combined with S469–S479, https://doi.org/10.3233/JAD-179912.
the classic plaque and tangle pathology, glucose hypometabolism and [13] E. Lauretti, J.G. Li, A. Di Meco, D. Pratico, Glucose deficit triggers tau pathology
associated pathophysiological alterations initiate a chain of factors and synaptic dysfunction in a tauopathy mouse model, Transl. Psychiatry 7 (1)
(2017), e1020, https://doi.org/10.1038/tp.2016.296.
contributing to the initiation and exacerbation of AD. Glucose hypo­ [14] E. Rebelos, J.O. Rinne, P. Nuutila, L.L. Ekblad, Brain glucose metabolism in
metabolism supplemented with other factors like insulin resistance and health, obesity, and cognitive decline-does insulin have anything to do with it? A
mitochondrial dysfunction plays crucial roles in AD pathogenesis. narrative review, J. Clin. Med. 10 (7) (2021), https://doi.org/10.3390/
jcm10071532.
Multiple mitophagy pathways co-exist for the clearance of non- [15] K. Shah, S. Desilva, T. Abbruscato, The role of glucose transporters in brain
functional mitochondria, contributing factors include genetics, protein disease: diabetes and Alzheimer’s Disease, Int. J. Mol. Sci. 13 (10) (2012)
expressions, and stress responses. AD pathology might be reversed by 12629–12655.
[16] D. Bosco, A. Fava, M. Plastino, T. Montalcini, A. Pujia, Possible implications of
inhibiting excessive mitochondrial division, or by enhancing mitophagy. insulin resistance and glucose metabolism in Alzheimer’s disease pathogenesis,
To translate the scientific knowledge gathered so far into investigative J. Cell Mol. Med. 15 (9) (2011) 1807–1821, https://doi.org/10.3390/
clinical practice, a revised focus on novel therapeutic and/or prophy­ ijms131012629.
[17] B.P. Austin, V.A. Nair, T.B. Meier, G. Xu, H.A. Rowley, C.M. Carlsson, S.
lactic advancements is urgently needed.
C. Johnson, V. Prabhakaran, Effects of hypoperfusion in Alzheimer’s disease,
J Alzheimers Dis 26 (Suppl 3) (2011) 123–133, https://doi.org/10.3233/JAD-
Acknowledgements 2011-0010.
[18] T.C. Hammond, A.L. Lin, Glucose metabolism is a better marker for predicting
clinical alzheimer’s disease than amyloid or tau, J Cell Immunol 4 (1) (2022)
The study project (grant number 02(0275)/16/EMR-II) that was 15–18.
given to S.D. by the Council of Scientific and Industrial Research, New [19] B.J. Neth, S. Craft, Insulin resistance and alzheimer’s disease: bioenergetic
linkages, Front. Aging Neurosci. 9 (2017) 345, https://doi.org/10.3389/
Delhi, India, is appreciated by the authors. For the State government fnagi.2017.00345.
Research Fellowship (Reference No. R-11/13/20), P.C. acknowledged [20] M. Gejl, J. Rungby, B. Brock, A. Gjedde, At the centennial of Michaelis and
Jadavpur University. The facilities provided by Jadavpur University in Menten, competing Michaelis-Menten steps explain effect of GLP-1 on blood-
brain transfer and metabolism of glucose, Basic Clin. Pharmacol. Toxicol. 115 (2)
India are gratefully acknowledged by all the authors. For the Ram­ (2014) 162–171, https://doi.org/10.1111/bcpt.12240.
alingaswami Fellowship, R.K. thanked DBT, India (NO. BT/RLF/Re- [21] E.A. Winkler, Y. Nishida, A.P. Sagare, S.V. Rege, R.D. Bell, D. Perlmutter, J.
entry/22/2016), Kakatiya Medical College, Warangal, SAN.No. 102/ D. Sengillo, S. Hillman, P. Kong, A.R. Nelson, J.S. Sullivan, Z. Zhao, H.
J. Meiselman, R.B. Wendy, J. Soto, E.D. Abel, J. Makshanoff, E. Zuniga, D.C. De
IFD/SAN/1117/2018–19, and CSIR-IICT Hyderabad. J.V. and R.K.
Vivo, B.V. Zlokovic, GLUT1 reductions exacerbate Alzheimer’s disease vasculo-
acknowledged DBT for the grant-No.BT/PR35841/MED/32/745/2020. neuronal dysfunction and degeneration, Nat. Neurosci. 18 (4) (2015) 521–530,
P.H.R. likes to express his heartfelt thanks to NIH for his projects https://doi.org/10.1038/nn.3966.
[22] Y. An, V.R. Varma, S. Varma, R. Casanova, E. Dammer, O. Pletnikova, C.W. Chia,
(R01AG042178, R01AG47812, R01NS105473, AG060767, AG069333,
J.M. Egan, L. Ferrucci, J. Troncoso, A.I. Levey, J. Lah, N.T. Seyfried, C. Legido-
and AG066347). Finally, the authors are grateful to the editor and re­ Quigley, R. O’Brien, M. Thambisetty, Evidence for brain glucose dysregulation in
viewers for their in-depth analyses and insightful criticism, which aided Alzheimer’s disease, Alzheimers Dement 14 (3) (2018) 318–329, https://doi.org/
the scientific quality of this project. 10.1016/j.jalz.2017.09.011.
[23] J.C. Ryu, E.R. Zimmer, P. Rosa-Neto, S.O. Yoon, Consequences of metabolic
disruption in alzheimer’s disease pathology, Neurotherapeutics 16 (3) (2019)
References 600–610, https://doi.org/10.1007/s13311-019-00755-y.
[24] T.M. Piers, K. Cosker, A. Mallach, G.T. Johnson, R. Guerreiro, J. Hardy, J.
M. Pocock, A locked immunometabolic switch underlies TREM2 R47H loss of
[1] P.J. Gallaway, H. Miyake, M.S. Buchowski, M. Shimada, Y. Yoshitake, A.S. Kim,
function in human iPSC-derived microglia, Faseb. J. 34 (2) (2020) 2436–2450,
N. Hongu, Physical activity: a viable way to reduce the risks of mild cognitive
https://doi.org/10.1096/fj.201902447R.
impairment, alzheimer’s disease, and vascular dementia in older adults, Brain Sci.
[25] C. Chinopoulos, T.N. Seyfried, Mitochondrial substrate-level phosphorylation as
7 (2) (2017) 22, https://doi.org/10.3390/brainsci7020022.
energy source for glioblastoma: review and hypothesis, ASN Neuro 10 (2018),
[2] J. Rayathala, K. Kumar, P. Venkatesh, Review on Alzheimer’s disease: past,
1759091418818261, https://doi.org/10.1177/1759091418818261.
present and future, J Innov Appl Pharm Sci 7 (1) (2022) 28–31, https://doi.org/
[26] R. Santangelo, M.L. Giuffrida, C. Satriano, M.F. Tomasello, S. Zimbone, A. Copani,
10.1017/S135561771700100X.
beta-amyloid monomers drive up neuronal aerobic glycolysis in response to
[3] P. Vercruysse, D. Vieau, D. Blum, A. Petersen, L. Dupuis, Hypothalamic
energy stressors, Aging (Albany NY) 13 (14) (2021) 18033–18050, https://doi.
alterations in neurodegenerative diseases and their relation to abnormal energy
org/10.18632/aging.203330.
metabolism, Front. Mol. Neurosci. 11 (2018) 2, https://doi.org/10.3389/
[27] L. Mosconi, Glucose metabolism in normal aging and Alzheimer’s disease:
fnmol.2018.00002.
methodological and physiological considerations for PET studies, Clin Transl
[4] A.I. Duarte, M.S. Santos, C.R. Oliveira, P.I. Moreira, Brain insulin signalling,
Imaging 1 (4) (2013), https://doi.org/10.1007/s40336-013-0026-y.
glucose metabolism and females’ reproductive aging: a dangerous triad in
[28] A.G. Vlassenko, S.N. Vaishnavi, L. Couture, D. Sacco, B.J. Shannon, R.H. Mach, J.
Alzheimer’s disease, Neuropharmacology 136 (Pt B) (2018) 223–242, https://
C. Morris, M.E. Raichle, M.A. Mintun, Spatial correlation between brain aerobic
doi.org/10.1016/j.neuropharm.2018.01.044.
glycolysis and amyloid-beta (Aβ) deposition, Proc. Natl. Acad. Sci. U. S. A. 107
[5] N. Raz, A.M. Daugherty, Pathways to brain aging and their modifiers: free-
(41) (2010) 17763–17767, https://doi.org/10.1073/pnas.1010461107.
radical-induced energetic and neural decline in senescence (FRIENDS) model - a
[29] M.S. Uddin, A.A. Mamun, S. Takeda, M.S. Sarwar, M.M. Begum, Analyzing the
mini-review, Gerontology 64 (1) (2018) 49–57, https://doi.org/10.1159/
chance of developing dementia among geriatric people: a cross-sectional pilot
000479508.

148
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

study in Bangladesh, Psychogeriatrics 19 (2) (2019) 87–94, https://doi.org/ susceptibility to amyloid-beta peptides modeling alzheimer’s disease,
10.1111/psyg.12368. J Alzheimers Dis 60 (2) (2017) 525–536, https://doi.org/10.3233/JAD-170356.
[30] T.T. Nguyen, Q.T.H. Ta, T.T.D. Nguyen, T.T. Le, V.G. Vo, Role of insulin [52] M. Oksanen, A.J. Petersen, N. Naumenko, K. Puttonen, S. Lehtonen, M. Gubert
resistance in the alzheimer’s disease progression, Neurochem. Res. 45 (7) (2020) Olive, A. Shakirzyanova, S. Leskela, T. Sarajarvi, M. Viitanen, J.O. Rinne,
1481–1491, https://doi.org/10.1007/s11064-020-03031-0. M. Hiltunen, A. Haapasalo, R. Giniatullin, P. Tavi, S.C. Zhang, K.M. Kanninen, R.
[31] L. Sousa, M. Guarda, M.J. Meneses, M.P. Macedo, H. Vicente Miranda, Insulin- H. Hamalainen, J. Koistinaho, PSEN1 mutant iPSC-derived model reveals severe
degrading enzyme: an ally against metabolic and neurodegenerative diseases, astrocyte pathology in alzheimer’s disease, Stem Cell Rep. 9 (6) (2017)
J. Pathol. 255 (4) (2021) 346–361, https://doi.org/10.1002/path.5777. 1885–1897, https://doi.org/10.1016/j.stemcr.2017.10.016.
[32] A.H. Dyer, C. Vahdatpour, A. Sanfeliu, D. Tropea, The role of Insulin-Like Growth [53] S. Hashimoto, T.C. Saido, Critical review: involvement of endoplasmic reticulum
Factor 1 (IGF-1) in brain development, maturation and neuroplasticity, stress in the aetiology of alzheimer’s disease, Open Biol 8 (4) (2018), 180024,
Neuroscience 325 (2016) 89–99, https://doi.org/10.1016/j. https://doi.org/10.1098/rsob.180024.
neuroscience.2016.03.056. [54] P. Jadiya, D.W. Kolmetzky, D. Tomar, A. Di Meco, A.A. Lombardi, J.P. Lambert,
[33] W. Ahmad, B. Ijaz, K. Shabbiri, F. Ahmed, S. Rehman, Oxidative toxicity in T.S. Luongo, M.H. Ludtmann, D. Pratico, J.W. Elrod, Impaired mitochondrial
diabetes and Alzheimer’s disease: mechanisms behind ROS/RNS generation, calcium efflux contributes to disease progression in models of Alzheimer’s
J. Biomed. Sci. 24 (1) (2017) 76, https://doi.org/10.1186/s12929-017-0379-z. disease, Nat. Commun. 10 (1) (2019) 3885, https://doi.org/10.1038/s41467-
[34] S. Zhang, B.B. Lachance, M.P. Mattson, X. Jia, Glucose metabolic crosstalk and 019-11813-6.
regulation in brain function and diseases, Prog. Neurobiol. 204 (2021), 102089, [55] N. Esteras, A.Y. Abramov, Mitochondrial calcium deregulation in the mechanism
https://doi.org/10.1016/j.pneurobio.2021.102089. of beta-amyloid and tau pathology, Cells 9 (9) (2020) 2135, https://doi.org/
[35] R.Z. Zhao, S. Jiang, L. Zhang, Z.B. Yu, Mitochondrial electron transport chain, 10.3390/cells9092135.
ROS generation and uncoupling, Int. J. Mol. Med. 44 (1) (2019) 3–15, https:// [56] A. Kubis-Kubiak, A. Dyba, A. Piwowar, The interplay between diabetes and
doi.org/10.3892/ijmm.2019.4188. alzheimer’s disease-in the hunt for biomarkers, Int. J. Mol. Sci. 21 (8) (2020)
[36] S.C. Correia, R.X. Santos, C. Carvalho, S. Cardoso, E. Candeias, M.S. Santos, C. 2744, https://doi.org/10.3390/ijms21082744.
R. Oliveira, P.I. Moreira, Insulin signaling, glucose metabolism and mitochondria: [57] C.V. Chandarana, S. Roy, Comprehensive review on neuro-degenerative type 3
major players in Alzheimer’s disease and diabetes interrelation, Brain Res. 1441 DM, Curr. Diabetes Rev. 18 (8) (2022), e131221198790, https://doi.org/
(2012) 64–78, https://doi.org/10.1016/j.brainres.2011.12.063. 10.2174/1573399818666211213103624.
[37] Y. Stern, Cognitive reserve in ageing and Alzheimer’s disease, Lancet Neurol. 11 [58] S. Cho, H. Lee, J. Seo, Impact of genetic risk factors for alzheimer’s disease on
(11) (2012) 1006–1012, https://doi.org/10.1016/S1474-4422(12)70191-6. brain glucose metabolism, Mol. Neurobiol. 58 (6) (2021) 2608–2619, https://doi.
[38] M. Gliozzi, V. Musolino, F. Bosco, M. Scicchitano, F. Scarano, S. Nucera, M. org/10.1007/s12035-021-02297-x.
C. Zito, S. Ruga, C. Carresi, R. Macri, L. Guarnieri, J. Maiuolo, A. Tavernese, A. [59] A.T. Balana, M.R. Pratt, Mechanistic roles for altered O-GlcNAcylation in
R. Coppoletta, C. Nicita, R. Mollace, E. Palma, C. Muscoli, C. Belzung, V. Mollace, neurodegenerative disorders, Biochem. J. 478 (14) (2021) 2733–2758, https://
Cholesterol homeostasis: researching a dialogue between the brain and peripheral doi.org/10.1042/BCJ20200609.
tissues, Pharmacol. Res. 163 (2021), 105215, https://doi.org/10.1016/j. [60] H.J. Park, K.W. Lee, S. Oh, R. Yan, J. Zhang, T.G. Beach, C.H. Adler, M. Voronkov,
phrs.2020.105215. S.P. Braithwaite, J.B. Stock, M.M. Mouradian, Protein phosphatase 2A and its
[39] S.A. Yuzwa, X. Shan, M.S. Macauley, T. Clark, Y. Skorobogatko, K. Vosseller, D. methylation modulating enzymes LCMT-1 and PME-1 are dysregulated in
J. Vocadlo, Increasing O-GlcNAc slows neurodegeneration and stabilizes tau tauopathies of progressive supranuclear palsy and alzheimer disease,
against aggregation, Nat. Chem. Biol. 8 (4) (2012) 393–399, https://doi.org/ J. Neuropathol. Exp. Neurol. 77 (2) (2018) 139–148, https://doi.org/10.1093/
10.1038/nchembio.797. jnen/nlx110.
[40] K. Batkulwar, R. Godbole, R. Banarjee, O. Kassaar, R.J. Williams, M.J. Kulkarni, [61] K. Iqbal, S. Bolognin, X. Wang, G. Basurto-Islas, J. Blanchard, Y.C. Tung, Animal
Advanced glycation end products modulate amyloidogenic APP processing and models of the sporadic form of Alzheimer’s disease: focus on the disease and not
tau phosphorylation: a mechanistic link between glycation and the development just the lesions, J Alzheimers Dis 37 (3) (2013) 469–474, https://doi.org/
of alzheimer’s disease, ACS Chem. Neurosci. 9 (5) (2018) 988–1000, https://doi. 10.3233/JAD-130827.
org/10.1021/acschemneuro.7b00410. [62] R. Kandimalla, V. Thirumala, P.H. Reddy, Is Alzheimer’s disease a Type 3
[41] G. Ogunmokun, S. Dewanjee, P. Chakraborty, C. Valupadas, A. Chaudhary, Diabetes? A critical appraisal, Biochim. Biophys. Acta, Mol. Basis Dis. 1863 (5)
V. Kolli, U. Anand, J. Vallamkondu, P. Goel, H.P.R. Paluru, K.D. Gill, P.H. Reddy, (2017) 1078–1089, https://doi.org/10.1016/j.bbadis.2016.08.018.
V. De Feo, R. Kandimalla, The potential role of cytokines and growth factors in [63] I. Sebastiao, E. Candeias, M.S. Santos, C.R. de Oliveira, P.I. Moreira, A.I. Duarte,
the pathogenesis of alzheimer’s disease, Cells 10 (10) (2021) 2790, https://doi. Insulin as a bridge between type 2 diabetes and alzheimer disease - how anti-
org/10.3390/cells10102790. diabetics could be a solution for dementia, Front. Endocrinol. 5 (2014) 110,
[42] M. Amaro, K. Kubiak-Ossowska, D.J.S. Birch, O.J. Rolinski, Initial stages of beta- https://doi.org/10.3389/fendo.2014.00110.
amyloid Abeta1-40 and Abeta1-42 oligomerization observed using fluorescence [64] S. Kullmann, A. Kleinridders, D.M. Small, A. Fritsche, H.U. Haring, H. Preissl,
decay and molecular dynamics analyses of tyrosine, Methods Appl. Fluoresc. 1 (1) M. Heni, Central nervous pathways of insulin action in the control of metabolism
(2013), 015006, https://doi.org/10.1088/2050-6120/1/1/015006. and food intake, Lancet Diabetes Endocrinol. 8 (6) (2020) 524–534, https://doi.
[43] S. Li, G.M. Shankar, D.J. Selkoe, How do soluble oligomers of amyloid beta- org/10.1016/S2213-8587(20)30113-3.
protein impair hippocampal synaptic plasticity? Front. Cell. Neurosci. 4 (2010) 5, [65] Y. Liu, F. Liu, I. Grundke-Iqbal, K. Iqbal, C.X. Gong, Deficient brain insulin
https://doi.org/10.3389/fncel.2010.00005. signalling pathway in Alzheimer’s disease and diabetes, J. Pathol. 225 (1) (2011)
[44] A. Emendato, G. Milordini, E. Zacco, A. Sicorello, F. Dal Piaz, R. Guerrini, 54–62, https://doi.org/10.1002/path.2912.
R. Thorogate, D. Picone, A. Pastore, Glycation affects fibril formation of Aβ [66] Y. Nakabeppu, Molecular pathophysiology of insulin depletion, mitochondrial
peptides, J. Biol. Chem. 293 (34) (2018) 13100–13111, https://doi.org/10.1074/ dysfunction, and oxidative stress in alzheimer’s disease brain, Adv. Exp. Med.
jbc.RA118.002275. Biol. 1128 (2019) 27–44, https://doi.org/10.1007/978-981-13-3540-2_3.
[45] M. Pera, D. Larrea, C. Guardia-Laguarta, J. Montesinos, K.R. Velasco, R. [67] S.E. Arnold, Z. Arvanitakis, S.L. Macauley-Rambach, A.M. Koenig, H.Y. Wang, R.
R. Agrawal, Y. Xu, R.B. Chan, G. Di Paolo, M.F. Mehler, G.S. Perumal, F. S. Ahima, S. Craft, S. Gandy, C. Buettner, L.E. Stoeckel, D.M. Holtzman, D.
P. Macaluso, Z.Z. Freyberg, R. Acin-Perez, J.A. Enriquez, E.A. Schon, E. Area- M. Nathan, Brain insulin resistance in type 2 diabetes and Alzheimer disease:
Gomez, Increased localization of APP-C99 in mitochondria-associated ER concepts and conundrums, Nat. Rev. Neurol. 14 (3) (2018) 168–181, https://doi.
membranes causes mitochondrial dysfunction in Alzheimer disease, EMBO J. 36 org/10.1038/nrneurol.2017.185.
(22) (2017) 3356–3371, https://doi.org/10.15252/embj.201796797. [68] C. Holscher, Insulin signaling impairment in the brain as a risk factor in
[46] S. Li, Z.H. Sheng, Energy matters: presynaptic metabolism and the maintenance of alzheimer’s disease, Front. Aging Neurosci. 11 (2019) 88, https://doi.org/
synaptic transmission, Nat. Rev. Neurosci. 23 (1) (2022) 4–22, https://doi.org/ 10.3389/fnagi.2019.00088.
10.1038/s41583-021-00535-8. [69] T. Scherer, K. Sakamoto, C. Buettner, Brain insulin signalling in metabolic
[47] M.C. Hughes, S.V. Ramos, P.C. Turnbull, I.A. Rebalka, A. Cao, C.M.F. Monaco, N. homeostasis and disease, Nat. Rev. Endocrinol. 17 (8) (2021) 468–483.
E. Varah, B.A. Edgett, J.S. Huber, P. Tadi, L.J. Delfinis, U. Schlattner, J. [70] G. Molnar, N. Farago, A.K. Kocsis, M. Rozsa, S. Lovas, E. Boldog, R. Baldi,
A. Simpson, T.J. Hawke, C.G.R. Perry, Early myopathy in Duchenne muscular E. Csajbok, J. Gardi, L.G. Puskas, G. Tamas, GABAergic neurogliaform cells
dystrophy is associated with elevated mitochondrial H2O2 emission during represent local sources of insulin in the cerebral cortex, J. Neurosci. 34 (4) (2014)
impaired oxidative phosphorylation, J Cachexia Sarcopenia Muscle 10 (3) (2019) 1133–1137, https://doi.org/10.1038/s41574-021-00498-x.
643–661, https://doi.org/10.1002/jcsm.12405. [71] H. Nagano, S. Ito, T. Masuda, S. Ohtsuki, Effect of insulin receptor-knockdown on
[48] P.H. Reddy, M.F. Beal, Amyloid beta, mitochondrial dysfunction and synaptic the expression levels of blood-brain barrier functional proteins in human brain
damage: implications for cognitive decline in aging and Alzheimer’s disease, microvascular endothelial cells, Pharm. Res. (N. Y.) 39 (7) (2022) 1561–1574,
Trends Mol. Med. 14 (2) (2008) 45–53, https://doi.org/10.3390/cells8050488. https://doi.org/10.1007/s11095-021-03131-8.
[49] M.J. Calkins, P.H. Reddy, Amyloid beta impairs mitochondrial anterograde [72] J. Levenga, H. Wong, R.A. Milstead, B.N. Keller, L.E. LaPlante, C.A. Hoeffer, AKT
transport and degenerates synapses in Alzheimer’s disease neurons, Biochim. isoforms have distinct hippocampal expression and roles in synaptic plasticity,
Biophys. Acta 1812 (4) (2011) 507–513, https://doi.org/10.1016/j. Elife 6 (2017), e30640, https://doi.org/10.7554/eLife.30640.
bbadis.2011.01.007. [73] M.P. Sajan, M.C. Lee, F. Foufelle, J. Sajan, C. Cleland, R.V. Farese, Coordinated
[50] M. Manczak, M.J. Calkins, P.H. Reddy, Impaired mitochondrial dynamics and regulation of hepatic FoxO1, PGC-1alpha and SREBP-1c facilitates insulin action
abnormal interaction of amyloid beta with mitochondrial protein Drp1 in neurons and resistance, Cell. Signal. 43 (2018) 62–70, https://doi.org/10.1016/j.
from patients with Alzheimer’s disease: implications for neuronal damage, Hum. cellsig.2017.12.005.
Mol. Genet. 20 (13) (2011) 2495–2509. [74] B.A. Pederson, Structure and regulation of glycogen synthase in the brain, Brain
[51] J.A. Amorim, P.M. Canas, A.R. Tome, A.P. Rolo, P. Agostinho, C.M. Palmeira, R. Glycogen Metabolism 23 (2019) 83–123, https://doi.org/10.1007/978-3-030-
A. Cunha, Mitochondria in excitatory and inhibitory synapses have similar 27480-1_3.

149
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

[75] S. Dewanjee, J. Vallamkondu, R.S. Kalra, A. John, P.H. Reddy, R. Kandimalla, receptor substrate 1 and inhibit tyrosine phosphorylation, J. Biol. Chem. 289 (18)
Autophagy in the diabetic heart: a potential pharmacotherapeutic target in (2014) 12467–12484, https://doi.org/10.1074/jbc.M114.554162.
diabetic cardiomyopathy, Ageing Res. Rev. 68 (2021), 101338, https://doi.org/ [101] K. Morino, S. Neschen, S. Bilz, S. Sono, D. Tsirigotis, R.M. Reznick, I. Moore,
10.1016/j.arr.2021.101338. Y. Nagai, V. Samuel, D. Sebastian, M. White, W. Philbrick, G.I. Shulman, Muscle-
[76] H.S. Brunetta, G.P. Holloway, A theoretical argument to support the biological specific IRS-1 Ser->Ala transgenic mice are protected from fat-induced insulin
benefits for insulin stimulating mitochondrial oxidative phosphorylation, Curr resistance in skeletal muscle, Diabetes 57 (10) (2008) 2644–2651, https://doi.
Opin Physiol 25 (2022), 100491, https://doi.org/10.1016/j. org/10.2337/db06-0454.
cophys.2022.100491. [102] K.D. Copps, N.J. Hancer, L. Opare-Ado, W. Qiu, C. Walsh, M.F. White, Irs1 serine
[77] J. Yip, X. Geng, J. Shen, Y. Ding, Cerebral gluconeogenesis and diseases, Front. 307 promotes insulin sensitivity in mice, Cell Metabol. 11 (1) (2010) 84–92,
Pharmacol. 7 (2017) 521, https://doi.org/10.3389/fphar.2016.00521. https://doi.org/10.1016/j.cmet.2009.11.003.
[78] T. Miyamoto, H. Amrein, Neuronal gluconeogenesis regulates systemic glucose [103] T.R. Bomfim, L. Forny-Germano, L.B. Sathler, J. Brito-Moreira, J.C. Houzel,
homeostasis in Drosophila melanogaster, Curr. Biol. 29 (8) (2019) 1263–1272, H. Decker, M.A. Silverman, H. Kazi, H.M. Melo, P.L. McClean, C. Holscher, S.
https://doi.org/10.1016/j.cub.2019.02.053. E. Arnold, K. Talbot, W.L. Klein, D.P. Munoz, S.T. Ferreira, F.G. De Felice, An anti-
[79] C.C. Martin, J.K. Oeser, C.A. Svitek, S.I. Hunter, J.C. Hutton, R.M. O’Brien, diabetes agent protects the mouse brain from defective insulin signaling caused
Identification and characterization of a human cdna and gene encoding a by Alzheimer’s disease- associated Abeta oligomers, J. Clin. Invest. 122 (4) (2012)
ubiquitously expressed glucose-6-phosphatase catalytic subunit-related protein, 1339–1353, https://doi.org/10.1172/JCI57256.
J. Mol. Endocrinol. 29 (2002) 205–222, https://doi.org/10.1677/jme.0.0290205. [104] M.V. Lourenco, J.R. Clarke, R.L. Frozza, T.R. Bomfim, L. Forny-Germano, A.
[80] A. Herrero-Mendez, A. Almeida, E. Fernández, C. Maestre, S. Moncada, J.P. Bola- F. Batista, L.B. Sathler, J. Brito-Moreira, O.B. Amaral, C.A. Silva, L. Freitas-Correa,
os, The bioenergetic and antioxidant status of neurons is controlled by continuous S. Espirito-Santo, P. Campello-Costa, J.C. Houzel, W.L. Klein, C. Holscher, J.
degradation of a key glycolytic enzyme by apc/C-Cdh1, Nat. Cell Biol. 11 (2009) B. Carvalheira, A.M. Silva, L.A. Velloso, D.P. Munoz, S.T. Ferreira, F.G. De Felice,
747–752, https://doi.org/10.1038/ncb1881. TNF-alpha mediates PKR-dependent memory impairment and brain IRS-1
[81] E.M. Schrijvers, J.C. Witteman, E.J. Sijbrands, A. Hofman, P.J. Koudstaal, M. inhibition induced by Alzheimer’s beta-amyloid oligomers in mice and monkeys,
M. Breteler, Insulin metabolism and the risk of alzheimer disease: the rotterdam Cell Metabol. 18 (6) (2013) 831–843, https://doi.org/10.1016/j.
study, Neurology 75 (22) (2010) 1982–1987, https://doi.org/10.1212/ cmet.2013.11.002.
WNL.0b013e3181ffe4f6. [105] Q.L. Ma, F. Yang, E.R. Rosario, O.J. Ubeda, W. Beech, D.J. Gant, P.P. Chen,
[82] M.A. Potenza, L. Sgarra, V. Desantis, C. Nacci, M. Montagnani, Diabetes and B. Hudspeth, C. Chen, Y. Zhao, H.V. Vinters, S.A. Frautschy, G.M. Cole, Beta-
alzheimer’s disease: might mitochondrial dysfunction help deciphering the amyloid oligomers induce phosphorylation of tau and inactivation of insulin
common path? Antioxidants 10 (8) (2021) 1257, https://doi.org/10.3390/ receptor substrate via c-Jun N-terminal kinase signaling: suppression by omega-3
antiox10081257. fatty acids and curcumin, J. Neurosci. 29 (28) (2009) 9078–9089, https://doi.
[83] K.D. Niswender, Basal insulin: beyond glycemia, Postgrad. Med. 123 (4) (2011) org/10.1523/JNEUROSCI.1071-09.2009.
27–37, https://doi.org/10.3810/pgm.2011.07.2301. [106] L. Liang, J. Chen, L. Zhan, X. Lu, X. Sun, H. Sui, L. Zheng, H. Xiang, F. Zhang,
[84] Y. Hou, X. Dan, M. Babbar, Y. Wei, S.G. Hasselbalch, D.L. Croteau, V.A. Bohr, Endoplasmic reticulum stress impairs insulin receptor signaling in the brains of
Ageing as a risk factor for neurodegenerative disease, Nat. Rev. Neurol. 15 (10) obese rats, PLoS One 10 (5) (2015), e0126384, https://doi.org/10.1371/journal.
(2019) 565–581, https://doi.org/10.1038/s41582-019-0244-7. pone.0126384.
[85] H.W. Querfurth, F.M. LaFerla, Alzheimer’s disease, N. Engl. J. Med. 362 (4) [107] E. Barone, F. Di Domenico, T. Cassano, A. Arena, A. Tramutola, M.A. Lavecchia,
(2010) 329–344, https://doi.org/10.1056/NEJMra0909142. R. Coccia, D.A. Butterfield, M. Perluigi, Impairment of biliverdin reductase-A
[86] L. Gasparini, G.K. Gouras, R. Wang, R.S. Gross, M.F. Beal, P. Greengard, H. Xu, promotes brain insulin resistance in Alzheimer disease: a new paradigm, Free
Stimulation of beta-amyloid precursor protein trafficking by insulin reduces Radic. Biol. Med. 91 (2016) 127–142, https://doi.org/10.1016/j.
intraneuronal beta-amyloid and requires mitogen-activated protein kinase freeradbiomed.2015.
signaling, J. Neurosci. 21 (8) (2001) 2561–2570, https://doi.org/10.1523/ [108] V. Kothari, Y. Luo, T. Tornabene, A.M. O’Neill, M.W. Greene, T. Geetha, J.
JNEUROSCI.21-08-02561.2001. R. Babu, High fat diet induces brain insulin resistance and cognitive impairment
[87] M. Vandal, P.J. White, C. Tremblay, I. St-Amour, G. Chevrier, V. Emond, in mice, Biochim. Biophys. Acta, Mol. Basis Dis. 1863 (2) (2017) 499–508,
D. Lefrancois, J. Virgili, E. Planel, Y. Giguere, A. Marette, F. Calon, Insulin https://doi.org/10.1016/j.bbadis.2016.10.006.
reverses the high-fat diet-induced increase in brain Abeta and improves memory [109] M. Yarchoan, J.B. Toledo, E.B. Lee, Z. Arvanitakis, H. Kazi, L.Y. Han, N. Louneva,
in an animal model of Alzheimer disease, Diabetes 63 (12) (2014) 4291–4301, V.M. Lee, S.F. Kim, J.Q. Trojanowski, S.E. Arnold, Abnormal serine
https://doi.org/10.2337/db14-0375. phosphorylation of insulin receptor substrate 1 is associated with tau pathology in
[88] O. Pivovarova, A. Hohn, T. Grune, A.F. Pfeiffer, N. Rudovich, Insulin-degrading Alzheimer’s disease and tauopathies, Acta Neuropathol. 128 (5) (2014) 679–689,
enzyme: new therapeutic target for diabetes and Alzheimer’s disease? Ann. Med. https://doi.org/10.1007/s00401-014-1328-5.
48 (8) (2016) 614–624, https://doi.org/10.1080/07853890.2016.1197416. [110] A.M. Moloney, R.J. Griffin, S. Timmons, R. O’Connor, R. Ravid, C. O’Neill,
[89] X. Ling, R.N. Martins, M. Racchi, S. Craft, E. Helmerhorst, Amyloid beta Defects in IGF-1 receptor, insulin receptor and IRS-1/2 in Alzheimer’s disease
antagonizes insulin promoted secretion of the amyloid beta protein precursor, indicate possible resistance to IGF-1 and insulin signalling, Neurobiol. Aging 31
J Alzheimers Dis 4 (5) (2002) 369–374, https://doi.org/10.3233/jad-2002-4504. (2) (2010) 224–243, https://doi.org/10.1016/j.neurobiolaging.2008.04.002.
[90] M. Fotuhi, V. Hachinski, P.J. Whitehouse, Changing perspectives regarding late- [111] A. Taguchi, L.M. Wartschow, M.F. White, Brain IRS2 signaling coordinates life
life dementia, Nat. Rev. Neurol. 5 (12) (2009) 649–658, https://doi.org/ span and nutrient homeostasis, Science 317 (5836) (2007) 369–372, https://doi.
10.1038/nrneurol.2009.175. org/10.1126/science.1142179.
[91] A. Tumminia, F. Vinciguerra, M. Parisi, L. Frittitta, Type 2 diabetes mellitus and [112] F.G. De Felice, R.A. Goncalves, S.T. Ferreira, Impaired insulin signalling and
alzheimer’s disease: role of insulin signalling and therapeutic implications, Int. J. allostatic load in Alzheimer disease, Nat. Rev. Neurosci. 23 (4) (2022) 215–230,
Mol. Sci. 19 (11) (2018) 3306, https://doi.org/10.3390/ijms19113306. https://doi.org/10.1038/s41583-022-00558-9.
[92] Y. Zhang, N.Q. Huang, F. Yan, H. Jin, S.Y. Zhou, J.S. Shi, F. Jin, Diabetes mellitus [113] S. Freude, M.M. Hettich, C. Schumann, O. Stohr, L. Koch, C. Kohler,
and Alzheimer’s disease: GSK-3beta as a potential link, Behav. Brain Res. 339 M. Udelhoven, U. Leeser, M. Muller, N. Kubota, T. Kadowaki, W. Krone,
(2018) 57–65, https://doi.org/10.1016/j.bbr.2017.11.015. H. Schroder, J.C. Bruning, M. Schubert, Neuronal IGF-1 resistance reduces Abeta
[93] K. Becker, S. Freude, J. Zemva, O. Stohr, W. Krone, M. Schubert, Chronic accumulation and protects against premature death in a model of Alzheimer’s
peripheral hyperinsulinemia has no substantial influence on tau phosphorylation disease, Faseb. J. 23 (10) (2009) 3315–3324, https://doi.org/10.1096/fj.09-
in vivo, Neurosci. Lett. 516 (2) (2012) 306–310, https://doi.org/10.1016/j. 132043.
neulet.2012.04.022. [114] S. Cardoso, I.P. Lopez, S. Pineiro-Hermida, J.G. Pichel, P.I. Moreira, IGF1R
[94] J.C. Shieh, P.T. Huang, Y.F. Lin, Alzheimer’s disease and diabetes: insulin deficiency modulates brain signaling pathways and disturbs mitochondria and
signaling as the bridge linking two pathologies, Mol. Neurobiol. 57 (4) (2020) redox homeostasis, Biomedicines 9 (2) (2021), https://doi.org/10.3390/
1966–1977, https://doi.org/10.1007/s12035-019-01858-5. biomedicines9020158.
[95] A. Jozefiak, M. Larska, M. Pomorska-Mol, J.J. Ruszkowski, The IGF-1 signaling [115] R. Killick, G. Scales, K. Leroy, M. Causevic, C. Hooper, E.E. Irvine, A.I. Choudhury,
pathway in viral infections, Viruses 13 (8) (2021) 1488, https://doi.org/10.3390/ L. Drinkwater, F. Kerr, H. Al-Qassab, J. Stephenson, Z. Yilmaz, K.P. Giese, J.
v13081488. P. Brion, D.J. Withers, S. Lovestone, Deletion of Irs2 reduces amyloid deposition
[96] M.F. White, Insulin signaling in health and disease, Science 302 (5651) (2003) and rescues behavioural deficits in APP transgenic mice, Biochem. Biophys. Res.
1710–1711, https://doi.org/10.1126/science.1092952. Commun. 386 (1) (2009) 257–262, https://doi.org/10.1016/j.bbrc.2009.06.032.
[97] M.F. White, C.R. Kahn, Insulin action at a molecular level - 100 years of progress, [116] D. Tanokashira, W. Fukuokaya, A. Taguchi, Involvement of insulin receptor
Mol. Metabol. 52 (2021), 101304, https://doi.org/10.1016/j. substrates in cognitive impairment and Alzheimer’s disease, Neural Regen Res 14
molmet.2021.101304. (8) (2019) 1330–1334, https://doi.org/10.4103/1673-5374.253535.
[98] W. Cai, M. Sakaguchi, A. Kleinridders, G. Gonzalez-Del Pino, J.M. Dreyfuss, B. [117] O. Stohr, K. Schilbach, L. Moll, M.M. Hettich, S. Freude, F.T. Wunderlich,
T. O’Neill, A.K. Ramirez, H. Pan, J.N. Winnay, J. Boucher, M.J. Eck, C.R. Kahn, M. Ernst, J. Zemva, J.C. Bruning, W. Krone, M. Udelhoven, M. Schubert, Insulin
Domain-dependent effects of insulin and IGF-1 receptors on signalling and gene receptor signaling mediates APP processing and beta-amyloid accumulation
expression, Nat. Commun. 8 (2017), 14892, https://doi.org/10.1038/ without altering survival in a transgenic mouse model of Alzheimer’s disease, Age
ncomms14892. (Dordr) 35 (1) (2013) 83–101, https://doi.org/10.1007/s11357-011-9333-2.
[99] L.R. Pearce, D. Komander, D.R. Alessi, The nuts and bolts of AGC protein kinases, [118] B. Kim, E.L. Feldman, Insulin resistance in the nervous system, Trends Endocrinol.
Nat. Rev. Mol. Cell Biol. 11 (1) (2010) 9–22, https://doi.org/10.1038/nrm2822. Metabol. 23 (3) (2012) 133–141, https://doi.org/10.1016/j.tem.2011.12.004.
[100] N.J. Hancer, W. Qiu, C. Cherella, Y. Li, K.D. Copps, M.F. White, Insulin and [119] S. Gabbouj, S. Ryhanen, M. Marttinen, R. Wittrahm, M. Takalo, S. Kemppainen,
metabolic stress stimulate multisite serine/threonine phosphorylation of insulin H. Martiskainen, H. Tanila, A. Haapasalo, M. Hiltunen, T. Natunen, Altered
insulin signaling in alzheimer’s disease brain - special emphasis on PI3K-Akt

150
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

pathway, Front. Neurosci. 13 (2019) 629, https://doi.org/10.3389/ Neuroscience 385 (2018) 246–254, https://doi.org/10.1016/j.
fnins.2019.00629. neuroscience.2018.05.002.
[120] K. Talbot, H.Y. Wang, H. Kazi, L.Y. Han, K.P. Bakshi, A. Stucky, R.L. Fuino, K. [142] J. Biswas, S. Gupta, D.K. Verma, P. Gupta, A. Singh, S. Tiwari, P. Goswami,
R. Kawaguchi, A.J. Samoyedny, R.S. Wilson, Z. Arvanitakis, J.A. Schneider, B. S. Sharma, S. Singh, Involvement of glucose related energy crisis and endoplasmic
A. Wolf, D.A. Bennett, J.Q. Trojanowski, S.E. Arnold, Demonstrated brain insulin reticulum stress: insinuation of streptozotocin induced Alzheimer’s like
resistance in Alzheimer’s disease patients is associated with IGF-1 resistance, IRS- pathology, Cell. Signal. 42 (2018) 211–226, https://doi.org/10.1016/j.
1 dysregulation, and cognitive decline, J. Clin. Invest. 122 (4) (2012) 1316–1338, cellsig.2017.10.018.
https://doi.org/10.1172/JCI59903. [143] E. Gil-Iturbe, M. Solas, M. Cuadrado-Tejedo, A. Garcia-Osta, X. Escote, M.
[121] S. Jimenez, M. Torres, M. Vizuete, R. Sanchez-Varo, E. Sanchez-Mejias, L. Trujillo- J. Ramirez, M.P. Lostao, GLUT12 expression in brain of mouse models of
Estrada, I. Carmona-Cuenca, C. Caballero, D. Ruano, A. Gutierrez, J. Vitorica, alzheimer’s disease, Mol. Neurobiol. 57 (2) (2020) 798–805, https://doi.org/
Age-dependent accumulation of soluble amyloid beta (Abeta) oligomers reverses 10.1007/s12035-019-01743-1.
the neuroprotective effect of soluble amyloid precursor protein-alpha (sAPP [144] Y. Hase, R. Ding, G. Harrison, E. Hawthorne, A. King, S. Gettings, C. Platten,
(alpha)) by modulating phosphatidylinositol 3-kinase (PI3K)/Akt-GSK-3beta W. Stevenson, L.J.L. Craggs, R.N. Kalaria, White matter capillaries in vascular and
pathway in Alzheimer mouse model, J. Biol. Chem. 286 (21) (2011) neurodegenerative dementias, Acta Neuropathol Commun 7 (1) (2019) 16,
18414–18425, https://doi.org/10.1074/jbc.M110.209718. https://doi.org/10.1111/jnc.14271.
[122] S.R. D’Mello, When good kinases go rogue: GSK3, p38 MAPK and CDKs as [145] Y. Liu, F. Liu, K. Iqbal, I. Grundke-Iqbal, C.X. Gong, Decreased glucose
therapeutic targets for alzheimer’s and huntington’s disease, Int. J. Mol. Sci. 22 transporters correlate to abnormal hyperphosphorylation of tau in Alzheimer
(11) (2021) 5911, https://doi.org/10.3390/ijms22115911. disease, FEBS Lett. 582 (2) (2008) 359–364, https://doi.org/10.1016/j.
[123] J.H. Son, J.H. Shim, K.H. Kim, J.Y. Ha, J.Y. Han, Neuronal autophagy and febslet.2007.12.035.
neurodegenerative diseases, Exp. Mol. Med. 44 (2) (2012) 89–98, https://doi. [146] M. Salkovic-Petrisic, J. Osmanovic-Barilar, A. Knezovic, S. Hoyer, K. Mosetter,
org/10.3858/emm.2012.44.2.031. W. Reutter, Long-term oral galactose treatment prevents cognitive deficits in male
[124] D. Heras-Sandoval, J.M. Perez-Rojas, J. Hernandez-Damian, J. Pedraza-Chaverri, Wistar rats treated intracerebroventricularly with streptozotocin,
The role of PI3K/AKT/mTOR pathway in the modulation of autophagy and the Neuropharmacology 77 (2014) 68–80, https://doi.org/10.1016/j.
clearance of protein aggregates in neurodegeneration, Cell. Signal. 26 (12) (2014) neuropharm.2013.09.002.
2694–2701, https://doi.org/10.1016/j.cellsig.2014.08.019. [147] J. Pujol-Gimenez, E. Martisova, A. Perez-Mediavilla, M.P. Lostao, M.J. Ramirez,
[125] L. Stoica, P.J. Zhu, W. Huang, H. Zhou, S.C. Kozma, M. Costa-Mattioli, Selective Expression of the glucose transporter GLUT12 in Alzheimer’s disease patients,
pharmacogenetic inhibition of mammalian target of Rapamycin complex I J Alzheimers Dis 42 (1) (2014) 97–101, https://doi.org/10.1007/s12035-019-
(mTORC1) blocks long-term synaptic plasticity and memory storage, Proc. Natl. 01743-1.
Acad. Sci. U. S. A. 108 (9) (2011) 3791–3796, https://doi.org/10.1073/ [148] L.M. Chua, M.L. Lim, P.R. Chong, Z.P. Hu, N.S. Cheung, B.S. Wong, Impaired
pnas.1014715108. neuronal insulin signaling precedes Abeta42 accumulation in female AbetaPPsw/
[126] H.K. Lee, B. Kwon, C.A. Lemere, S. de la Monte, K. Itamura, A.Y. Ha, H. PS1DeltaE9 mice, J Alzheimers Dis 29 (4) (2012) 783–791, https://doi.org/
W. Querfurth, mTORC2 (rictor) in alzheimer’s disease and reversal of amyloid- 10.3233/JAD-2012-111880.
beta expression-induced insulin resistance and toxicity in rat primary cortical [149] B.V. Zlokovic, Neurovascular pathways to neurodegeneration in Alzheimer’s
neurons, J Alzheimers Dis 56 (3) (2017) 1015–1036, https://doi.org/10.3233/ disease and other disorders, Nat. Rev. Neurosci. 12 (12) (2011) 723–738, https://
JAD-161029. doi.org/10.1038/nrn3114.
[127] M.C. Calle, M.L. Fernandez, Inflammation and type 2 diabetes, Diabetes Metab. [150] Z. Cai, L.J. Yan, K. Li, S.H. Quazi, B. Zhao, Roles of AMP-activated protein kinase
38 (3) (2012) 183–191, https://doi.org/10.1016/j.diabet.2011.11.006. in Alzheimer’s disease, NeuroMolecular Med. 14 (1) (2012) 1–14, https://doi.
[128] M.T. Heneka, D.T. Golenbock, E. Latz, Innate immunity in Alzheimer’s disease, org/10.1007/s12017-012-8173-2.
Nat. Immunol. 16 (3) (2015) 229–236, https://doi.org/10.1038/ni.3102. [151] F. Yao, M. Zhang, L. Chen, 5’-Monophosphate-activated protein kinase (AMPK)
[129] C.A. Magalhaes, C.N. Ferreira, C.M.G. Loures, V.G. Fraga, A.C. Chaves, A.C. improves autophagic activity in diabetes and diabetic complications, Acta Pharm.
R. Oliveira, L.C. de Souza, E.P.F. Resende, K.C. Carmona, H.C. Guimaraes, M.T. Sin. B 6 (1) (2016) 20–25, https://doi.org/10.1016/j.apsb.2015.07.009.
G. Cintra, I.N. Lanna, D.A.G. Zauli, M.A. Bicalho, M.G. Carvalho, L.P. Sousa, [152] S.M. Jeon, Regulation and function of AMPK in physiology and diseases, Exp.
P. Caramelli, K.B. Gomes, Leptin, hsCRP, TNF-alpha and IL-6 levels from normal Mol. Med. 48 (7) (2016) e245, https://doi.org/10.1038/emm.2016.81.
aging to dementia: relationship with cognitive and functional status, J. Clin. [153] R. Khanra, N. Bhattacharjee, T.K. Dua, A. Nandy, A. Saha, J. Kalita, P. Manna,
Neurosci. 56 (2018) 150–155, https://doi.org/10.1016/j.jocn.2018.08.027. S. Dewanjee, Taraxerol, a pentacyclic triterpenoid, from Abroma augusta leaf
[130] S. Craft, Insulin resistance syndrome and Alzheimer’s disease: age- and obesity- attenuates diabetic nephropathy in type 2 diabetic rats, Biomed. Pharmacother.
related effects on memory, amyloid, and inflammation, Neurobiol. Aging 26 94 (2017) 726–741, https://doi.org/10.1016/j.biopha.2017.07.112.
(Suppl 1) (2005) 65–69, https://doi.org/10.1016/j.neurobiolaging.2005.08.021. [154] D. Das, S. Sarkar, A. Dihingia, N.U. Afzal, S.B. Wann, J. Kalita, S. Dewanjee,
[131] S. Dewanjee, N. Bhattacharjee, MicroRNA: a new generation therapeutic target in P. Manna, A popular fermented soybean food of Northeast India exerted
diabetic nephropathy, Biochem. Pharmacol. 155 (2018) 32–47, https://doi.org/ promising antihyperglycemic potential via stimulating PI3K/AKT/AMPK/GLUT4
10.1016/j.bcp.2018.06.017. signaling pathways and regulating muscle glucose metabolism in type 2 diabetes,
[132] M. Banerjee, M. Saxena, Interleukin-1 (IL-1) family of cytokines: role in type 2 J. Food Biochem. 7 (2022), e14385, https://doi.org/10.1111/jfbc.14385.
diabetes, Clin. Chim. Acta 413 (15–16) (2012) 1163–1170, https://doi.org/ [155] M.P. Guarino, M.J. Ribeiro, J.F. Sacramento, S.V. Conde, Chronic caffeine intake
10.1016/j.cca.2012.03.021. reverses age-induced insulin resistance in the rat: effect on skeletal muscle Glut4
[133] W. Fang, D. Bi, R. Zheng, N. Cai, H. Xu, R. Zhou, J. Lu, M. Wan, X. Xu, transporters and AMPK activity, Age (Dordr) 35 (5) (2013) 1755–1765, https://
Identification and activation of TLR4-mediated signalling pathways by alginate- doi.org/10.1007/s11357-012-9475-x.
derived guluronate oligosaccharide in RAW264.7 macrophages, Sci. Rep. 7 (1) [156] H.R. Zimmermann, W. Yang, N.P. Kasica, X. Zhou, X. Wang, B.C. Beckelman,
(2017) 1663, https://doi.org/10.1038/s41598-017-01868-0. J. Lee, C.M. Furdui, C.D. Keene, T. Ma, Brain-specific repression of AMPKalpha1
[134] N. Kyrtata, H.C.A. Emsley, O. Sparasci, L.M. Parkes, B.R. Dickie, A systematic alleviates pathophysiology in Alzheimer’s model mice, J. Clin. Invest. 130 (7)
review of glucose transport alterations in alzheimer’s disease, Front. Neurosci. 15 (2020) 3511–3527, https://doi.org/10.1172/JCI133982.
(2021), 626636, https://doi.org/10.3389/fnins.2021.626636. [157] X. Wang, X. Zhou, B. Uberseder, J. Lee, C.S. Latimer, C.M. Furdui, C.D. Keene, T.
[135] A. Chadt, H. Al-Hasani, Glucose transporters in adipose tissue, liver, and skeletal J. Montine, T.C. Register, S. Craft, C.A. Shively, T. Ma, Isoform-specific
muscle in metabolic health and disease, Pflügers Archiv 472 (9) (2020) dysregulation of AMP-activated protein kinase signaling in a non-human primate
1273–1298, https://doi.org/10.1007/s00424-020-02417-x. model of Alzheimer’s disease, Neurobiol. Dis. 158 (2021), 105463, https://doi.
[136] C. Iadecola, Sugar and Alzheimer’s disease: a bittersweet truth, Nat. Neurosci. 18 org/10.1016/j.nbd.2021.105463.
(4) (2015) 477–478, https://doi.org/10.1038/nn.3986. [158] Y. Xiao, X. Zheng, G. Li, C. Zhou, C. Wu, Z. Xu, G. Hu, X. Guo, L. Li, H. Cao,
[137] R.J. Mullins, T.C. Diehl, C.W. Chia, D. Kapogiannis, Insulin resistance as a link V. Latigo, P. Liu, Investigation of the effects of dichlorvos poisoning on AMPK
between amyloid-beta and tau pathologies in alzheimer’s disease, Front. Aging signaling pathway in chicken brain tissues, Environ. Pollut. 261 (2020), 114109,
Neurosci. 9 (2017) 118, https://doi.org/10.3389/fnagi.2017.00118. https://doi.org/10.1016/j.envpol.2020.114109.
[138] Y. Deng, B. Li, Y. Liu, K. Iqbal, I. Grundke-Iqbal, C.X. Gong, Dysregulation of [159] S. Ramesh, M. Govindarajulu, T. Lynd, G. Briggs, D. Adamek, E. Jones, J. Heiner,
insulin signaling, glucose transporters, O-GlcNAcylation, and phosphorylation of M. Majrashi, T. Moore, R. Amin, V. Suppiramaniam, M. Dhanasekaran, SIRT3
tau and neurofilaments in the brain: implication for Alzheimer’s disease, Am. J. activator Honokiol attenuates beta-Amyloid by modulating amyloidogenic
Pathol. 175 (5) (2009) 2089–2098, https://doi.org/10.2353/ pathway, PLoS One 13 (1) (2018), e0190350, https://doi.org/10.1371/journal.
ajpath.2009.090157. pone.0190350.
[139] A. Knezovic, A. Loncar, J. Homolak, U. Smailovic, J. Osmanovic Barilar, [160] M. Cui, T. Yoshimori, S. Nakamura, Autophagy system as a potential therapeutic
L. Ganoci, N. Bozina, P. Riederer, M. Salkovic-Petrisic, Rat brain glucose target for neurodegenerative diseases, Neurochem. Int. 155 (2022), 105308,
transporter-2, insulin receptor and glial expression are acute targets of https://doi.org/10.1016/j.neuint.2022.105308.
intracerebroventricular streptozotocin: risk factors for sporadic Alzheimer’s [161] Z. Ou, X. Kong, X. Sun, X. He, L. Zhang, Z. Gong, J. Huang, B. Xu, D. Long, J. Li,
disease? J. Neural. Transm. 124 (6) (2017) 695–708, https://doi.org/10.1007/ Q. Li, L. Xu, A. Xuan, Metformin treatment prevents amyloid plaque deposition
s00702-017-1727-6. and memory impairment in APP/PS1 mice, Brain Behav. Immun. 69 (2018)
[140] R.N. Kalaria, S.I. Harik, Abnormalities of the glucose transporter at the blood- 351–363, https://doi.org/10.1016/j.bbi.2017.12.009.
brain barrier and in brain in Alzheimer’s disease, Prog. Clin. Biol. Res. 317 (1989) [162] Y. Cao, X. Tan, Q. Lu, K. Huang, X. Tang, Z. He, miR-590-3 and SP1 promote
415–421. neuronal apoptosis in patients with alzheimer’s disease via AMPK signaling
[141] K.C. Ahn, C.R. Learman, G.L. Dunbar, P. Maiti, W.C. Jang, H.C. Cha, M.S. Song, pathway, Contrast Media Mol. Imaging 2021 (2021), 6010362, https://doi.org/
Characterization of impaired cerebrovascular structure in APP/PS1 mouse brains, 10.1155/2021/6010362.

151
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

[163] A. Salminen, K. Kaarniranta, AMP-activated protein kinase (AMPK) controls the [186] M.A. DeTure, D.W. Dickson, The neuropathological diagnosis of Alzheimer’s
aging process via an integrated signaling network, Ageing Res. Rev. 11 (2) (2012) disease, Mol. Neurodegener. 14 (1) (2019) 32, https://doi.org/10.1186/s13024-
230–241, https://doi.org/10.1016/j.arr.2011.12.005. 019-0333-5.
[164] E. Barone, F. Di Domenico, M. Perluigi, D.A. Butterfield, The interplay among [187] W. Rungratanawanich, Y. Qu, X. Wang, M.M. Essa, B.J. Song, Advanced glycation
oxidative stress, brain insulin resistance and AMPK dysfunction contribute to end products (AGEs) and other adducts in aging-related diseases and alcohol-
neurodegeneration in type 2 diabetes and Alzheimer disease, Free Radic. Biol. mediated tissue injury, Exp. Mol. Med. 53 (2) (2021) 168–188, https://doi.org/
Med. 176 (2021) 16–33, https://doi.org/10.1016/j.freeradbiomed.2021.09.006. 10.1038/s12276-021-00561-7.
[165] B. Boland, W.H. Yu, O. Corti, B. Mollereau, A. Henriques, E. Bezard, G. [188] N. Barnea-Goraly, M. Raman, P. Mazaika, M. Marzelli, T. Hershey, S.
M. Pastores, D.C. Rubinsztein, R.A. Nixon, M.R. Duchen, G.R. Mallucci, A. Weinzimer, T. Aye, B. Buckingham, N. Mauras, N.H. White, L.A. Fox,
G. Kroemer, B. Levine, E.L. Eskelinen, F. Mochel, M. Spedding, C. Louis, O. M. Tansey, R.W. Beck, K.J. Ruedy, C. Kollman, P. Cheng, A.L. Reiss, N. Diabetes
R. Martin, M.J. Millan, Promoting the clearance of neurotoxic proteins in Research in Children, Alterations in white matter structure in young children with
neurodegenerative disorders of ageing, Nat. Rev. Drug Discov. 17 (9) (2018) type 1 diabetes, Diabetes Care 37 (2) (2014) 332–340, https://doi.org/10.2337/
660–688, https://doi.org/10.1038/nrd.2018.109. dc13-1388.
[166] Y. Wang, J. Lv, J. He, G. Wen, X. Wu, Mechanism of psychoactive substance- [189] P. Mergenthaler, U. Lindauer, G.A. Dienel, A. Meisel, Sugar for the brain: the role
induced cognitive disorders: does tau protein play a role? Front Biosci (Landmark of glucose in physiological and pathological brain function, Trends Neurosci. 36
Ed) 27 (1) (2022) 6, https://doi.org/10.31083/j.fbl2701006. (10) (2013) 587–597, https://doi.org/10.1016/j.tins.2013.07.001.
[167] A. Sleigh, P. Raymond-Barker, K. Thackray, D. Porter, M. Hatunic, A. Vottero, [190] M. Takeuchi, A. Sakasai-Sakai, T. Takata, J.I. Takino, Y. Koriyama, C. Kikuchi,
C. Burren, C. Mitchell, M. McIntyre, S. Brage, T.A. Carpenter, P.R. Murgatroyd, K. A. Furukawa, K. Nagamine, T. Hori, T. Matsunaga, Intracellular toxic AGEs
M. Brindle, G.J. Kemp, S. O’Rahilly, R.K. Semple, D.B. Savage, Mitochondrial (TAGE) triggers numerous types of cell damage, Biomolecules 11 (3) (2021) 387,
dysfunction in patients with primary congenital insulin resistance, J. Clin. Invest. https://doi.org/10.3390/biom11030387.
121 (6) (2011) 2457–2461, https://doi.org/10.1172/JCI46405. [191] K.V. Ramana, Aldose reductase: new insights for an old enzyme, Biomol. Concepts
[168] N. Abolhassani, J. Leon, Z. Sheng, S. Oka, H. Hamasaki, T. Iwaki, Y. Nakabeppu, 2 (1–2) (2011) 103–114, https://doi.org/10.1515/BMC.2011.002.
Molecular pathophysiology of impaired glucose metabolism, mitochondrial [192] N. Niimi, H. Yako, S. Takaku, S.K. Chung, K. Sango, Aldose reductase and the
dysfunction, and oxidative DNA damage in Alzheimer’s disease brain, Mech. polyol pathway in schwann cells: old and new problems, Int. J. Mol. Sci. 22 (3)
Ageing Dev. 161 (Pt A) (2017) 95–104, https://doi.org/10.1016/j. (2021) 1031, https://doi.org/10.3390/ijms22031031.
mad.2016.05.005. [193] T. Sirota, N. Sirota, On the mechanism of oxygen activation in chemical and
[169] A. Kleinridders, W. Cai, L. Cappellucci, A. Ghazarian, W.R. Collins, S.G. Vienberg, biological systems, Biophysics 67 (1) (2022) 1–7, https://doi.org/10.1134/
E.N. Pothos, C.R. Kahn, Insulin resistance in brain alters dopamine turnover and S000635092201016X.
causes behavioral disorders, Proc. Natl. Acad. Sci. U. S. A. 112 (11) (2015) [194] P.J. Yao, E. Eren, E.J. Goetzl, D. Kapogiannis, Mitochondrial electron transport
3463–3468, https://doi.org/10.1073/pnas.1500877112. chain protein abnormalities detected in plasma extracellular vesicles in
[170] Z. Chen, C. Zhong, Decoding Alzheimer’s disease from perturbed cerebral glucose alzheimer’s disease, Biomedicines 9 (11) (2021), https://doi.org/10.3390/
metabolism: implications for diagnostic and therapeutic strategies, Prog. biomedicines9111587.
Neurobiol. 108 (2013) 21–43, https://doi.org/10.1016/j.pneurobio.2013.06.004. [195] X. Wang, W. Wang, L. Li, G. Perry, H.G. Lee, X. Zhu, Oxidative stress and
[171] L. Mosconi, J. Murray, W.H. Tsui, Y. Li, N. Spector, A. Goldowsky, S. Williams, mitochondrial dysfunction in Alzheimer’s disease, Biochim. Biophys. Acta 1842
R. Osorio, P. McHugh, L. Glodzik, S. Vallabhajosula, M.J. de Leon, Brain imaging (8) (2014) 1240–1247, https://doi.org/10.1016/j.bbadis.2013.10.015.
of cognitively normal individuals with 2 parents affected by late-onset AD, [196] M. Naziroglu, S. Muhamad, L. Pecze, Nanoparticles as potential clinical
Neurology 82 (9) (2014) 752–760, https://doi.org/10.1212/ therapeutic agents in Alzheimer’s disease: focus on selenium nanoparticles, Expet
WNL.0000000000000181. Rev. Clin. Pharmacol. 10 (7) (2017) 773–782, https://doi.org/10.1080/
[172] G. Vial, D. Detaille, B. Guigas, Role of mitochondria in the mechanism (s) of 17512433.2017.1324781.
action of metformin, Front. Endocrinol. 10 (2019) 294, https://doi.org/10.3389/ [197] K. Cao, Y.T. Dong, J. Xiang, Y. Xu, W. Hong, H. Song, Z.Z. Guan, Reduced
fendo.2019.00294. expression of SIRT1 and SOD-1 and the correlation between these levels in
[173] M.C. Wingard, C.R. Frasier, M. Singh, K. Singh, Heart failure and diabetes: role of various regions of the brains of patients with Alzheimer’s disease, J. Clin. Pathol.
ATM, Curr. Opin. Pharmacol. 54 (2020) 27–35, https://doi.org/10.1016/j. 71 (12) (2018) 1090–1099, https://doi.org/10.1136/jclinpath-2018-205320.
coph.2020.06.007. [198] P. Youssef, B. Chami, J. Lim, T. Middleton, G.T. Sutherland, P.K. Witting,
[174] C.H. Wang, T.F. Tsai, Y.H. Wei, Role of mitochondrial dysfunction and Evidence supporting oxidative stress in a moderately affected area of the brain in
dysregulation of Ca(2+) homeostasis in insulin insensitivity of mammalian cells, Alzheimer’s disease, Sci. Rep. 8 (1) (2018), 11553, https://doi.org/10.1038/
Ann. N. Y. Acad. Sci. 1350 (2015) 66–76, https://doi.org/10.1111/nyas.12838. s41598-018-29770-3.
[175] S. Dewanjee, S. Das, A.K. Das, N. Bhattacharjee, A. Dihingia, T.K. Dua, J. Kalita, [199] A.A. Vilcaes, N.L. Chanaday, E.T. Kavalali, Interneuronal exchange and functional
P. Manna, Molecular mechanism of diabetic neuropathy and its integration of synaptobrevin via extracellular vesicles, Neuron 109 (6) (2021)
pharmacotherapeutic targets, Eur. J. Pharmacol. 833 (2018) 472–523, https:// 971–983, https://doi.org/10.1016/j.neuron.2021.01.007, e5.
doi.org/10.1016/j.ejphar.2018.06.034. [200] R.H. Swerdlow, Mitochondria and mitochondrial cascades in alzheimer’s disease,
[176] J. Satoh, Y. Yamamoto, N. Asahina, S. Kitano, Y. Kino, RNA-Seq data mining: J Alzheimers Dis 62 (3) (2018) 1403–1416, https://doi.org/10.3233/JAD-
downregulation of NeuroD6 serves as a possible biomarker for alzheimer’s 170585.
disease brains, Dis. Markers 2014 (2014), 123165, https://doi.org/10.1155/ [201] S.S. Adav, J.E. Park, S.K. Sze, Quantitative profiling brain proteomes revealed
2014/123165. mitochondrial dysfunction in Alzheimer’s disease, Mol. Brain 12 (1) (2019) 8,
[177] H.C. Kung, K.J. Lin, C.T. Kung, T.K. Lin, Oxidative stress, mitochondrial https://doi.org/10.1186/s13041-019-0430-y.
dysfunction, and neuroprotection of polyphenols with respect to resveratrol in [202] K. Hirai, G. Aliev, A. Nunomura, H. Fujioka, R.L. Russell, C.S. Atwood, A.
Parkinson’s disease, Biomedicines 9 (8) (2021) 918, https://doi.org/10.3390/ B. Johnson, Y. Kress, H.V. Vinters, M. Tabaton, S. Shimohama, A.D. Cash, S.
biomedicines9080918. L. Siedlak, P.L. Harris, P.K. Jones, R.B. Petersen, G. Perry, M.A. Smith,
[178] N. Bhattacharjee, S. Barma, N. Konwar, S. Dewanjee, P. Manna, Mechanistic Mitochondrial abnormalities in Alzheimer’s disease, J. Neurosci. 21 (9) (2001)
insight of diabetic nephropathy and its pharmacotherapeutic targets: an update, 3017–3023, https://doi.org/10.1523/JNEUROSCI.21-09-03017.2001.
Eur. J. Pharmacol. 791 (2016) 8–24, https://doi.org/10.1016/j. [203] M. Kriebel, J. Ebel, F. Battke, S. Griesbach, H. Volkmer, Interference with complex
ejphar.2016.08.022. IV as a model of age-related decline in synaptic connectivity, Front. Mol.
[179] A.E. Stinghen, Z.A. Massy, H. Vlassara, G.E. Striker, A. Boullier, Uremic toxicity of Neurosci. 13 (2020) 43, https://doi.org/10.3389/fnmol.2020.00043.
advanced glycation end products in CKD, J. Am. Soc. Nephrol. 27 (2) (2016) [204] D. Prokopenko, S.L. Morgan, K. Mullin, O. Hofmann, B. Chapman, R. Kirchner,
354–370, https://doi.org/10.1681/ASN.2014101047. I. Alzheimer’s Disease Neuroimaging, S. Amberkar, I. Wohlers, C. Lange, W. Hide,
[180] B. Wu, L. Yu, P. Hu, Y. Lu, J. Li, Y. Wei, R. He, GRP78 protects CHO cells from L. Bertram, R.E. Tanzi, Whole-genome sequencing reveals new Alzheimer’s
ribosylation, Biochim. Biophys. Acta Mol. Cell Res. 1865 (4) (2018) 629–637, disease-associated rare variants in loci related to synaptic function and neuronal
https://doi.org/10.1016/j.bbamcr.2018.02.001. development, Alzheimers Dement 17 (9) (2021) 1509–1527, https://doi.org/
[181] R. Mastrocola, D. Nigro, A.S. Cento, F. Chiazza, M. Collino, M. Aragno, High- 10.1002/alz.12319.
fructose intake as risk factor for neurodegeneration: key role for carboxy [205] B.G. Trist, D.J. Hare, K.L. Double, Oxidative stress in the aging substantia nigra
methyllysine accumulation in mice hippocampal neurons, Neurobiol. Dis. 89 and the etiology of Parkinson’s disease, Aging Cell 18 (6) (2019), e13031,
(2016) 65–75, https://doi.org/10.1016/j.nbd.2016.02.005. https://doi.org/10.1111/acel.13031.
[182] A. Gugliucci, formation of fructose-mediated advanced glycation end products [206] E. Llanos-Gonzalez, A.A. Henares-Chavarino, C.M. Pedrero-Prieto, S. Garcia-
and their roles in metabolic and inflammatory diseases, Adv. Nutr. 8 (1) (2017) Carpintero, J. Frontinan-Rubio, F.J. Sancho-Bielsa, F.J. Alcain, J.R. Peinado,
54–62, https://doi.org/10.3945/an.116.013912. Y. Rabanal-Ruiz, M. Duran-Prado, Interplay between mitochondrial oxidative
[183] D.N. Hauser, A. Mamais, M.M. Conti, C.T. Primiani, R. Kumaran, A.A. Dillman, R. disorders and proteostasis in alzheimer’s disease, Front. Neurosci. 13 (2019)
G. Langston, A. Beilina, J.H. Garcia, A. Diaz-Ruiz, M. Bernier, F.C. Fiesel, X. Hou, 1444, https://doi.org/10.3389/fnins.2019.01444.
W. Springer, Y. Li, R. de Cabo, M.R. Cookson, Hexokinases link DJ-1 to the [207] Y. Yamazaki, S. Fujii, Extracellular ATP modulates synaptic plasticity induced by
PINK1/parkin pathway, Mol. Neurodegener. 12 (1) (2017) 70, https://doi.org/ activation of metabotropic glutamate receptors in the hippocampus, Biomed. Res.
10.1186/s13024-017-0212-x. 36 (1) (2015) 1–9, https://doi.org/10.2220/biomedres.36.1.
[184] A. Kuzan, Toxicity of advanced glycation end products, Biomed Rep 14 (5) (2021) [208] M.D. Sweeney, A.P. Sagare, B.V. Zlokovic, Blood-brain barrier breakdown in
46, https://doi.org/10.3892/br.2021.1422. Alzheimer disease and other neurodegenerative disorders, Nat. Rev. Neurol. 14
[185] M. Takeuchi, J.I. Takino, A. Sakasai-Sakai, T. Takata, M. Tsutsumi, Toxic AGE (3) (2018) 133–150, https://doi.org/10.1038/nrneurol.2017.188.
(TAGE) theory for the pathophysiology of the onset/progression of NAFLD and
ALD, Nutrients 9 (6) (2017) 634, https://doi.org/10.3390/nu9060634.

152
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

[209] G. Lou, K. Palikaras, S. Lautrup, M. Scheibye-Knudsen, N. Tavernarakis, E.F. Fang, [231] K.R. Valasani, Q. Sun, D. Fang, Z. Zhang, Q. Yu, Y. Guo, J. Li, A. Roy, S. ShiDu
Mitophagy and neuroprotection, Trends Mol. Med. 26 (1) (2020) 8–20, https:// Yan, Identification of a small molecule cyclophilin D inhibitor for rescuing abeta-
doi.org/10.1016/j.molmed.2019.07.002. mediated mitochondrial dysfunction, ACS Med. Chem. Lett. 7 (3) (2016)
[210] O. Pansarasa, M. Bordoni, L. Diamanti, D. Sproviero, S. Gagliardi, C. Cereda, 294–299, https://doi.org/10.1021/acsmedchemlett.5b00451.
SOD1 in amyotrophic lateral sclerosis: "ambivalent" behavior connected to the [232] M.P. Kummer, M.T. Heneka, Truncated and modified amyloid-beta species,
disease, Int. J. Mol. Sci. 19 (5) (2018) 1345, https://doi.org/10.3390/ Alzheimer’s Res. Ther. 6 (3) (2014) 28, https://doi.org/10.1186/alzrt258.
ijms19051345. [233] M. Panel, B. Ghaleh, D. Morin, Mitochondria and aging: a role for the
[211] R.A. Cavallaro, V. Nicolia, M.T. Fiorenza, S. Scarpa, A. Fuso, S- mitochondrial transition pore? Aging Cell 17 (4) (2018), e12793 https://doi.org/
adenosylmethionine and superoxide dismutase 1 synergistically counteract 10.1111/acel.12793.
alzheimer’s disease features progression in TgCRND8 mice, Antioxidants 6 (4) [234] S.J. Beck, L. Guo, A. Phensy, J. Tian, L. Wang, N. Tandon, E. Gauba, L. Lu, J.
(2017) 76, https://doi.org/10.3390/antiox6040076. M. Pascual, S. Kroener, H. Du, Deregulation of mitochondrial F1FO-ATP synthase
[212] R. Sahu, T.K. Dua, S. Das, V. De Feo, S. Dewanjee, Wheat phenolics suppress via OSCP in Alzheimer’s disease, Nat. Commun. 7 (2016), 11483, https://doi.org/
doxorubicin-induced cardiotoxicity via inhibition of oxidative stress, MAP kinase 10.1038/ncomms11483.
activation, NF-κB pathway, PI3K/Akt/mTOR impairment, and cardiac apoptosis, [235] V.S. Sukhorukov, N.M. Mudzhiri, A.S. Voronkova, T.I. Baranich, V.V. Glinkina, S.
Food Chem. Toxicol. 125 (2019) 503–519, https://doi.org/10.1016/j. N. Illarioshkin, Mitochondrial disorders in alzheimer’s disease, Biochemistry
fct.2019.01.034. (Mosc.) 86 (6) (2021) 667–679, https://doi.org/10.1134/S0006297921060055.
[213] T.K. Dua, S. Joardar, P. Chakraborty, S. Bhowmick, A. Saha, V. De Feo, [236] E.E. Congdon, E.M. Sigurdsson, Tau-targeting therapies for Alzheimer disease,
S. Dewanjee, Myricitrin, a glycosyloxyflavone in myrica esculenta bark Nat. Rev. Neurol. 14 (7) (2018) 399–415, https://doi.org/10.1038/s41582-018-
ameliorates diabetic nephropathy via improving glycemic status, reducing 0013-z.
oxidative stress, and suppressing inflammation, Molecules 26 (6) (2021) 258, [237] D.C. David, S. Hauptmann, I. Scherping, K. Schuessel, U. Keil, P. Rizzu, R. Ravid,
https://doi.org/10.3390/molecules26020258. S. Drose, U. Brandt, W.E. Muller, A. Eckert, J. Gotz, Proteomic and functional
[214] S. Das, S. Dewanjee, T.K. Dua, S. Joardar, P. Chakraborty, S. Bhowmick, A. Saha, analyses reveal a mitochondrial dysfunction in P301L tau transgenic mice, J. Biol.
S. Bhattacharjee, V. De Feo, Carnosic acid attenuates cadmium induced Chem. 280 (25) (2005) 23802–23814, https://doi.org/10.1074/jbc.
nephrotoxicity by inhibiting oxidative stress, promoting Nrf2/HO-1 signalling M500356200.
and impairing TGF-β1/smad/collagen IV signalling, Molecules 24 (22) (2019) [238] G. Amadoro, V. Corsetti, A. Atlante, F. Florenzano, S. Capsoni, R. Bussani,
4176, https://doi.org/10.3390/molecules24224176. D. Mercanti, P. Calissano, Interaction between NH(2)-tau fragment and Abeta in
[215] V.K. Rao, E.A. Carlson, S.S. Yan, Mitochondrial permeability transition pore is a Alzheimer’s disease mitochondria contributes to the synaptic deterioration,
potential drug target for neurodegeneration, Biochim. Biophys. Acta 1842 (8) Neurobiol. Aging 33 (4) (2012) 833 e1–e25, https://doi.org/10.1016/j.
(2014) 1267–1272, https://doi.org/10.1016/j.bbadis.2013.09.003. neurobiolaging.2011.08.001.
[216] M. Bonora, P. Pinton, A new current for the mitochondrial permeability [239] C. Jara, A. Aranguiz, W. Cerpa, C. Tapia-Rojas, R.A. Quintanilla, Genetic ablation
transition, Trends Biochem. Sci. 44 (7) (2019) 559–561, https://doi.org/ of tau improves mitochondrial function and cognitive abilities in the
10.1016/j.tibs.2019.04.009. hippocampus, Redox Biol. 18 (2018) 279–294, https://doi.org/10.1016/j.
[217] K. Jia, H. Du, Mitochondrial permeability transition: a pore intertwines brain redox.2018.07.010.
aging and alzheimer’s disease, Cells 10 (3) (2021) 649, https://doi.org/10.3390/ [240] B. Xiao, J.Y. Goh, L. Xiao, H. Xian, K.L. Lim, Y.C. Liou, Reactive oxygen species
cells10030649. trigger Parkin/PINK1 pathway-dependent mitophagy by inducing mitochondrial
[218] S. Joardar, S. Dewanjee, S. Bhowmick, T.K. Dua, S. Das, A. Saha, V. De Feo, recruitment of Parkin, J. Biol. Chem. 292 (40) (2017) 16697–16708, https://doi.
Rosmarinic acid attenuates cadmium-induced nephrotoxicity via inhibition of org/10.1074/jbc.M117.787739.
oxidative stress, apoptosis, inflammation and fibrosis, Int. J. Mol. Sci. 20 (8) [241] H. Murali Mahadevan, A. Hashemiaghdam, G. Ashrafi, A.B. Harbauer,
(2019 2027), https://doi.org/10.3390/ijms20082027. Mitochondria in neuronal health: from energy metabolism to Parkinson’s disease,
[219] R. Peruzzo, I. Szabo, Contribution of mitochondrial ion channels to chemo- Adv Biol (Weinh) 5 (9) (2021), e2100663, https://doi.org/10.1002/
resistance in cancer cells, Cancers 11 (6) (2019) 761, https://doi.org/10.3390/ adbi.202100663.
cancers11060761. [242] S.C. Cunnane, E. Trushina, C. Morland, A. Prigione, G. Casadesus, Z.B. Andrews,
[220] B.M. Dunyak, J.E. Gestwicki, Peptidyl-proline isomerases (PPIases): targets for M.F. Beal, L.H. Bergersen, R.D. Brinton, S. de la Monte, A. Eckert, J. Harvey,
natural products and natural product-inspired compounds, J. Med. Chem. 59 (21) R. Jeggo, J.H. Jhamandas, O. Kann, C.M. la Cour, W.F. Martin, G. Mithieux, P.
(2016) 9622–9644, https://doi.org/10.1021/acs.jmedchem.6b00411. I. Moreira, M.P. Murphy, K.A. Nave, T. Nuriel, S.H.R. Oliet, F. Saudou, M.
[221] W. Lin, M. Bonin, A. Boden, R. Wieduwild, P. Murawala, M. Wermke, H. Andrade, P. Mattson, R.H. Swerdlow, M.J. Millan, Brain energy rescue: an emerging
M. Bornhauser, Y. Zhang, Peptidyl prolyl cis/trans isomerase activity on the cell therapeutic concept for neurodegenerative disorders of ageing, Nat. Rev. Drug
surface correlates with extracellular matrix development, Commun Biol 2 (2019) Discov. 19 (9) (2020) 609–633, https://doi.org/10.1038/s41573-020-0072-x.
58, https://doi.org/10.1038/s42003-019-0315-8. [243] G. Amadoro, V. Corsetti, F. Florenzano, A. Atlante, M.T. Ciotti, M.P. Mongiardi,
[222] S. Dutta, P. Chakraborty, S. Basak, S. Ghosh, N. Ghosh, S. Chatterjee, S. Dewanjee, R. Bussani, V. Nicolin, S.L. Nori, M. Campanella, P. Calissano, AD-linked, toxic
P.C. Sil, Synthesis, characterization, and evaluation of in vitro cytotoxicity and in NH2 human tau affects the quality control of mitochondria in neurons, Neurobiol.
vivo antitumor activity of asiatic acid-loaded poly lactic-co-glycolic acid Dis. 62 (2014) 489–507, https://doi.org/10.1016/j.nbd.2013.10.018.
nanoparticles: a strategy of treating breast cancer, Life Sci. 307 (2022), 120876, [244] L. Barazzuol, F. Giamogante, M. Brini, T. Cali, PINK1/Parkin mediated
https://doi.org/10.1016/j.lfs.2022.120876. mitophagy, Ca(2+) signalling, and ER-mitochondria contacts in Parkinson’s
[223] C. Penna, M.G. Perrelli, P. Pagliaro, Mitochondrial pathways, permeability disease, Int. J. Mol. Sci. 21 (5) (2020), https://doi.org/10.3390/ijms21051772.
transition pore, and redox signaling in cardioprotection: therapeutic implications, [245] C.A. Gautier, T. Kitada, J. Shen, Loss of PINK1 causes mitochondrial functional
Antioxidants Redox Signal. 18 (5) (2013) 556–599, https://doi.org/10.1089/ defects and increased sensitivity to oxidative stress, Proc. Natl. Acad. Sci. U. S. A.
ars.2011.4459. 105 (32) (2008) 11364–11369, https://doi.org/10.1073/pnas.0802076105.
[224] S. Chakraborty, Z.Y. Dlie, S. Chakraborty, S. Roy, B. Mukherjee, B.E. Besra, [246] G. Arena, E.M. Valente, PINK1 in the limelight: multiple functions of an eclectic
S. Dewanjee, A. Mukherjee, P.K. Ojha, V. Kumar, R. Sen, Aptamer-functionalized protein in human health and disease, J. Pathol. 241 (2) (2017) 251–263, https://
drug nanocarrier improves hepatocellular carcinoma toward normal by targeting doi.org/10.1002/path.4815.
neoplastic hepatocytes, Mol. Ther. Nucleic Acids 20 (2020) 34–49, https://doi. [247] R.J. Youle, A.M. van der Bliek, Mitochondrial fission, fusion, and stress, Science
org/10.1016/j.omtn.2020.01.034. 337 (6098) (2012) 1062–1065, https://doi.org/10.1126/science.1219855.
[225] H. Du, L. Guo, F. Fang, D. Chen, A.A. Sosunov, G.M. McKhann, Y. Yan, C. Wang, [248] K. Panchal, A.K. Tiwari, Mitochondrial dynamics, a key executioner in
H. Zhang, J.D. Molkentin, F.J. Gunn-Moore, J.P. Vonsattel, O. Arancio, J.X. Chen, neurodegenerative diseases, Mitochondrion 47 (2019) 151–173, https://doi.org/
S.D. Yan, Cyclophilin D deficiency attenuates mitochondrial and neuronal 10.1016/j.mito.2018.11.002.
perturbation and ameliorates learning and memory in Alzheimer’s disease, Nat [249] M.K. McCoy, A. Kaganovich, I.N. Rudenko, J. Ding, M.R. Cookson, Hexokinase
Med 14 (10) (2008) 1097–1105, https://doi.org/10.1038/nm.1868. activity is required for recruitment of parkin to depolarized mitochondria, Hum.
[226] E. Gauba, L. Guo, H. Du, Cyclophilin D promotes brain mitochondrial F1FO ATP Mol. Genet. 23 (1) (2014) 145–156, https://doi.org/10.1093/hmg/ddt407.
synthase dysfunction in aging mice, J Alzheimers Dis 55 (4) (2017) 1351–1362, [250] K. Banerjee, S. Munshi, D.E. Frank, G.E. Gibson, Abnormal glucose metabolism in
https://doi.org/10.3233/JAD-160822. alzheimer’s disease: relation to autophagy/mitophagy and therapeutic
[227] B.C. Lynn, J. Wang, W.R. Markesbery, M.A. Lovell, Quantitative changes in the approaches, Neurochem. Res. 40 (12) (2015) 2557–2569, https://doi.org/
mitochondrial proteome from subjects with mild cognitive impairment, early 10.1007/s11064-015-1631-0.
stage, and late stage Alzheimer’s disease, J Alzheimers Dis 19 (1) (2010) [251] C. Ploumi, I. Daskalaki, N. Tavernarakis, Mitochondrial biogenesis and clearance:
325–339, https://doi.org/10.3233/JAD-2010-1254. a balancing act, FEBS J. 284 (2) (2017) 183–195.
[228] H. Du, L. Guo, W. Zhang, M. Rydzewska, S. Yan, Cyclophilin D deficiency [252] K.A. Boyle, J. Van Wickle, R.B. Hill, A. Marchese, B. Kalyanaraman, M.B. Dwinell,
improves mitochondrial function and learning/memory in aging Alzheimer Mitochondria-targeted drugs stimulate mitophagy and abrogate colon cancer cell
disease mouse model, Neurobiol. Aging 32 (3) (2011) 398–406, https://doi.org/ proliferation, J. Biol. Chem. 293 (38) (2018) 14891–14904, https://doi.org/
10.1016/j.neurobiolaging.2009.03.003. 10.1074/jbc.RA117.001469.
[229] E. Gauba, H. Chen, L. Guo, H. Du, Cyclophilin D deficiency attenuates [253] N. Bhattacharjee, T.K. Dua, R. Khanra, S. Joardar, A. Nandy, A. Saha, V. De Feo,
mitochondrial F1Fo ATP synthase dysfunction via OSCP in Alzheimer’s disease, S. Dewanjee, Protocatechuic acid, a phenolic from sansevieria roxburghiana
Neurobiol. Dis. 121 (2019) 138–147, https://doi.org/10.1016/j. leaves, suppresses diabetic cardiomyopathy via stimulating glucose metabolism,
nbd.2018.09.020. ameliorating oxidative stress, and inhibiting inflammation, Front. Pharmacol. 8
[230] L. Guo, H. Du, S. Yan, X. Wu, G.M. McKhann, J.X. Chen, S.S. Yan, Cyclophilin D (2017) 251, https://doi.org/10.3389/fphar.2017.00251.
deficiency rescues axonal mitochondrial transport in Alzheimer’s neurons, PLoS [254] S. Brun, N. Schall, S.R. Bonam, K. Bigaut, A.G. Mensah-Nyagan, J. de Seze,
One 8 (1) (2013), e54914, https://doi.org/10.1371/journal.pone.0054914. S. Muller, An autophagy-targeting peptide to treat chronic inflammatory

153
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

demyelinating polyneuropathies, J. Autoimmun. 92 (2018) 114–125, https://doi. transfectants with induced wild-type tau expression, Eur. J. Neurosci. 27 (5)
org/10.1016/j.jaut.2018.05.009. (2008) 1119–1130, https://doi.org/10.1111/j.1460-9568.2008.06084.x.
[255] R. Sanchez-Varo, L. Trujillo-Estrada, E. Sanchez-Mejias, M. Torres, D. Baglietto- [274] A. Piras, L. Collin, F. Gruninger, C. Graff, A. Ronnback, Autophagic and lysosomal
Vargas, I. Moreno-Gonzalez, V. De Castro, S. Jimenez, D. Ruano, M. Vizuete, J. defects in human tauopathies: analysis of post-mortem brain from patients with
C. Davila, J.M. Garcia-Verdugo, A.J. Jimenez, J. Vitorica, A. Gutierrez, Abnormal familial Alzheimer disease, corticobasal degeneration and progressive
accumulation of autophagic vesicles correlates with axonal and synaptic supranuclear palsy, Acta Neuropathol Commun 4 (2016) 22, https://doi.org/
pathology in young Alzheimer’s mice hippocampus, Acta Neuropathol. 123 (1) 10.1186/s40478-016-0292-9.
(2012) 53–70, https://doi.org/10.1007/s00401-011-0896-x. [275] J.L. Guo, A. Buist, A. Soares, K. Callaerts, S. Calafate, F. Stevenaert, J.P. Daniels,
[256] W.A. Eimer, R. Vassar, Neuron loss in the 5XFAD mouse model of Alzheimer’s B.E. Zoll, A. Crowe, K.R. Brunden, D. Moechars, V.M. Lee, The dynamics and
disease correlates with intraneuronal Abeta42 accumulation and Caspase-3 turnover of tau aggregates in cultured cells: insights into therapies for
activation, Mol. Neurodegener. 8 (2013) 2, https://doi.org/10.1186/1750-1326- tauopathies, J. Biol. Chem. 291 (25) (2016) 13175–13193, https://doi.org/
8-2. 10.1074/jbc.M115.712083.
[257] G. Gontier, C. George, Z. Chaker, M. Holzenberger, S. Aid, Blocking IGF signaling [276] K. Inoue, J. Rispoli, H. Kaphzan, E. Klann, E.I. Chen, J. Kim, M. Komatsu,
in adult neurons alleviates alzheimer’s disease pathology through amyloid-beta A. Abeliovich, Macroautophagy deficiency mediates age-dependent
clearance, J. Neurosci. 35 (33) (2015) 11500–11513, https://doi.org/10.1523/ neurodegeneration through a phospho-tau pathway, Mol. Neurodegener. 7 (2012)
JNEUROSCI.0343-15.2015. 48, https://doi.org/10.1186/1750-1326-7-48.
[258] V. Lachance, Q. Wang, E. Sweet, I. Choi, C.Z. Cai, X.X. Zhuang, Y. Zhang, J. [277] A. Caccamo, S. Majumder, A. Richardson, R. Strong, S. Oddo, Molecular interplay
L. Jiang, R.D. Blitzer, O. Bozdagi-Gunal, B. Zhang, J.H. Lu, Z. Yue, Autophagy between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects
protein NRBF2 has reduced expression in Alzheimer’s brains and modulates on cognitive impairments, J. Biol. Chem. 285 (17) (2010) 13107–13120, https://
memory and amyloid-beta homeostasis in mice, Mol. Neurodegener. 14 (1) doi.org/10.1074/jbc.M110.100420.
(2019) 43, https://doi.org/10.1186/s13024-019-0342-4. [278] S. Majumder, A. Richardson, R. Strong, S. Oddo, Inducing autophagy by
[259] B.L. Heckmann, B.J.W. Teubner, E. Boada-Romero, B. Tummers, C. Guy, rapamycin before, but not after, the formation of plaques and tangles ameliorates
P. Fitzgerald, U. Mayer, S. Carding, S.S. Zakharenko, T. Wileman, D.R. Green, cognitive deficits, PLoS One 6 (9) (2011), e25416, https://doi.org/10.1371/
Noncanonical function of an autophagy protein prevents spontaneous journal.pone.0025416.
Alzheimer’s disease, Sci. Adv. 6 (33) (2020), eabb9036, https://doi.org/10.1126/ [279] R.A. Nixon, Amyloid precursor protein and endosomal-lysosomal dysfunction in
sciadv.abb9036. Alzheimer’s disease: inseparable partners in a multifactorial disease, Faseb. J. 31
[260] J.A. Pradeepkiran, P.H. Reddy, Defective mitophagy in Alzheimer’s disease, (7) (2017) 2729–2743, https://doi.org/10.1096/fj.201700359.
Ageing Res. Rev. 64 (2020), 101191, https://doi.org/10.1016/j. [280] A. Caccamo, A. Magri, D.X. Medina, E.V. Wisely, M.F. Lopez-Aranda, A.J. Silva,
arr.2020.101191. S. Oddo, mTOR regulates tau phosphorylation and degradation: implications for
[261] D.J. Selkoe, J. Hardy, The amyloid hypothesis of Alzheimer’s disease at 25 years, Alzheimer’s disease and other tauopathies, Aging Cell 12 (3) (2013) 370–380,
EMBO Mol. Med. 8 (6) (2016) 595–608, https://doi.org/10.15252/ https://doi.org/10.1111/acel.12057.
emmm.201606210. [281] V. Schaeffer, M. Goedert, Stimulation of autophagy is neuroprotective in a mouse
[262] Z. Zhang, X. Yang, Y.Q. Song, J. Tu, Autophagy in Alzheimer’s disease model of human tauopathy, Autophagy 8 (11) (2012) 1686–1687, https://doi.
pathogenesis: therapeutic potential and future perspectives, Ageing Res. Rev. 72 org/10.4161/auto.21488.
(2021), 101464, https://doi.org/10.1016/j.arr.2021.101464. [282] M.C. Silva, G.A. Nandi, S. Tentarelli, I.K. Gurrell, T. Jamier, D. Lucente, B.
[263] P. Nilsson, K. Loganathan, M. Sekiguchi, Y. Matsuba, K. Hui, S. Tsubuki, C. Dickerson, D.G. Brown, N.J. Brandon, S.J. Haggarty, Prolonged tau clearance
M. Tanaka, N. Iwata, T. Saito, T.C. Saido, Aβ secretion and olig formation depend and stress vulnerability rescue by pharmacological activation of autophagy in
on autophagy, Cell Rep. 5 (1) (2013) 61–69, https://doi.org/10.1016/j. tauopathy neurons, Nat. Commun. 11 (1) (2020) 3258, https://doi.org/10.1038/
celrep.2013.08.042. s41467-020-16984-1.
[264] P. Nilsson, M. Sekiguchi, T. Akagi, S. Izumi, T. Komori, K. Hui, K. Sorgjerd, [283] H. Sun, Y. Zhong, X. Zhu, H. Liao, J. Lee, Y. Chen, L. Ma, J. Ren, M. Zhao, M. Tu,
M. Tanaka, T. Saito, N. Iwata, T.C. Saido, Autophagy-related protein 7 deficiency F. Li, H. Zhang, M. Tian, D. Ling, A tauopathy-homing and autophagy-activating
in amyloid beta (Abeta) precursor protein transgenic mice decreases Abeta in the nanoassembly for specific clearance of pathogenic tau in alzheimer’s disease, ACS
multivesicular bodies and induces Abeta accumulation in the Golgi, Am. J. Pathol. Nano 15 (3) (2021) 5263–5275, https://doi.org/10.1021/acsnano.0c10690.
185 (2) (2015) 305–313, https://doi.org/10.1016/j.ajpath.2014.10.011. [284] S. Kim, D. Lee, J.C. Song, S.J. Cho, S.M. Yun, Y.H. Koh, J. Song, G.V. Johnson,
[265] J.J. Collier, C. Guissart, M. Olahova, S. Sasorith, F. Piron-Prunier, F. Suomi, C. Jo, NDP52 associates with phosphorylated tau in brains of an Alzheimer
D. Zhang, N. Martinez-Lopez, N. Leboucq, A. Bahr, S. Azzarello-Burri, S. Reich, disease mouse model, Biochem. Biophys. Res. Commun. 454 (1) (2014) 196–201,
L. Schols, T.M. Polvikoski, P. Meyer, L. Larrieu, A.M. Schaefer, H.S. Alsaif, https://doi.org/10.1016/j.bbrc.2014.10.066.
S. Alyamani, S. Zuchner, I.A. Barbosa, C. Deshpande, A. Pyle, A. Rauch, [285] S. Malampati, J.-X. Song, B. Chun-Kit Tong, A. Nalluri, C.-B. Yang, Z. Wang,
M. Synofzik, F.S. Alkuraya, F. Rivier, M. Ryten, R. McFarland, A. Delahodde, T. S. Gopalkrishnashetty Sreenivasmurthy, Z. Zhu, J. Liu, C. Su, Targeting
G. McWilliams, M. Koenig, R.W. Taylor, Developmental consequences of defective aggrephagy for the treatment of Alzheimer’s disease, Cells 9 (2) (2020) 311,
ATG7-mediated autophagy in humans, N. Engl. J. Med. 384 (25) (2021) https://doi.org/10.3390/cells9020311.
2406–2417, https://doi.org/10.1056/NEJMoa1915722. [286] V.A. Polito, H. Li, H. Martini-Stoica, B. Wang, L. Yang, Y. Xu, D.B. Swartzlander,
[266] Z.G. Zhang, Y. Li, C.T. Ng, Y.Q. Song, Inflammation in alzheimer’s disease and M. Palmieri, A. di Ronza, V.M. Lee, M. Sardiello, A. Ballabio, H. Zheng, Selective
molecular genetics: recent update, Arch. Immunol. Ther. Exp. 63 (5) (2015) clearance of aberrant tau proteins and rescue of neurotoxicity by transcription
333–344, https://doi.org/10.1007/s00005-015-0351-0. factor EB, EMBO Mol. Med. 6 (9) (2014) 1142–1160, https://doi.org/10.15252/
[267] B.L. Heckmann, B.J.W. Teubner, B. Tummers, E. Boada-Romero, L. Harris, emmm.201303671.
M. Yang, C.S. Guy, S.S. Zakharenko, D.R. Green, LC3-Associated endocytosis [287] M. Do, J. Park, Y. Chen, S.Y. Rah, T.T. Nghiem, J.H. Gong, S.A. Ju, B.S. Kim,
facilitates beta-amyloid clearance and mitigates neurodegeneration in murine R. Yu, J.W. Park, S.W. Ryter, Y.J. Surh, U.H. Kim, Y. Joe, H.T. Chung, PERK
alzheimer’s disease, Cell 178 (3) (2019) 536–551, https://doi.org/10.1016/j. activation by SB202190 ameliorates amyloidogenesis via the TFEB-induced
cell.2019.05.056, e14. autophagy-lysosomal pathway, Aging (Albany NY) 14 (3) (2022) 1233–1252,
[268] Y. Huang, K.E. Happonen, P.G. Burrola, C. O’Connor, N. Hah, L. Huang, https://doi.org/10.18632/aging.203899.
A. Nimmerjahn, G. Lemke, Microglia use TAM receptors to detect and engulf [288] Y. Xu, S. Du, J.A. Marsh, K. Horie, C. Sato, A. Ballabio, C.M. Karch, D.
amyloid beta plaques, Nat. Immunol. 22 (5) (2021) 586–594, https://doi.org/ M. Holtzman, H. Zheng, TFEB regulates lysosomal exocytosis of tau and its loss of
10.1038/s41590-021-00913-5. function exacerbates tau pathology and spreading, Mol. Psychiatr. 26 (10) (2021)
[269] A. Rocchi, S. Yamamoto, T. Ting, Y. Fan, K. Sadleir, Y. Wang, W. Zhang, S. Huang, 5925–5939, https://doi.org/10.1038/s41380-020-0738-0.
B. Levine, R. Vassar, C. He, A Becn1 mutation mediates hyperactive autophagic [289] P. Jiang, M. Gan, W.L. Lin, S.H. Yen, Nutrient deprivation induces alpha-
sequestration of amyloid oligomers and improved cognition in Alzheimer’s synuclein aggregation through endoplasmic reticulum stress response and
disease, PLoS Genet. 13 (8) (2017), e1006962, https://doi.org/10.1371/journal. SREBP2 pathway, Front. Aging Neurosci. 6 (2014) 268, https://doi.org/10.3389/
pgen.1006962. fnagi.2014.00268.
[270] F. Pickford, E. Masliah, M. Britschgi, K. Lucin, R. Narasimhan, P.A. Jaeger, [290] Y. Wang, M. Martinez-Vicente, U. Kruger, S. Kaushik, E. Wong, E.M. Mandelkow,
S. Small, B. Spencer, E. Rockenstein, B. Levine, T. Wyss-Coray, The autophagy- A.M. Cuervo, E. Mandelkow, Tau fragmentation, aggregation and clearance: the
related protein beclin 1 shows reduced expression in early Alzheimer disease and dual role of lysosomal processing, Hum. Mol. Genet. 18 (21) (2009) 4153–4170,
regulates amyloid beta accumulation in mice, J. Clin. Invest. 118 (6) (2008) https://doi.org/10.1093/hmg/ddp367.
2190–2199, https://doi.org/10.1172/JCI33585. [291] B. Caballero, M. Bourdenx, E. Luengo, A. Diaz, P.D. Sohn, X. Chen, C. Wang, Y.
[271] S. Mueller-Steiner, Y. Zhou, H. Arai, E.D. Roberson, B. Sun, J. Chen, X. Wang, R. Juste, S. Wegmann, B. Patel, Z.T. Young, S.Y. Kuo, J.A. Rodriguez-Navarro,
G. Yu, L. Esposito, L. Mucke, L. Gan, Antiamyloidogenic and neuroprotective H. Shao, M.G. Lopez, C.M. Karch, A.M. Goate, J.E. Gestwicki, B.T. Hyman, L. Gan,
functions of cathepsin B: implications for Alzheimer’s disease, Neuron 51 (6) A.M. Cuervo, Acetylated tau inhibits chaperone-mediated autophagy and
(2006) 703–714, https://doi.org/10.1016/j.neuron.2006.07.027. promotes tau pathology propagation in mice, Nat. Commun. 12 (1) (2021) 2238,
[272] F. Bellia, V. Lanza, S. Garcia-Vinuales, I.M.M. Ahmed, A. Pietropaolo, https://doi.org/10.1038/s41467-021-22501-9.
C. Iacobucci, G. Malgieri, G. D’Abrosca, R. Fattorusso, V.G. Nicoletti, D. Sbardella, [292] M. Bourdenx, A. Martin-Segura, A. Scrivo, J.A. Rodriguez-Navarro, S. Kaushik,
G.R. Tundo, M. Coletta, L. Pirone, E. Pedone, D. Calcagno, G. Grasso, D. Milardi, I. Tasset, A. Diaz, N.J. Storm, Q. Xin, Y.R. Juste, E. Stevenson, E. Luengo, C.
Ubiquitin binds the amyloid beta peptide and interferes with its clearance C. Clement, S.J. Choi, N.J. Krogan, E.V. Mosharov, L. Santambrogio,
pathways, Chem. Sci. 10 (9) (2019) 2732–2742, https://doi.org/10.1039/ F. Grueninger, L. Collin, D.L. Swaney, D. Sulzer, E. Gavathiotis, A.M. Cuervo,
c8sc03394c. Chaperone-mediated autophagy prevents collapse of the neuronal metastable
[273] T. Hamano, T.F. Gendron, E. Causevic, S.H. Yen, W.L. Lin, C. Isidoro, M. Deture, proteome, Cell 184 (10) (2021) 2696–2714, https://doi.org/10.1016/j.
L.W. Ko, Autophagic-lysosomal perturbation enhances tau aggregation in cell.2021.03.048, e25.

154
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

[293] E.F. Fang, M. Scheibye-Knudsen, L.E. Brace, H. Kassahun, T. SenGupta, H. Nilsen, dementia in diabetes, Faseb. J. 33 (3) (2019) 4458–4472, https://doi.org/
J.R. Mitchell, D.L. Croteau, V.A. Bohr, Defective mitophagy in XPA via PARP-1 10.1096/fj.201802043R.
hyperactivation and NAD(+)/SIRT1 reduction, Cell 157 (4) (2014) 882–896, [315] A. Tyagi, S. Pugazhenthi, Targeting insulin resistance to treat cognitive
https://doi.org/10.1016/j.cell.2014.03.026. dysfunction, Mol. Neurobiol. 58 (6) (2021) 2672–2691, https://doi.org/10.1007/
[294] J.S. Kerr, B.A. Adriaanse, N.H. Greig, M.P. Mattson, M.Z. Cader, V.A. Bohr, E. s12035-021-02283-3.
F. Fang, Mitophagy and alzheimer’s disease: cellular and molecular mechanisms, [316] G.E. Gibson, Q. Shi, A mitocentric view of Alzheimer’s disease suggests multi-
Trends Neurosci. 40 (3) (2017) 151–166, https://doi.org/10.1016/j. faceted treatments, J Alzheimers Dis 20 (Suppl 2) (2010) S591–S607, https://doi.
tins.2017.01.002. org/10.3233/JAD-2010-100336.
[295] D. Gatica, V. Lahiri, D.J. Klionsky, Cargo recognition and degradation by selective [317] J.S. Bhatti, G.K. Bhatti, P.H. Reddy, Mitochondrial dysfunction and oxidative
autophagy, Nat. Cell Biol. 20 (3) (2018) 233–242, https://doi.org/10.1038/ stress in metabolic disorders - a step towards mitochondria based therapeutic
s41556-018-0037-z. strategies, Biochim. Biophys. Acta, Mol. Basis Dis. 1863 (5) (2017) 1066–1077,
[296] C. Kondapalli, A. Kazlauskaite, N. Zhang, H.I. Woodroof, D.G. Campbell, https://doi.org/10.1016/j.bbadis.2016.11.010.
R. Gourlay, L. Burchell, H. Walden, T.J. Macartney, M. Deak, A. Knebel, D. [318] M.F. Galindo, I. Ikuta, X. Zhu, G. Casadesus, J. Jordan, Mitochondrial biology in
R. Alessi, M.M. Muqit, PINK1 is activated by mitochondrial membrane potential Alzheimer’s disease pathogenesis, J. Neurochem. 114 (4) (2010) 933–945,
depolarization and stimulates Parkin E3 ligase activity by phosphorylating Serine https://doi.org/10.1111/j.1471-4159.2010.06814.x.
65, Open Biol 2 (5) (2012), 120080, https://doi.org/10.1098/rsob.120080. [319] M. Webb, D.P. Sideris, M. Biddle, Modulation of mitochondrial dysfunction for
[297] L. Aerts, K. Craessaerts, B. De Strooper, V.A. Morais, PINK1 kinase catalytic treatment of disease, Bioorg. Med. Chem. Lett 29 (11) (2019) 1270–1277, https://
activity is regulated by phosphorylation on serines 228 and 402, J. Biol. Chem. doi.org/10.1016/j.bmcl.2019.03.041.
290 (5) (2015) 2798–2811, https://doi.org/10.1074/jbc.M114.620906. [320] M. Jazvinscak Jembrek, P.R. Hof, G. Simic, Ceramides in alzheimer’s disease: key
[298] L.A. Kane, M. Lazarou, A.I. Fogel, Y. Li, K. Yamano, S.A. Sarraf, S. Banerjee, R. mediators of neuronal apoptosis induced by oxidative stress and abeta
J. Youle, PINK1 phosphorylates ubiquitin to activate Parkin E3 ubiquitin ligase accumulation, Oxid. Med. Cell. Longev. 2015 (2015), 346783, https://doi.org/
activity, J. Cell Biol. 205 (2) (2014) 143–153, https://doi.org/10.1083/ 10.1155/2015/346783.
jcb.201402104. [321] A. Kumar, B. Yegla, T.C. Foster, Redox signaling in neurotransmission and
[299] F. Koyano, K. Okatsu, H. Kosako, Y. Tamura, E. Go, M. Kimura, Y. Kimura, cognition during aging, Antioxidants Redox Signal. 28 (18) (2018) 1724–1745,
H. Tsuchiya, H. Yoshihara, T. Hirokawa, T. Endo, E.A. Fon, J.F. Trempe, Y. Saeki, https://doi.org/10.1089/ars.2017.7111.
K. Tanaka, N. Matsuda, Ubiquitin is phosphorylated by PINK1 to activate parkin, [322] Q. Yu, F. Du, J.T. Douglas, H. Yu, S.S. Yan, S.F. Yan, Mitochondrial dysfunction
Nature 510 (7503) (2014) 162–166, https://doi.org/10.1038/nature13392. triggers synaptic deficits via activation of p38 MAP kinase signaling in
[300] J.F. Trempe, V. Sauve, K. Grenier, M. Seirafi, M.Y. Tang, M. Menade, S. Al-Abdul- differentiated alzheimer’s disease trans-mitochondrial cybrid cells, J Alzheimers
Wahid, J. Krett, K. Wong, G. Kozlov, B. Nagar, E.A. Fon, K. Gehring, Structure of Dis 59 (1) (2017) 223–239, https://doi.org/10.3233/JAD-170283.
parkin reveals mechanisms for ubiquitin ligase activation, Science 340 (6139) [323] K. Sinha, J. Das, P.B. Pal, P.C. Sil, Oxidative stress: the mitochondria-dependent
(2013) 1451–1455, https://doi.org/10.1126/science.1237908. and mitochondria-independent pathways of apoptosis, Arch. Toxicol. 87 (7)
[301] C.T. Chu, Mechanisms of selective autophagy and mitophagy: implications for (2013) 1157–1180, https://doi.org/10.1007/s00204-013-1034-4.
neurodegenerative diseases, Neurobiol. Dis. 122 (2019) 23–34, https://doi.org/ [324] A. Tarafdar, G. Pula, The role of NADPH oxidases and oxidative stress in
10.1016/j.nbd.2018.07.015. neurodegenerative disorders, Int. J. Mol. Sci. 19 (12) (2018) 3824, https://doi.
[302] M. Lazarou, D.A. Sliter, L.A. Kane, S.A. Sarraf, C. Wang, J.L. Burman, D.P. Sideris, org/10.3390/ijms19123824.
A.I. Fogel, R.J. Youle, The ubiquitin kinase PINK1 recruits autophagy receptors to [325] N. Koju, A. Taleb, J. Zhou, G. Lv, J. Yang, X. Cao, H. Lei, Q. Ding, Pharmacological
induce mitophagy, Nature 524 (7565) (2015) 309–314, https://doi.org/10.1038/ strategies to lower crosstalk between nicotinamide adenine dinucleotide
nature14893. phosphate (NADPH) oxidase and mitochondria, Biomed. Pharmacother. 111
[303] L. Wang, Y.L. Cho, Y. Tang, J. Wang, J.E. Park, Y. Wu, C. Wang, Y. Tong, (2019) 1478–1498, https://doi.org/10.1016/j.biopha.2018.11.128.
R. Chawla, J. Zhang, Y. Shi, S. Deng, G. Lu, Y. Wu, H.W. Tan, P. Pawijit, G.G. Lim, [326] R.L. Friede, Enzyme histochemical studies of senile plaques, J. Neuropathol. Exp.
H.Y. Chan, J. Zhang, L. Fang, H. Yu, Y.C. Liou, M. Karthik, B.H. Bay, K.L. Lim, S. Neurol. 24 (3) (1965) 477–491, https://doi.org/10.1097/00005072-196507000-
K. Sze, C.T. Yap, H.M. Shen, PTEN-L is a novel protein phosphatase for ubiquitin 00008.
dephosphorylation to inhibit PINK1-Parkin-mediated mitophagy, Cell Res. 28 (8) [327] M. Kowalska, T. Piekut, M. Prendecki, A. Sodel, W. Kozubski, J. Dorszewska,
(2018) 787–802, https://doi.org/10.1038/s41422-018-0056-0. Mitochondrial and nuclear DNA oxidative damage in physiological and
[304] Y. Wang, Y. Huang, J. Yang, F.Q. Zhou, L. Zhao, H. Zhou, Pyruvate is a pathological aging, DNA Cell Biol. 39 (8) (2020) 1410–1420, https://doi.org/
prospective alkalizer to correct hypoxic lactic acidosis, Mil Med Res 5 (1) (2018) 10.1089/dna.2019.5347.
13, https://doi.org/10.1186/s40779-018-0160-y. [328] R.S. Kalra, S.A. Bapat, Enhanced levels of double-strand DNA break repair
[305] S. Pickles, P. Vigie, R.J. Youle, Mitophagy and quality control mechanisms in proteins protect ovarian cancer cells against genotoxic stress-induced apoptosis,
mitochondrial maintenance, Curr. Biol. 28 (4) (2018) R170–R185, https://doi. J. Ovarian Res. 6 (1) (2013) 1–10, https://doi.org/10.1186/1757-2215-6-66.
org/10.1016/j.cub.2018.01.004. [329] P. Mecocci, U. MacGarvey, M.F. Beal, Oxidative damage to mitochondrial DNA is
[306] A. Kobro-Flatmoen, M.J. Lagartos-Donate, Y. Aman, P. Edison, M.P. Witter, E. increased in Alzheimer’s disease, Ann. Neurol. 36 (5) (1994) 747–751, https://
F. Fang, Re-emphasizing early Alzheimer’s disease pathology starting in select doi.org/10.1002/ana.410360510.
entorhinal neurons, with a special focus on mitophagy, Ageing Res. Rev. 67 [330] K. Ohno-Matsui, Parallel findings in age-related macular degeneration and
(2021), 101307, https://doi.org/10.1016/j.arr.2021.101307. Alzheimer’s disease, Prog. Retin. Eye Res. 30 (4) (2011) 217–238, https://doi.
[307] E.F. Fang, Y. Hou, K. Palikaras, B.A. Adriaanse, J.S. Kerr, B. Yang, S. Lautrup, M. org/10.1016/j.preteyeres.2011.02.004.
M. Hasan-Olive, D. Caponio, X. Dan, P. Rocktaschel, D.L. Croteau, M. Akbari, N. [331] S.M. de la Monte, T. Luong, T.R. Neely, D. Robinson, J.R. Wands, Mitochondrial
H. Greig, T. Fladby, H. Nilsen, M.Z. Cader, M.P. Mattson, N. Tavernarakis, V. DNA damage as a mechanism of cell loss in Alzheimer’s disease, Lab. Invest. 80
A. Bohr, Mitophagy inhibits amyloid-beta and tau pathology and reverses (8) (2000) 1323–1335, https://doi.org/10.1038/labinvest.3780140.
cognitive deficits in models of Alzheimer’s disease, Nat. Neurosci. 22 (3) (2019) [332] S. Oka, M. Ohno, D. Tsuchimoto, K. Sakumi, M. Furuichi, Y. Nakabeppu, Two
401–412, https://doi.org/10.1038/s41593-018-0332-9. distinct pathways of cell death triggered by oxidative damage to nuclear and
[308] G.L. Dharmaraj, F.D. Arigo, K.A. Young, R. Martins, R.L. Mancera, P. Bharadwaj, mitochondrial DNAs, EMBO J. 27 (2) (2008) 421–432, https://doi.org/10.1038/
Novel amylin analogues reduce amyloid-beta cross-seeding aggregation and sj.emboj.7601975.
neurotoxicity, J Alzheimers Dis 87 (1) (2022) 373–390, https://doi.org/10.3233/ [333] K.Y. Wong, J. Roy, M.L. Fung, B.C. Heng, C. Zhang, L.W. Lim, Relationships
JAD-215339. between mitochondrial dysfunction and neurotransmission failure in alzheimer’s
[309] Z.W. Yu, Y. Wang, X. Li, Y.H. Fu, Y. Yuan, H.Y. Li, X.Y. Gao, Brainstem auditory disease, Aging Dis 11 (5) (2020) 1291–1316, https://doi.org/10.14336/
evoked potential in cognitive impairment in patients with type 2 diabetes AD.2019.1125.
mellitus, Exp. Gerontol. 159 (2022), 111684, https://doi.org/10.1016/j. [334] S.C. Correia, P.I. Moreira, Role of Mitochondria in Neurodegenerative Diseases:
exger.2021.111684. the Dark Side of the “Energy Factory”, Mitochondrial Biology and Experimental
[310] R.H. Swerdlow, S. Koppel, I. Weidling, C. Hayley, Y. Ji, H.M. Wilkins, Therapeutics, Springer, 2018, pp. 213–239, https://doi.org/10.1007/978-3-319-
Mitochondria, cybrids, aging, and alzheimer’s disease, Prog Mol Biol Transl Sci 73344-9_11.
146 (2017) 259–302, https://doi.org/10.1016/bs.pmbts.2016.12.017. [335] A.M. Pisoschi, A. Pop, F. Iordache, L. Stanca, G. Predoi, A.I. Serban, Oxidative
[311] C. Carvalho, M.S. Santos, C.R. Oliveira, P.I. Moreira, Alzheimer’s disease and type stress mitigation by antioxidants - an overview on their chemistry and influences
2 diabetes-related alterations in brain mitochondria, autophagy and synaptic on health status, Eur. J. Med. Chem. 209 (2021), 112891, https://doi.org/
markers, Biochim. Biophys. Acta 1852 (8) (2015) 1665–1675, https://doi.org/ 10.1016/j.ejmech.2020.112891.
10.1016/j.bbadis.2015.05.001. [336] V. Bhatia, S. Sharma, Role of mitochondrial dysfunction, oxidative stress and
[312] I. Merdzo, I. Rutkai, V. Sure, P.V.G. Katakam, D.W. Busija, Effects of prolonged autophagy in progression of Alzheimer’s disease, J. Neurol. Sci. 421 (2021),
type 2 diabetes on mitochondrial function in cerebral blood vessels, Am. J. 117253, https://doi.org/10.1016/j.jns.2020.117253.
Physiol. Heart Circ. Physiol. 317 (5) (2019) H1086–H1092, https://doi.org/ [337] K. Takuma, J. Yao, J. Huang, H. Xu, X. Chen, J. Luddy, A.C. Trillat, D.M. Stern,
10.1152/ajpheart.00341.2019. O. Arancio, S.S. Yan, ABAD enhances Abeta-induced cell stress via mitochondrial
[313] N. Zhao, C.C. Liu, A.J. Van Ingelgom, Y.A. Martens, C. Linares, J.A. Knight, M. dysfunction, Faseb. J. 19 (6) (2005) 597–598, https://doi.org/10.1096/fj.04-
M. Painter, P.M. Sullivan, G. Bu, Apolipoprotein E4 impairs neuronal insulin 2582fje.
signaling by trapping insulin receptor in the endosomes, Neuron 96 (1) (2017) [338] K. Leuner, T. Schutt, C. Kurz, S.H. Eckert, C. Schiller, A. Occhipinti, S. Mai,
115–129, https://doi.org/10.1016/j.neuron.2017.09.003, e5. M. Jendrach, G.P. Eckert, S.E. Kruse, R.D. Palmiter, U. Brandt, S. Drose, I. Wittig,
[314] G.N. Ruegsegger, S. Manjunatha, P. Summer, S. Gopala, P. Zabeilski, S. Dasari, P. M. Willem, C. Haass, A.S. Reichert, W.E. Muller, Mitochondrion-derived reactive
M. Vanderboom, I.R. Lanza, K.A. Klaus, K.S. Nair, Insulin deficiency and oxygen species lead to enhanced amyloid beta formation, Antioxidants Redox
intranasal insulin alter brain mitochondrial function: a potential factor for Signal. 16 (12) (2012) 1421–1433, https://doi.org/10.1089/ars.2011.4173.

155
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

[339] Q. Cai, P. Tammineni, Mitochondrial aspects of synaptic dysfunction in [363] L. Mosconi, S. De Santi, J. Li, W.H. Tsui, Y. Li, M. Boppana, E. Laska, H. Rusinek,
alzheimer’s disease, J Alzheimers Dis 57 (4) (2017) 1087–1103, https://doi.org/ M.J. de Leon, Hippocampal hypometabolism predicts cognitive decline from
10.3233/JAD-160726. normal aging, Neurobiol. Aging 29 (5) (2008) 676–692, https://doi.org/10.1016/
[340] M.A. Ansari, S.W. Scheff, Oxidative stress in the progression of Alzheimer disease j.neurobiolaging.2006.12.008.
in the frontal cortex, J. Neuropathol. Exp. Neurol. 69 (2) (2010) 155–167, [364] Q.Y. Dong, T.R. Li, X.Y. Jiang, X.N. Wang, Y. Han, J.H. Jiang, Glucose metabolism
https://doi.org/10.1097/NEN.0b013e3181cb5af4. in the right middle temporal gyrus could be a potential biomarker for subjective
[341] N. Alikhani, L. Guo, S. Yan, H. Du, C.M. Pinho, J.X. Chen, E. Glaser, S.S. Yan, cognitive decline: a study of a Han population, Alzheimer’s Res. Ther. 13 (1)
Decreased proteolytic activity of the mitochondrial amyloid-beta degrading (2021) 74, https://doi.org/10.1186/s13195-021-00811-w.
enzyme, PreP peptidasome, in Alzheimer’s disease brain mitochondria, [365] M. Li, C. Long, L. Yang, Hippocampal-prefrontal circuit and disrupted functional
J Alzheimers Dis 27 (1) (2011) 75–87, https://doi.org/10.3233/JAD-2011- connectivity in psychiatric and neurodegenerative disorders, BioMed Res. Int.
101716. 2015 (2015), 810548, https://doi.org/10.1155/2015/810548.
[342] M. Calvo-Rodriguez, B.J. Bacskai, Mitochondria and calcium in alzheimer’s [366] M.K. Jha, S. Jeon, K. Suk, Pyruvate dehydrogenase kinases in the nervous system:
disease: from cell signaling to neuronal cell death, Trends Neurosci. 44 (2) (2021) their principal functions in neuronal-glial metabolic interaction and neuro-
136–151, https://doi.org/10.1016/j.tins.2020.10.004. metabolic disorders, Curr. Neuropharmacol. 10 (4) (2012) 393–403, https://doi.
[343] A.K. Ramachandran, S. Das, A. Joseph, G.G. Shenoy, A.T. Alex, J. Mudgal, org/10.2174/157015912804143586.
Neurodegenerative pathways in alzheimer’s disease: a review, Curr. [367] A. Nir, B. Barak, White matter alterations in Williams syndrome related to
Neuropharmacol. 19 (5) (2021) 679–692, https://doi.org/10.2174/ behavioral and motor impairments, Glia 69 (1) (2021) 5–19, https://doi.org/
1570159X18666200807130637. 10.1002/glia.23868.
[344] A. Konar, R.S. Kalra, A. Chaudhary, A. Nayak, K.P. Guruprasad, K. Satyamoorthy, [368] D.H. Silverman, C.T. Truong, S.K. Kim, C.Y. Chang, W. Chen, A.P. Kowell, J.
Y. Ishida, K. Terao, S.C. Kaul, R. Wadhwa, Identification of caffeic acid phenethyl L. Cummings, J. Czernin, G.W. Small, M.E. Phelps, Prognostic value of regional
ester (CAPE) as a potent neurodifferentiating natural compound that improves cerebral metabolism in patients undergoing dementia evaluation: comparison to a
cognitive and physiological functions in animal models of neurodegenerative quantifying parameter of subsequent cognitive performance and to prognostic
diseases, Front. Aging Neurosci. 12 (2020), 561925, https://doi.org/10.3389/ assessment without PET, Mol. Genet. Metabol. 80 (3) (2003) 350–355, https://
fnagi.2020.561925. doi.org/10.1016/S1096-7192(03)00139-2.
[345] L.A.S. Neves, Linking Mitochondrial Src and Hippocampal Dendritic Changes in [369] K. Herholz, Cerebral glucose metabolism in preclinical and prodromal
Alzheimer’s Disease, Universidade de Coimbra, 2021 available at: https://eg.uc. Alzheimer’s disease, Expert Rev. Neurother. 10 (11) (2010) 1667–1673, https://
pt/handle/10316/94314. (Accessed 17 July 2022). doi.org/10.1586/ern.10.136.
[346] F.L. Heppner, R.M. Ransohoff, B. Becher, Immune attack: the role of inflammation [370] G. Chetelat, B. Desgranges, B. Landeau, F. Mezenge, J.B. Poline, V. de la Sayette,
in Alzheimer disease, Nat. Rev. Neurosci. 16 (6) (2015) 358–372, https://doi.org/ F. Viader, F. Eustache, J.C. Baron, Direct voxel-based comparison between grey
10.1038/nrn3880. matter hypometabolism and atrophy in Alzheimer’s disease, Brain 131 (Pt 1)
[347] P.I. Moreira, P.L. Harris, X. Zhu, M.S. Santos, C.R. Oliveira, M.A. Smith, G. Perry, (2008) 60–71, https://doi.org/10.1093/brain/awm288.
Lipoic acid and N-acetyl cysteine decrease mitochondrial-related oxidative stress [371] J. Yao, R.T. Hamilton, E. Cadenas, R.D. Brinton, Decline in mitochondrial
in Alzheimer disease patient fibroblasts, J Alzheimers Dis 12 (2) (2007) 195–206, bioenergetics and shift to ketogenic profile in brain during reproductive
https://doi.org/10.3233/jad-2007-12210. senescence, Biochim. Biophys. Acta 1800 (10) (2010) 1121–1126, https://doi.
[348] P. Picone, D. Nuzzo, L. Caruana, V. Scafidi, M. Di Carlo, Mitochondrial org/10.1016/j.bbagen.2010.06.002.
dysfunction: different routes to Alzheimer’s disease therapy, Oxid. Med. Cell. [372] T.G. Demarest, V.R. Varma, D. Estrada, M. Babbar, S. Basu, U.V. Mahajan,
Longev. 2014 (2014), 780179, https://doi.org/10.1155/2014/780179. R. Moaddel, D.L. Croteau, M. Thambisetty, M.P. Mattson, V.A. Bohr, Biological
[349] P. Norat, S. Soldozy, J.D. Sokolowski, C.M. Gorick, J.S. Kumar, Y. Chae, sex and DNA repair deficiency drive Alzheimer’s disease via systemic metabolic
K. Yagmurlu, F. Prada, M. Walker, M.R. Levitt, R.J. Price, P. Tvrdik, M.Y. remodeling and brain mitochondrial dysfunction, Acta Neuropathol. 140 (1)
S. Kalani, Mitochondrial dysfunction in neurological disorders: exploring (2020) 25–47, https://doi.org/10.1007/s00401-020-02152-8.
mitochondrial transplantation, NPJ Regen Med 5 (1) (2020) 22, https://doi.org/ [373] A. Liskova, M. Samec, L. Koklesova, E. Kudela, P. Kubatka, O. Golubnitschaja,
10.1038/s41536-020-00107-x. Mitochondriopathies as a clue to systemic disorders-analytical tools and
[350] G. Monzio Compagnoni, A. Di Fonzo, S. Corti, G.P. Comi, N. Bresolin, E. Masliah, mitigating measures in context of predictive, preventive, and personalized (3P)
The role of mitochondria in neurodegenerative diseases: the lesson from medicine, Int. J. Mol. Sci. 22 (4) (2021), https://doi.org/10.3390/ijms22042007,
alzheimer’s disease and Parkinson’s disease, Mol. Neurobiol. 57 (7) (2020) 2007.
2959–2980, https://doi.org/10.1007/s12035-020-01926-1. [374] J. Woo, H. Cho, Y. Seol, S.H. Kim, C. Park, A. Yousefian-Jazi, S.J. Hyeon, J. Lee,
[351] W. Wang, F. Zhao, X. Ma, G. Perry, X. Zhu, Mitochondria dysfunction in the H. Ryu, Power failure of mitochondria and oxidative stress in neurodegeneration
pathogenesis of Alzheimer’s disease: recent advances, Mol. Neurodegener. 15 (1) and its computational models, Antioxidants 10 (2) (2021) 229, https://doi.org/
(2020) 30, https://doi.org/10.1186/s13024-020-00376-6. 10.3390/antiox10020229.
[352] I.W. Weidling, R.H. Swerdlow, Mitochondria in Alzheimer’s disease and their [375] A. Salminen, A. Haapasalo, A. Kauppinen, K. Kaarniranta, H. Soininen,
potential role in Alzheimer’s proteostasis, Exp. Neurol. 330 (2020), 113321, M. Hiltunen, Impaired mitochondrial energy metabolism in Alzheimer’s disease:
https://doi.org/10.1016/j.expneurol.2020.113321. impact on pathogenesis via disturbed epigenetic regulation of chromatin
[353] C. Shao, S. Xiong, G.M. Li, L. Gu, G. Mao, W.R. Markesbery, M.A. Lovell, Altered landscape, Prog. Neurobiol. 131 (2015) 1–20, https://doi.org/10.1016/j.
8-oxoguanine glycosylase in mild cognitive impairment and late-stage pneurobio.2015.05.001.
Alzheimer’s disease brain, Free Radic. Biol. Med. 45 (6) (2008) 813–819, https:// [376] L. Hedskog, S. Zhang, M. Ankarcrona, Strategic role for mitochondria in
doi.org/10.1016/j.freeradbiomed.2008.06.003. Alzheimer’s disease and cancer, Antioxidants Redox Signal. 16 (12) (2012)
[354] N. Shah, R. Singh, U. Sarangi, N. Saxena, A. Chaudhary, G. Kaur, S.C. Kaul, 1476–1491, https://doi.org/10.1089/ars.2011.4259.
R. Wadhwa, Combinations of Ashwagandha leaf extracts protect brain-derived [377] A. Grimm, K. Friedland, A. Eckert, Mitochondrial dysfunction: the missing link
cells against oxidative stress and induce differentiation, PLoS One 10 (3) (2015), between aging and sporadic Alzheimer’s disease, Biogerontology 17 (2) (2016)
e0120554, https://doi.org/10.1371/journal.pone.0120554. 281–296, https://doi.org/10.1007/s10522-015-9618-4.
[355] V. Parikh, D.A. Bangasser, Cholinergic signaling dynamics and cognitive control [378] R.S. Kalra, J.K. Dhanjal, M. Das, B. Singh, R. Naithani, Cell transdifferentiation
of attention, Curr Top Behav Neurosci 45 (2020) 71–87, https://doi.org/ and reprogramming in disease modeling: insights into the neuronal and cardiac
10.1007/7854_2020_133. disease models and current translational strategies, Cells 10 (10) (2021) 2558,
[356] P. Sharma, P. Srivastava, A. Seth, P.N. Tripathi, A.G. Banerjee, S.K. Shrivastava, https://doi.org/10.3390/cells10102558.
Comprehensive review of mechanisms of pathogenesis involved in Alzheimer’s [379] R. Kandimalla, P.H. Reddy, Multiple faces of dynamin-related protein 1 and its
disease and potential therapeutic strategies, Prog. Neurobiol. 174 (2019) 53–89, role in Alzheimer’s disease pathogenesis, Biochim. Biophys. Acta 1862 (4) (2016)
https://doi.org/10.1016/j.pneurobio.2018.12.006. 814–828, https://doi.org/10.1016/j.bbadis.2015.12.018.
[357] E. Zenaro, G. Piacentino, G. Constantin, The blood-brain barrier in Alzheimer’s [380] W.I. Ryu, M.K. Bormann, M. Shen, D. Kim, B. Forester, Y. Park, J. So, H. Seo, K.
disease, Neurobiol. Dis. 107 (2017) 41–56, https://doi.org/10.1016/j. C. Sonntag, B.M. Cohen, Brain cells derived from Alzheimer’s disease patients
nbd.2016.07.007. have multiple specific innate abnormalities in energy metabolism, Mol. Psychiatr.
[358] B.D. Freudenthal, W.A. Beard, L. Perera, D.D. Shock, T. Kim, T. Schlick, S. 26 (10) (2021) 5702–5714, https://doi.org/10.1038/s41380-021-01068-3.
H. Wilson, Uncovering the polymerase-induced cytotoxicity of an oxidized [381] C.R. Jack Jr., D.A. Bennett, K. Blennow, M.C. Carrillo, B. Dunn, S.B. Haeberlein,
nucleotide, Nature 517 (7536) (2015) 635–639, https://doi.org/10.1038/ D.M. Holtzman, W. Jagust, F. Jessen, J. Karlawish, E. Liu, J.L. Molinuevo,
nature13886. T. Montine, C. Phelps, K.P. Rankin, C.C. Rowe, P. Scheltens, E. Siemers, H.
[359] T. Lindahl, Instability and decay of the primary structure of DNA, Nature 362 M. Snyder, R. Sperling, Contributors, NIA-AA Research Framework: toward a
(6422) (1993) 709–715, https://doi.org/10.1038/362709a0. biological definition of Alzheimer’s disease, Alzheimers Dement 14 (4) (2018)
[360] S.H. Shin, W.Y. Park, D. Park, Characterization of DNA lesions associated with 535–562, https://doi.org/10.1016/j.jalz.2018.02.018.
cell-free DNA by targeted deep sequencing, BMC Med. Genom. 14 (1) (2021) 192, [382] L.R. Gray, S.C. Tompkins, E.B. Taylor, Regulation of pyruvate metabolism and
https://doi.org/10.1186/s12920-021-01040-8. human disease, Cell. Mol. Life Sci. 71 (14) (2014) 2577–2604, https://doi.org/
[361] C. Duran-Aniotz, C. Hetz, Glucose metabolism: a sweet relief of alzheimer’s 10.1007/s00018-013-1539-2.
disease, Curr. Biol. 26 (17) (2016) R806–R809, https://doi.org/10.1016/j. [383] S. Tacconi, R. Perri, E. Balestrieri, S. Grelli, S. Bernardini, R. Annichiarico,
cub.2016.07.060. A. Mastino, C. Caltagirone, B. Macchi, Increased caspase activation in peripheral
[362] V.K. Sharma, T.G. Singh, V. Mehta, Stressed mitochondria: a target to intrude blood mononuclear cells of patients with Alzheimer’s disease, Exp. Neurol. 190
alzheimer’s disease, Mitochondrion 59 (2021) 48–57, https://doi.org/10.1016/j. (1) (2004) 254–262, https://doi.org/10.1016/j.expneurol.2004.07.009.
mito.2021.04.004. [384] L. Gatta, A. Cardinale, F. Wannenes, C. Consoli, A. Armani, F. Molinari,
C. Mammi, F. Stocchi, M. Torti, G.M. Rosano, M. Fini, Peripheral blood

156
S. Dewanjee et al. Free Radical Biology and Medicine 193 (2022) 134–157

mononuclear cells from mild cognitive impairment patients show deregulation of [398] J. Mendiola-Precoma, L.C. Berumen, K. Padilla, G. Garcia-Alcocer, Therapies for
Bax and Sod1 mRNAs, Neurosci. Lett. 453 (1) (2009) 36–40, https://doi.org/ prevention and treatment of alzheimer’s disease, BioMed Res. Int. 2016 (2016),
10.1016/j.neulet.2009.02.003. 2589276, https://doi.org/10.1155/2016/2589276.
[385] Y. Peng, P. Gao, L. Shi, L. Chen, J. Liu, J. Long, Central and peripheral metabolic [399] J. Gotz, L.M. Ittner, Animal models of Alzheimer’s disease and frontotemporal
defects contribute to the pathogenesis of alzheimer’s disease: targeting dementia, Nat. Rev. Neurosci. 9 (7) (2008) 532–544, https://doi.org/10.1038/
mitochondria for diagnosis and prevention, Antioxidants Redox Signal. 32 (16) nrn2420.
(2020) 1188–1236, https://doi.org/10.1089/ars.2019.7763. [400] W. Fu, D. Shi, D. Westaway, J.H. Jhamandas, Bioenergetic mechanisms in
[386] S. Maynard, A.M. Hejl, T.S. Dinh, G. Keijzers, A.M. Hansen, C. Desler, M. Moreno- astrocytes may contribute to amyloid plaque deposition and toxicity, J. Biol.
Villanueva, A. Burkle, L.J. Rasmussen, G. Waldemar, V.A. Bohr, Defective Chem. 290 (20) (2015) 12504–12513, https://doi.org/10.1074/jbc.
mitochondrial respiration, altered dNTP pools and reduced AP endonuclease 1 M114.618157.
activity in peripheral blood mononuclear cells of Alzheimer’s disease patients, [401] G.R. Tundo, D. Sbardella, C. Ciaccio, G. Grasso, M. Gioia, A. Coletta, F. Polticelli,
Aging (Albany NY) 7 (10) (2015) 793–815, https://doi.org/10.18632/ D. Di Pierro, D. Milardi, P. Van Endert, S. Marini, M. Coletta, Multiple functions of
aging.100810. insulin-degrading enzyme: a metabolic crosslight? Crit. Rev. Biochem. Mol. Biol.
[387] E. Candeias, A.I. Duarte, C. Carvalho, S.C. Correia, S. Cardoso, R.X. Santos, A. 52 (5) (2017) 554–582, https://doi.org/10.1080/10409238.2017.1337707.
I. Placido, G. Perry, P.I. Moreira, The impairment of insulin signaling in [402] M.M. Anwar, Oxidative stress-A direct bridge to central nervous system
Alzheimer’s disease, IUBMB Life 64 (12) (2012) 951–957, https://doi.org/ homeostatic dysfunction and Alzheimer’s disease, Cell Biochem. Funct. 40 (1)
10.1002/iub.1098. (2022) 17–27, https://doi.org/10.1002/cbf.3673.
[388] J. Raskin, J. Cummings, J. Hardy, K. Schuh, R.A. Dean, Neurobiology of [403] T.J. Huat, J. Camats-Perna, E.A. Newcombe, N. Valmas, M. Kitazawa,
alzheimer’s disease: integrated molecular, physiological, anatomical, biomarker, R. Medeiros, Metal toxicity links to alzheimer’s disease and neuroinflammation,
and cognitive dimensions, Curr. Alzheimer Res. 12 (8) (2015) 712–722, https:// J. Mol. Biol. 431 (9) (2019) 1843–1868, https://doi.org/10.1016/j.
doi.org/10.2174/1567205012666150701103107. jmb.2019.01.018.
[389] Z. Cheng, Y. Tseng, M.F. White, Insulin signaling meets mitochondria in [404] F. Nikbakht, Y. Khadem, S. Haghani, H. Hoseininia, A. Moein Sadat, P. Heshemi,
metabolism, Trends Endocrinol. Metabol. 21 (10) (2010) 589–598, https://doi. N. Jamali, Protective role of apigenin against abeta 25-35 toxicity via inhibition
org/10.1016/j.tem.2010.06.005. of mitochondrial cytochrome c release, Basic Clin. Neurosci. 10 (6) (2019)
[390] Y. Tong, S. Xu, L. Huang, C. Chen, Obesity and insulin resistance: pathophysiology 557–566, https://doi.org/10.32598/BCN.9.10.385.
and treatment, Drug Discov. Today 27 (3) (2022) 822–830, https://doi.org/ [405] J.Y. Wang, L.L. Wen, Y.N. Huang, Y.T. Chen, M.C. Ku, Dual effects of antioxidants
10.1016/j.drudis.2021.11.001. in neurodegeneration: direct neuroprotection against oxidative stress and indirect
[391] R.S. Kalra, A. Chaudhary, A. Omar, C.T. Cheung, S. Garg, S.C. Kaul, R. Wadhwa, protection via suppression of glia-mediated inflammation, Curr. Pharmaceut. Des.
Stress-induced changes in CARF expression determine cell fate to death, survival, 12 (27) (2006) 3521–3533, https://doi.org/10.2174/138161206778343109.
or malignant transformation, Cell Stress & Chaperones 25 (3) (2020) 481–494, [406] M.A. Daulatzai, Cerebral hypoperfusion and glucose hypometabolism: key
https://doi.org/10.1007/s12192-020-01088-y. pathophysiological modulators promote neurodegeneration, cognitive
[392] L. Liu, J. Bai, F. Liu, Y. Xu, M. Zhao, C. Zhao, Z. Zhou, Cross-talking pathways of impairment, and Alzheimer’s disease, J. Neurosci. Res. 95 (4) (2017) 943–972,
forkhead box O1 (FOXO1) are involved in the pathogenesis of alzheimer’s disease https://doi.org/10.1002/jnr.23777.
and huntington’s disease, Oxid. Med. Cell. Longev. 2022 (2022), 7619255, [407] S. Mitra, A. Banik, S. Saurabh, M. Maulik, S.N. Khatri, Neuroimmunometabolism:
https://doi.org/10.1155/2022/7619255. a new pathological nexus underlying neurodegenerative disorders, J. Neurosci. 42
[393] J. Spaas, L. van Veggel, M. Schepers, A. Tiane, J. van Horssen, D.M. Wilson 3rd, P. (10) (2022) 1888–1907, https://doi.org/10.1523/JNEUROSCI.0998-21.2022.
R. Moya, E. Piccart, N. Hellings, B.O. Eijnde, W. Derave, R. Schreiber, [408] S. Zulfiqar, P. Garg, K. Nieweg, Contribution of astrocytes to metabolic
T. Vanmierlo, Oxidative stress and impaired oligodendrocyte precursor cell dysfunction in the Alzheimer’s disease brain, Biol. Chem. 400 (9) (2019)
differentiation in neurological disorders, Cell. Mol. Life Sci. 78 (10) (2021) 1113–1127, https://doi.org/10.1515/hsz-2019-0140.
4615–4637, https://doi.org/10.1007/s00018-021-03802-0. [409] K.K. Dey, H. Wang, M. Niu, B. Bai, X. Wang, Y. Li, J.H. Cho, H. Tan, A. Mishra, A.
[394] N. Pathak, S.K. Vimal, I. Tandon, L. Agrawal, C. Hongyi, S. Bhattacharyya, A. High, P.C. Chen, Z. Wu, T.G. Beach, J. Peng, Deep undepleted human serum
Neurodegenerative disorders of alzheimer, parkinsonism, amyotrophic lateral proteome profiling toward biomarker discovery for Alzheimer’s disease, Clin.
sclerosis and multiple sclerosis: an early diagnostic approach for precision Proteonomics 16 (2019) 16, https://doi.org/10.1186/s12014-019-9237-1.
treatment, Metab. Brain Dis. 37 (1) (2022) 67–104, https://doi.org/10.1007/ [410] P.G. Ridge, J.S.K. Kauwe, Mitochondria and alzheimer’s disease: the role of
s11011-021-00800-w. mitochondrial genetic variation, Curr Genet Med Rep 6 (1) (2018) 1–10, https://
[395] S.M. de la Monte, Brain insulin resistance and deficiency as therapeutic targets in doi.org/10.1007/s40142-018-0132-2.
Alzheimer’s disease, Curr. Alzheimer Res. 9 (1) (2012) 35–66, https://doi.org/ [411] A. Bobba, V.A. Petragallo, E. Marra, A. Atlante, Alzheimer’s proteins, oxidative
10.2174/156720512799015037. stress, and mitochondrial dysfunction interplay in a neuronal model of
[396] P. Edison, Neuroinflammation, microglial activation, and glucose metabolism in Alzheimer’s disease, Int. J. Alzheimer’s Dis. 2010 (2010), 621870, https://doi.
neurodegenerative diseases, Int. Rev. Neurobiol. 154 (2020) 325–344, https:// org/10.4061/2010/621870.
doi.org/10.1016/bs.irn.2020.03.017. [412] A. Eckert, R. Nisbet, A. Grimm, J. Gotz, March separate, strike together–role of
[397] G. Morris, A.F. Carvalho, G. Anderson, P. Galecki, M. Maes, The many phosphorylated TAU in mitochondrial dysfunction in Alzheimer’s disease,
neuroprogressive actions of tryptophan catabolites (TRYCATs) that may be Biochim. Biophys. Acta 1842 (8) (2014) 1258–1266, https://doi.org/10.1016/j.
associated with the pathophysiology of neuro-immune disorders, Curr. bbadis.2013.08.01.
Pharmaceut. Des. 22 (8) (2016) 963–977, https://doi.org/10.2174/ [413] D. Daugherty, J. Goldberg, W. Fischer, R. Dargusch, P. Maher, D. Schubert,
1381612822666151215102420. A novel Alzheimer’s disease drug candidate targeting inflammation and fatty acid
metabolism, Alzheimer’s Res. Ther. 9 (1) (2017) 50, https://doi.org/10.1186/
s13195-017-0277-3.

157

You might also like