Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Journal of Chemical Neuroanatomy 95 (2019) 43–53

Contents lists available at ScienceDirect

Journal of Chemical Neuroanatomy


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

Review

Mitochondrial SIRT3 and neurodegenerative brain disorders T


Anamika, Archita Khanna, Papia Acharjee, Arup Acharjee, Surendra Kumar Trigun

Biochemistry Section, Department of Zoology, Institute of Science, Banaras Hindu University Varanasi, 221005, India

ARTICLE INFO ABSTRACT

Keywords: Sirtuins are highly conserved NAD+ dependent class III histone deacetylases and catalyze deacetylation and ADP
Sirtuins ribosylation of a number of non-histone proteins. Since, they require NAD+ for their activity, the cellular level of
SIRT3 Sirtuins represents redox status of the cells and thereby serves as bona fide metabolic stress sensors. Out of seven
Mitochondrial derangement homologues of Sirtuins identified in mammals, SIRT3, 4 & 5 have been found to be localized and active in
Excitotoxicity
mitochondria. During recent past, clusters of protein substrates for SIRT3 have been identified in mitochondria
Neurodegenerative brain disorders
and thereby advocating SIRT3 as the main mitochondrial Sirtuin which could be involved in protecting stress
induced mitochondrial integrity and energy metabolism. As mitochondrial dysfunction underlies the patho-
genesis of almost all neurodegenerative diseases, a role of SIRT3 becomes an arguable speculation in such brain
disorders. Some recent findings demonstrate that SIRT3 over expression could prevent neuronal derangements in
certain in vivo and in vitro models of aging and neurodegenerative brain disorders like; Alzheimer’s disease,
Huntington’s disease, stroke etc. Similarly, loss of SIRT3 has been found to accelerate neurodegeneration in the
brain challenged with excitotoxicity. Therefore, it is argued that SIRT3 could be a relevant target to understand
pathogenesis of neurodegenerative brain disorders. This review is an attempt to summarize recent findings on
(1) the implication of SIRT3 in neurodegenerative brain disorders and (2) whether SIRT3 modulation could
ameliorate neuropathologies in relevant models.

1. Introduction et al., 2007; Haigis and Sinclair, 2010; Cantó and Auwerx, 2012). In-
deed, these proteins are now described to regulate key cellular pro-
A protein, first identified in brewer’s yeast S. cerevisiae, was found to cesses of metabolic adaptations and stress responses and thereby
be primarily involved in delaying cell senescence in this organism by maintain health and longevity (Haigis and Sinclair, 2010).
gene silencing mechanisms and it was named as silent information Importantly, reports from last decade emphasize that some of the
regulator 2 (SIR2) (Haigis and Sinclair, 2010). Later SIR2 was found to Sirtuins, SIRT1 and SIRT3 in particular, are expressed in most of the
be a member of an evolutionarily conserved family of proteins, known brain regions (Sidorova-Darmos et al., 2014) and they help maintain
as Sirtuins, found in almost all life forms and catalyze NAD+ dependent higher order brain functions and protect brain cells from un-
protein deacetylation. Notably, a direct link of Sirtuin activity with that physiological insults and neurodegeneration (Donmez and Outeiro,
of calorie restriction (CR) and aging in many organisms (Imai et al., 2013). Therefore, the present review is organized to first present an
2000; Lin et al., 2000) strongly advocated about roles of such protein overview on Sirtuin biochemistry in general and then to describe their
deacetylases in maintaining normal cellular functions, cell and/or or- roles in maintaining brain functions and neuropathologies. Since, mi-
ganism senescence and age-related diseases in mammals. tochondrial dysfunction underlies the pathogenesis of most of the
A rapid progress in Sirtuin research, thereafter, emphasized that neurodegenerative brain disorders; much emphasis has been given on
these protein deacetylases target an array of metabolic and cell sig- this mitochondria active deacetylase in the brain.
naling proteins and thus, act as an important epigenetic regulator of cell
functions. Use of NAD+ for each cycle of their catalytic activity cate- 1.1. Biology of SIRTUINS
gorized them as a bona fide metabolic/bioenergetic sensor of the cells
(Berger et al., 2005; Haigis and Sinclair, 2010). Presence of conserved As illustrated in Fig. 1, the catalytic deacetylation by all the Sirtuins,
allosteric sites for activators (resveratrol and related compounds) and including SIRT3, begins with hydrolysis of the glycosidic bond of NAD+
inhibitor (nicotinamide) and identification of many protein modifica- to generate nicotinamide (NAM) and an ADP-ribose intermediate. This
tion sites make them a target of multimodal regulations as well (Tanno follows transfer of the acetyl group from a lysine residue of an


Corresponding author.
E-mail address: sktrigun@gmail.com (S.K. Trigun).

https://doi.org/10.1016/j.jchemneu.2017.11.009
Received 20 June 2017; Received in revised form 16 September 2017; Accepted 8 November 2017
Available online 09 November 2017
0891-0618/ © 2017 Elsevier B.V. All rights reserved.
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

Table 1
Biochemical classification of Sirtuins: Catalytic specificity and structural details (Carafa
et al., 2012; Herskovits and Guarente, 2013).

Class Sirtuins Acitvity Structure MW (kDa): AA (Catalytic


domain)

Class I SIRT1 Deacetylase 81.7: 747 (244–498)


SIRT2 41.5: 389 (65–340)
SIRT3 43.6: 399 (139–382)
Class II SIRT4 ADP- ribosyltransferase 35.2: 314 (45–314)
Class III SIRT5 Demalonylase 33.9: 310 (41–309)
Desuccinylase
Deacetylase
Class IV SIRT6 ADP-ribosyltransferase 39.1: 355 (35–274)
SIRT7 Deacetylase () 44.9: 400 (90–331)

genes involved in promoting cell survival by deacetylating an array of


transcription factors (Pillai et al., 2010a). SIRT2 resides in the cyto-
plasm and plays an important role in oxidative stress resistance (Wang
et al., 2007), cell motility and cell division by deacetylating α-tubulin
(North et al., 2003). The Sirtuin isoforms targeted to mitochondria are
Fig. 1. Protein deacetylation reaction pathway of Sirtuons including SIRT3. The catalytic SIRT3, 4 and 5. These Sirtuins evidently serve as stress sensors and
reaction of all the Sirtuins is completed in two steps. It starts with cleavage of the NAD+
modulate the activity of several key mitochondrial proteins to induce
glycoside to produce NAM and an ADP-ribose which, in the next step, accepts acetate
group transferred from the susceptible lysine residue of an acetylated protein to syn-
adaptive changes during bioenergetic deficits, ROS insult, apoptosis,
thesize 2′-O-acetyl-ADP-ribose and thus leaving behind the substrate protein deacety- aberrant signal transduction and deranged intermediary metabolism
lated. Since cells cannot afford to lose NAD+ after each Sirtuins reaction, they operate an (Verdin et al., 2010). As evident from a bigger list of protein substrates
enzymatic mechanism to reuse NAM, the NAD+ cleavage product, to re-generate NAD+. (Table 2), SIRT3 could be considered as the main mitochondrial NAD+-
NAM is first converted into NMN by the enzyme NAMPT which, subsequently is converted dependent stress responsive protein deacetylase (Verdin et al., 2010).
back to NAD+ by another enzyme, NMNAT.

1.2. Sirtuins in brain


acetylated protein to the 2′-OH of ADP-ribose intermediate to produce
2′-O-acetyl-ADP-ribose. Thus finally Sirtuins activity ends up with As illustrated in Table 3, out of all the Sirtuins, SIRT1 & SIRT3 re-
generating a deacetylated protein, a NAM and a 2′-O-acetyl-ADP-ribose present maximum Sirtuins activity in the neurons and are reported to
(Haigis and Guarente, 2006). regulate most of the critical brain functions and thus, considered to be
Each cycle of the Sirtuin catalyzed reaction consumes one equiva- involved in the pathogenesis of almost all age related and neurode-
lent of NAD+ and thereby likely to swing the NAD+: NADH ratio in the generative brain disorders. In the rodent brain, SIRT1 has been the most
cells. Thus, the level of the Sirtuins activity is considered to directly studied Sirtuins wherein, it is demonstrated to be mainly expressed in
relate with the redox and bioenergetic status of the cell. For example, a the neurons (Ramadori et al., 2008). SIRT1 expression has been de-
reduced NAD+: NADH ratio becomes inevitable during increased tected as early as 3 day postnatal mice brain localized in both nucleus
Sirtuins activity and therefore, mammalian cells have been evolved and cytosol whereas, in adult brain, it is predominantly cytosolic (Li
with an enzymatic mechanism to regenerate NAD+ from the NAM. As et al., 2008). SIRT1 has been detected in CA1 to CA4 regions of the
illustrated in Fig. 1, this happens in two steps: Nampt (Nicotinamide hippocampus, basal ganglia circuits (e.g. Substantia nigra), brain stem
phosphoribosyltransferase) converts NAM to nicotinamide mono- (e.g. raphe nucleus) and cerebellum (Zakhary et al., 2010). Brain region
nucleotide (NMN) (Revollo et al., 2004). Subsequently, NMNAT (ni- specific differential distribution of this protein deacetylase is also on
cotinamide mononucleotide adenyl transferase), an another enzyme, record. The expression of SIRT1, at both transcript and protein levels, is
regenerate NAD+ from NMN (Berger et al., 2005; Haigis and Sinclair, found to be highest in cerebellum, moderate in cortex, hippocampus,
2010). Such NAD+ dependency of Sirtuins including mitochondrial striatum and olfactory bulb and lowest in the spinal cord (Sidorova-
SIRT3 qualify them as bona fide bioenerergetic sensors of the cells. Darmos et al., 2014). Moreover, it was important to note that absence of
In mammals, seven homologues of sirtuins (SIRT1-7) have so far SIRT1 could impair cognitive abilities including immediate memory,
been identified sharing a conserved catalytic core domain, however, classical conditioning and spatial learning (Michán et al., 2010) and
with slightly different N and C termini extensions (North and Verdin, thus suggested a critical role of SIRT1 in maintaining higher order brain
2004). The N terminal extension is responsible for their differential functions and synaptic plasticity including its role in promoting dif-
subcellular localization and substrate specificity whereas, variable C- ferentiation of neuronal progenitor cells (NPCs) (Wang et al., 2011).
termini, as extended region of the catalytic domain, probably con- Although SIRT1 has been the main Sirtuins target to understand
tribute in determining reaction specificity of different Sirtuins (North pathogenesis of many neurological diseases caused due to protein ag-
and Verdin, 2004; Moniot et al., 2012; Flick and Luscher, 2012). gregation or due to toxicity of α-synuclein, tau, huntingtin and Aβ
Accordingly, based on the type of reactions they catalyze, these peptide (Duan, 2013), some recent reports do argue for significant roles
Sirtuins have conveniently been classified into four classes. Sirtuins of other sirtuins in neurodegeneration (Donmez and Outeiro, 2013).
classification and specific structural details is given in Table 1. As Since, mitochondrial dysfunction lies at the centre of many neurode-
evident from the table data, with the robust deacetylase activity, class I generative disorders, as listed in Table 3, the SIRT3, primarily active in
Sirtuins; SIRT1, SIRT2 & SIRT3, are evident to regulate diversified cell neuronal mitochondria, could be considered another hot spot for un-
functions and thus, derive much scientific merit to study class I Sirtuins derstanding pathogenesis and therapeutic strategies against neurode-
as relevant targets to understand metabolic adaptations under normal generative diseases. Some reports during recent past suggest about a
and diseased conditions. direct association of SIRT3 deficiency with the induction of certain
Importance of Sirtuins got further highlighted due to their specific classical neurological diseases. For example, SIRT3 deficiency in Hu-
sub-cellular locatizations. As illustrated in Table 2, SIRT1, 6 and 7 are tingtons protein expressing brain cells (Fu et al., 2012), down regula-
found in the nucleus and known to regulate expression of many critical tion of SIRT3 in the frontal cortices of APOE4 carrier human brain (Yin

44
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

Table 2
Subcellular localization, target proteins and cellular/physiological functions known to be regulated by different Sirtuin homologues reported in mammals.

Sirtuins Subcellular location Protein Substrates Functions References

SIRT1 Nuclear p53, FOX03a, Bax, HIF-1α, STAT3, Energy metabolism, differentiation, neuroprotection Haigis and Sinclair, 2010
PGC1α, NFƙB, PPARγ
SIRT2 Cytoplasmic α-tubulin, H4, FOXO Cell cycle regulation North et al., 2003
SIRT3 Mitochondrial matrix LCAD, SDH, GDH, LKB1, NDUFA9, Fatty acid oxidation, amino acids catabolism, TCA cycle, Ahn et al., 2008; Sundaresan et al.,
IDH2, SOD2, FOXO3a, CypD, Ku70, ATP production, mito- protein synthesis ROS homeostasis, 2009; Qiu et al., 2010; Verdin et al.,
p53, MRPL10, PGC1α balance of survival and apoptosis, 2010; Hallows et al., 2011;
SIRT4 Mitochondrial matrix GDH Amino acid catabolism Verdin et al., 2010;
SIRT5 Inner Mitochondrial CPSI Urea Cycle Verdin et al., 2010;
Membrane Du et al., 2011
SIRT6 Nuclear H3K9, H3K56, HIF1α, NFƙB, DNA DNA repair and inflammation Nakagawa and Guarente, 2011; Jiang
polymerase β et al., 2013
SIRT7 Nucleolus RNA pol I, SMAD6, p53 RNA pol I activity Michan and Sinclair, 2007;
Haigis and Sinclair, 2010

et al., 2015a; Ansari et al., 2017), SIRT3 deficiency and increased loss of brain shows similar pattern in cortex, hippocampus, striatum, spinal
dopaminergic neurons in substantia nigra and striatum of MPTP (1- cord and brain stem and at modestly lower levels in the cerebellum
methyl-4-phenyl-1,2,3,6-tetrahydropyridine) induced PD mice (Liu (Sidorova-Darmos et al., 2014). However, SIRT3 expression has been
et al., 2015). found to be declined significantly in the cortex and hippocampus of
aging rats (Braidy et al., 2015) and thus, indicating its significant role in
age-associated brain disorders.
2. SIRT3
Therefore, to highlight mechanistic aspects about neuroprotective
roles of SIRT3, in the present review the structural and regulatory as-
SIRT3 has been reported to regulate almost every aspect of mi-
pects of this deacetylase have been given due space followed by sum-
tochondrial functions like; mitochondrial biogenesis and dynamics,
marizing the current status about how SIRT3 could be implicated in
ROS metabolism, ATP production and maintenance of mitochondrial
neuropathogenesis and neuroprotection.
integrity (Bause and Haigis 2013). In addition, SIRT3 is directly linked
to the human longevity also (Kong et al., 2010; Brown et al., 2013;
Kincaid & Bossy-Wetzel, 2013; Ansari et al., 2017). Though, SIRT3 2.1. SIRT3 structure and organelle targeting
Knockout mice exhibited a normal physiology under normal condition
but under stress condition or with aging, these animals develop age- Like other sirtuins, SIRT3 also utilizes NAD+ as the main cofactor
related diseases at an accelerated pace (Lombard and Zwaans, 2014; for its activity and hence, its activity directly relates with the increased
McDonnell et al., 2015). Similarly, SIRT3 knockout animals have been NAD+/NADH ratio in the cells (Haigis and Sinclair, 2010; Kincaid and
shown to have increased acetylation of mitochondrial proteins (Fritz Bossy-Wetzel, 2013). Furthermore, SIRT3 also contains a conserved
et al., 2012; Hebert et al., 2013) and altered mitochondrial metabolism core domain for the binding of NAD+ and the protein substrate.
like, fatty acid oxidation, urea cycle defects, declined ATP production Schematic arrangement of different functional and regulatory domains
and increased oxidative damage from ROS (Ahn et al., 2008; Qiu et al., of a full length SIRT3 has been illustrated in Fig. 2. A conserved cata-
2010; Hallows et al., 2011). Involvement of SIRT3 in metabolic adap- lytic core domain of ∼275 aa is consisting of a large NAD+ binding
tations of mitochondria against various metabolic disorders, cardiac Rossmann fold made up of several inverted classical open α/β struc-
dysfunction, cancer, aging and neurological disorder have been ex- tures followed by a substrate binding site and a groove of several loops
amined by various groups as well (Lombard et al., 2007; Someya et al., forming a connecting cleft between NAD+ and acetylated peptide
2007; Anderson et al., 2009; Nicholls, 2009). Moreover, it is becoming substrate (Carafa et al., 2012; Moniot et al., 2012). Many regulatory
clearer that SIRT3 could act as a potential therapeutic target with re- sites including Zn2+ binding domains have also been reported in SIRT3
spect to mitochondrial dysfunction led neurological disorders. (Min et al., 2013). Importantly, the amino and carboxy terminal ex-
This is further supported by the fact that the level of SIRT3 changes tensions autoregulate enzymatic activity of this protein, e.g. C terminal
according to the nutrient status of the cell in a tissue specific manner extended loop region is known to interact with the NAD+ binding
(Hirschey et al., 2011; Hebert et al., 2013). It is expressed maximally in pocket of large domain while N terminal extension help maintains
metabolically active tissues like brain, heart, kidney, liver, brown adi- substrate specificity (North and Verdin, 2004).
pose tissue and skeletal muscles (Onyango et al., 2002; Schwer et al., Despite some initial controversies about subcellular localization of
2002; Shi et al., 2005). In particular, SIRT3 expression in the adult SIRT3, majority of evidences suggest mitochondrial matrix as primary

Table 3
Sirtuins in Brain: Brain cellular specificity and implication in age related neurodegenerative disorder.

SIRTUINS Brain cell specificity Age Related Brain Disorder References

SIRT1 Neurons and astrocyte AD, PD, HD, ALS, Wallerian degeneration, Multiple Duan 2013; Braidy et al., 2015
sclerosis
SIRT2 Myelin producing cells; Oligodendrocytes and schwann ↓Gliomas; Its inhibition prevents AD, PD, HD Michan and Sinclair, 2007; de Oliveira et al.,
cells 2012
SIRT3 Neurons, rare in gliocytes AD,PD,HD, ALS, ARHL Herskovits and Guarente, 2013; Huang et al.,
2016
SIRT4 Detected in Glial cells (astrocytes) Unknown Komlos et al., 2013
SIRT5 Detected in neuron and astrocyte Unknown Braidy et al., 2015
SIRT6 Neuron and astrocyte AD Braidy et al., 2015
Kaluski et al., 2017
SIRT7 Detected in neuron and astrocyte Unknown Braidy et al., 2015

45
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

Fig. 2. SIRT3 functional domains: SIRT3 is synthe-


sized as a 44 kDa protein and remains inactive due to
the presence of additional N terminal sequences. The
first 25 aa N-terminal sequences serve as MLS which,
after entry into mitochondria, gets cleaved by a mi-
tochondrial matrix peptidase at a conserved site
around 142 aa to generate an active 28 kDa mi-
tochondria active SIRT3. A stretch between positions
101–118 is reported to contain ∼6 ser phosphor-
ylation sites which are speculated to prevent sub-
strate binding to SIRT3 in a phosphorylation status
dependent manner before it gets transported to the
organelle. A catalytic core domain of ∼275 aa is
comprised of one NAD+ binding Rossmann fold, a
substrate protein binding site and a groove of con-
necting cleft between these two substrate binding
sites. Many regulatory sites including Zn2+ binding
+
sites are also located within the catalytic domain. The C terminal extended region is known to interact with the NAD binding pocket of the catalytic domain while N terminal extension is
known to help maintain substrate specificity.

site of SIRT3 activity (Lombard et al., 2007; Cooper and Spelbrink, evidently promotes mitochondrial biogenesis and ROS detoxification by
2008; Sundaresan et al., 2008). Nonetheless, short length and full deacetylating many protein substrates. As an immediate action, SIRT3
length SIRT3, found in humans and mice, as two different isoforms, deacetylates Mn-SOD and increases its activity to dismutate oxygen free
appear to be differentially distributed in a tissue-specific manner (Scher radicals. Also, SIRT3 protects cells during oxidative stress by preventing
and Vaquero, 2007). As depicted in Fig. 2, SIRT3 is synthesized as a mPTP (mitochondrial permeability transition pore) opening via deac-
44 kDa full length inactive protein with a stretch of N terminal 142 aa, tivating Cyclophilin D (Cyp D) which blocks the release of GSH and cyt
The first 25 aa sequences act as mitochondrial localizing sequences c and thereby prevents cell apoptosis (Bause and Haigis, 2013). In ad-
(MLS) (Onyango et al., 2002). After this protein enters into the mi- dition, SIRT3 is also known to deacetylate FOXO3a and facilitates its
tochondrion, the MLS stretch is removed by a mitochondrial matrix nuclear translocation which in turn accounts for increased transcription
peptidase at a conserved site around 142 aa resulting into generation of of the antioxidant enzymes (Jacobs et al., 2008; Tseng et al., 2013).
an active 28 kDa short form of SIRT3 (Schwer et al., 2002; Scher and Furthermore, PGC-1α is known as a primary regulator of genes in-
Vaquero, 2007). It has been reported that this N-terminal additional volved in mitochondrial biogenesis, metabolism, effective ROS meta-
stretch might regulate the access of protein substrates to the active site bolism and stress responses (Herzig et al., 2001; Yoon et al., 2001;
of the enzyme (Schlicker et al., 2008) and thus, imparting its functional Palacios et al., 2009). It has been described that PGC-1α, arbitrated by
inactivation before it reaches to the mitochondrial matrix. Additionally, ERRα, stimulates SIRT3 expression by binding to the SIRT3 promoter
high-resolution mass spectrometry-based phosphor-proteome analysis region (Kong et al., 2010; Giralt et al., 2011; Ansari et al., 2017).
has identified six phosphorylated serine residues between positions Satterstrom et al. (2015) proposed that NRF-2 (Nuclear Respiratory
101–118 closer to the mitochondrial cleavage site (Olsen et al., 2010) Factor-2) could be a novel regulator of SIRT3 expression. Notably, NRF-
and thus, suggesting for phosphorylation status dependent prevention 2 is already known to be co-activated by PGC-1α, leading towards en-
of substrate binding to SIRT3 active site before its mitochondrial entry hanced expression of its target genes (Mootha et al., 2004; Baldelli
(Flick and Lüscher, 2012). et al., 2013). Intriguingly, SIRT3, in turn, stimulates PGC-1α expression
involving CREB and AMPK feedback loop (Shi et al., 2005; Palacios
et al., 2009; Pillai et al., 2010b). Taking together, SIRT3 adapts more
2.2. SIRT3 and mitochondrial integrity
than one mechanism to protect mitochondria under metabolic stress
challenges.
It is known from ages that calorie restriction (CR), fasting or ex-
ercise boost cellular repair mechanism and endorse healthy lifespan.
During recent past, some studies have shown that CR promotes dea- 2.3. SIRT3 and energy metabolism
cetylation of many mitochondrial proteins by upregulating SIRT3 ex-
pression resulting into increased oxidative phosphorylation and indu- In general, SIRT3 is activated with increased NAD+/NADH ratio
cing multimodal adaptations in mitochondrial functions (Lombard and regulate complex interactions between metabolic pathways in a
et al., 2007; Palacios et al., 2009). Mitochondria are the primary site of multimodal way. SIRT3 is reported to up regulate TCA cycle by acti-
generating ROS and thus become immediate target of oxidative da- vating PDH (pyruvate dehydrogenase complex), the first enzyme that
mage. SIRT3 is now emerging as an important regulator of mitochon- catalyzes pyruvate entry into oxidative energy production pathway.
drial antioxidant defense mechanism (Chen et al., 2014) and it does so And it does so mainly by deacetylating E1α subunit (Jing et al., 2013).
by employing more than one mechanisms. SIRT3 mediated activation of In addition, it targets almost all critical TCA cycle enzymes (Schwer and
mitochondrial Mn-SOD is considered to be the most plausible one (Qiu Verdin, 2008) and has been described to physically associate with and
et al., 2010). It is now becoming clearer that CR and ROS imbalance vis deacetylate NADH dehydrogenase (ubiquinone) 1 α sub complex 9
a vis SIRT3 level are mutual in maintaining mitochondrial structure and (NDUFA9) (Ahn et al., 2008; Lombard et al., 2011). SIRT3 also interacts
functions. with 2 subunit of Fo-F1 ATP synthase leading to deacetylation of alpha
As illustrated in Fig. 3, the bioenergetic deficit reflected by in- and OSCP subunits (Law et al., 2009; Duan, 2013). In addition, SIRT3 is
creased AMP/ATP ratio activates AMPK/CREB signaling to induce likely to regulate expression of some of the sub units of complex IV. It
SIRT3 expression by involving PGC-1α (peroxisome- proliferator acti- has been reported that PGC-1α induced expression of complex IV sub-
vated receptor gamma co activator 1-alpha) and nuclear respiratory units is decreased in SIRT3 knockdown condition (Bause and Haigis
factor 2 (NRF2)/estrogen related receptor α (ERRα) trans activation et al., 2013). Thus, it is evident that SIRT3 activity critically associates
mechanisms (Kong et al., 2010; Giralt et al., 2011; Satterstrom et al., with the mitochondrial ATP synthesizing machinery (Fig. 3).
2015; Ansari et al., 2017). In addition to its role in enhancing nuclear SIRT3 is also involved in activating alternate energy pathways like,
transcription of ROS detoxification genes (Shi et al., 2005) in a feed- fatty acids oxidation. In fact SIRT3 deficient mice are reported to have
back loop mechanism, after it enters into mitochondria, SIRT3 reduced ATP level, defect in thermogenesis and hypoglycemia during

46
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

Fig. 3. SIRT3 and mitochondrial integrity: Though complete signaling


cascades of the mitochondrial SIRT3 regulation remain unexplored,
based on some recent evidences, it is suggested that increased AMP/
ATP ratio, as a marker of bioenergetic deficit, could activate AMPK/
CREB signaling resulting into induction of SIRT3 expression by trans
activation of PGC-1α and NRF2/ERRα. After SIRT3 enters into mi-
tochondria, it is known to promote mitochondrial ROS detoxification
by deacetylating many proteins; including Mn-SOD, most of the TCA
cycle enzymes, ETS assemblies and Fo-F1 ATPase subunits, resulting
into increased rate of oxidative phosphorylation. It also inhibits mPTP
opening via deactivating Cyp D which blocks the release of GSH and
cyt c and thereby prevents cell apoptosis. In addition, SIRT3 is known
to deacetylate mitochondrial FOXO3a and facilitates its nuclear
translocation for enhanced transcription of the antioxidant enzymes.
In addition, SIRT3 is also found to activate alternate energy pathways
like, fatty acids oxidation and lactate utilization.

starvation (Hirschey et al., 2010). SIRT3 deacetylates and activates long post synaptic membrane receptors; Metabotropic (mGlu 1–5) and io-
chain acyl CoA dehydrogenase and 3-Hydroxy-3-Methylglutaryl-CoA notropic receptors (NMDA, AMPA and Kainate receptor). Under normal
Synthase 2 (HMGCS2) and thus promotes utilization of fat as gluco- physiological conditions, glutamate released into the synaptic cleft
neogenic source (Shimazu et al., 2010). Also, SIRT3 promotes gluco- binds to the post synaptic NMDA receptor resulting into release of
neogenesis from amino acids by deacetylating and activating glutamate Mg2+ blockage and opening of the Ca2+ channel and thereby initiating
dehydrogenase (GDH) that converts glutamate into the TCA cycle in- Ca2+ dependent intracellular signaling (Spandou et al., 2007; Chen and
termediate α-ketoglutarate (Schlicker et al., 2008). SIRT3 also aug- Lipton, 2006). AMPA receptors are usually co-expressed with the
ments glycolysis by deacetylating Cyclophilin D (Cyp D) which on one NMDA receptors and they jointly process the synaptic functions like;
hand keeps hexokinase II inactive and on the other hand prevents its potentiation, learning and memory & excitotoxicity. Prolonged depo-
association with ANT and thereby, blocks mPTP formation (Shulga larization of the postsynaptic neurons is associated with the deranged
et al., 2010). glutamatergic activity which in turn is likely to induce excitotoxic
neuronal derangement in a variety of neurodegenerative diseases such
2.4. Exogenous modulation of SIRT3 as stroke, Alzheimer’s disease and Parkinson’s disease (Nicholas and
Rothstein, 2006).
Though information is limited about SIRT3 specific modulators Fig. 4 illustrates the most plausible neurochemical events that ul-
during diseased conditions, some studies describe that resveratrol, a timately converge at neuronal mitochondria to induce excitotoxicity led
polyphenolic compound and a known activator of SIRT1, could also neurodegeneration. The probable role of mitochondrial SIRT3 in neu-
activate SIRT3 (Schirmer et al., 2012). In particular, a dimer of this rodegeneration is an evolving concept backed up by some recent re-
compound, a trans-(−)-є-Viniferine, was found to show strong neuro- ports, using neuronal cell culture model, that describe prevention of
protective effects by activating SIRT3 (Fu et al., 2012). Another excitotoxicity in the SIRT3 over expressing neuronal cells (Kim et al.,
bioactive compound, roxylin A, was demonstrated to activate SIRT3 in 2011; Han et al., 2014). This goes in line with some earlier reports as
human breast cell carcinoma (Wei et al., 2013). Rhamnetin, an o-me- well which described the role of SIRT3 in maintaining almost every
thylated flavonoid and Honokiol, derived from the bark of magnolia aspects of mitochondrial function like, energy metabolism, biogenesis
trees, also showed cardio protective effects by activating SIRT3 (Park (Shi et al., 2005; Ahn et al., 2008) and protection from oxidative stress
et al., 2014; Pillai et al., 2015). led induction of cell death mechanisms (Sundaresan et al., 2009).
So far SIRT3 inhibitors are concerned, apart from the NAM, a known Excitotoxicity associated with glutamatergic system mainly involves
noncompetitive inhibitor of the Sirtuin family enzymes, 5-amino-2 increased Ca2+ influx that activates neuronal nitric oxide synthase
phenyl-benzoxazole have been reported as a novel and specific SIRT3 (nNOS) to produce excess of NO and other RNS (reactive nitrogen
inhibitor (Chen et al., 2014). These findings, thus, do argue that SIRT3 species). This in turn may adapt more than one mechanisms like; via
could be a relevant candidate to undergo modulation by exogenous altered NO signaling and/or by directly affecting mitochondrial bio-
factors including natural compounds in the conditions of brain disorder. chemistry for enhanced ROS production and declined energy status of
Indeed, regulation of SIRT3 activity by exogenous compounds could be the organelle. Together, such aberrant mitochondrial metabolism is
an important issue of future research. considered to open mPTP leading into cytochrome c release and in-
duction of apoptosis and thus drive the cell to undergo death process
(Lipton, 2008; Lee et al., 2009; Nicholls, 2009). Moreover, as depicted
3. SIRT3 and excitotoxicity
in Fig. 4 and as described explicitly in the previous text (2.3), all such
mitochondrial derangements could be prevented due to over expression
Excitotoxicity refers to the prolonged activation of the excitatory
and sustained activity of SIRT3. Thus, it is speculated that during glu-
amino acid receptors in the brain leading to neuronal damage or death
tamate excitotoxicity, neuronal cell mitochondria might be challenged
(Lipton, 2008; Vincent and Mulle, 2009). Glutamate is the principle
with a declined level of SIRT3 as well.
excitatory neurotransmitter involved in regulating synaptic plasticity,
To bring SIRT3 level down, the excitotoxic cells might adapt two
learning, memory and other cognitive functions. However, when pro-
mechanisms; one, the Ca2+ load dependent altered NO signaling may
duced in excess, it may develop a condition of neuro-excitotoxicity in
significantly affect nuclear activity responsible to transcribe SIRT3 and/
the post synaptic neurons. Glutamate acts through two major classes of

47
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

Fig. 4. SIRT3, excitotoxicity and neurodegeneration: Glutamatergic


excitotoxicity gets induced mainly due to over activation of NMDAR
at postsynaptic neurons. The resultant influx of Ca2+ is known to
activate nNOS in those neurons, which in turn may adapt more than
one mechanism of NO dependent alterations in neuronal biochem-
istry. Over production of NO in combination with ROS load has been
considered a common pathogenic mechanism of most of the neuro-
degenerative brain disorders. Since neurodegeneration finally begins
due to mitochondrial dysfunction led apoptosis, as depicted in Fig. 3
about SIRT3 dependent prevention of mitochondrial derangement, it
is argued that SIRT3 could also play a significant role in the patho-
physiology of neurodegeneration. Though this aspect needs to be ex-
plored in detail, based on some recent reports, it is speculated that
NMDAR over activation induced nNOS activation could either affect
nuclear mechanism to decline SIRT3 level or could directly, via NO &
ROS interaction, may decrease SIRT3 activity. Since SIRT3 activity
correlates with preventing events leading towards mPTP opening,
cytochrome c release and induction of apoptosis, it is argued that
SIRT3 either alone or in combination with enhanced RNS & ROS load,
might be implicated in the pathogenesis of the neurodegenerative
brain disorders. The argument gets support from some findings that
describe that over expression and activation of SIRT3 could protect
neurons from undergoing neurodegenerative changes.

or the mitochondrial transport of the SIRT3 protein. Second, the altered function. Thus, strongly advocate about signficant roles of SIRT3 in the
NO signaling and cumulative RNS & ROS load in mitochondria together pathogenesis of AD.
may decline SIRT3 activity and thereby depriving mitochondria from its Indeed some studies report roles of SIRT3 in AD at different stages of
endogenous protection mechanisms. Although, to delineate the me- the disease progression. For example, in a primary cortical neurons
chanism by which glutamate excitotoxicity declines SIRT3 activity is a model, neurons were induced to undergo apoptosis after treatment with
relevant hot spot for future research, some reports do suggest that over beta-amyloid (Aβ) in a manner similar to the Alzheimer’s disease (AD).
expression and activation of SIRT3 could protect neurons from under- However, co-treatment with pituitary adenylate cyclase-activating
going neurodegenerative changes (Kim et al., 2011; Fu et al., 2012; Han polypeptide (PACAP) could rescue these neurons mainly by upregu-
et al., 2014). lating SIRT3 expression (Han et al., 2014). Also, the PACAP mediated
Importantly, some reports from recent past, do advocate that neu- neuroprotective effect was found to be lost in the condition of SIRT3
ronal excitotoxicity constitutes a common neurochemical event asso- knocked down by shRNA in primary cultured neurons (Han et al.,
ciated with pathogenesis of most of the neurodegenerative brain dis- 2014). Furthermore, SIRT3 overexpression has been reported to confer
orders and with the brain dysfunctions developed due to neuro- resistance against oxidative stress and thereby extended neuronal
physiological challenges. The most important ones are; Alzheimer’s longevity in primary hippocampal cultures (Weir et al., 2012). Apoli-
disease, Parkinson’s disease, Huntington’s disease, ALS, stroke etc poprotein E4 (APOE4) is a major genetic factor associated with late-
(Raymond et al., 2011; Cozzolino and Carrì, 2012; Salvatore et al., onset of AD and it has been reported that SIRT3 is down regulated in
2012; Ong et al., 2013; Kalogeris et al., 2014; Prentice et al., 2015) and the frontal cortices of APOE4 carrier human brain as compared to the
metabolic brain disorders like hepatic encephalopathy (HE) (Singh and non-carrier individuals (Yin et al., 2015a; Ansari et al., 2017). Yin et al.
Trigun, 2014; Mondal and Trigun, 2015). Therefore, for clarity about (2015a) have further proposed that SIRT3 level may aid in the diagnosis
roles of SIRT3 in the pathogenesis of a specific brain disorder and for of AD.
SIRT3 associated different neuronal/molecular markers and substrates,
the text on SIRT3 and common neurodegenerative brain disorders are 3.2. SIRT3 and Huntington’s disease
being summarized in a disease specific manner (Fig. 4).
Huntington’s disease (HD) is an inherited, autosomal dominant
3.1. SIRT3 and Alzheimer’s disease neurodegenerative disorder characterized by cognitive dysfunction, loss
of coordination and motor functions. It is caused when the gene for the
Alzheimer’s disease (AD) is a complex, late-onset, progressive neu- huntingtin (Htt) protein, located at 4p16.3, shows an expansion of CAG
rodegenerative disorder associated with memory impairment and se- repeat that codes for a stretch of glutamine residues, affecting the
vere dementia (Farooqui, 2010). AD is characterized by accumulation protein conformation resulting into aggregation of the mutant Htt in
of amyloid β peptide (Aβ) neuritic plaques, neurofibrillary tangles, nucleus and cytoplasm of the affected neurons (Arrasate and
synaptic failure and mitochondrial dysfunction. Although the under- Finkbeiner, 2012). Normal expression of Htt is reported throughout the
lying molecular mechanism for AD pathogenesis is not clearly eluci- body and is required for gene transcription, protein trafficking and
dated, but it is believed that Aβ mediated oxidative stress, induction of mitochondrial function (Arrasate and Finkbeiner, 2012). But the mu-
neuroinflammation and abnormal glutamate metabolism could be im- tant Htt (mHtt) in the brain, particularly in the caudate-putamen
plicated in onset and progression of this disease (Kontush, 2001; (striatum) and cortex, leads to neurodegeneration by implicating in-
Butterfield, 2002). creased glutamatergic signaling and mitochondrial dysfunction in the
AD is an example of slow excitotoxicity (Ong et al., 2013) where corticostriatal neurons (Raymond et al., 2011).
NMDA receptor overactivation is found to be in tonic rather than in Notably, cells expressing mHTT have been reported to have de-
phasic manner which allows glutamate to become neurotoxic at con- clined SIRT3 levels. This reduced SIRT3 activity levels was recovered
centrations that normally shows no toxicity (Putcha et al., 2011). Also, after ε-viniferin, a stilbene resveratrol dimer, activation of SIRT3 (Fu
mitochondrial dysfunction associated with AD involves Aβ entry into et al., 2012). This SIRT3 activator increases SIRT3 protein level and
the mitochondria leading to disruption of ETC, generation of ROS and activates AMPK by SIRT3-dependent deacetylation of AMPK kinase.
decreased ATP production (Mungarro-Menchaca et al., 2002; Manczak Also it activates liver kinase B1 (LKB1), also known as serine/threonine
et al., 2006). Taking together, all these precipitating events either ori- kinase 11 (STK11), which in turn enhances PGC-1α level and promotes
ginate from mitochondria and/or converge to derange mitochondrial mitochondrial biogenesis, electron transport activity, ROS

48
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

detoxification mechanisms and preserves NAD+/NADH ratio in mHtt ALS, effectively slows down the disease progression and prolongs sur-
cells (Fu et al., 2012). SIRT3 also directly interacts with SOD2 and vival by inhibiting glutamate release and its receptor (Lacomblez et al.,
enhances its antioxidant activity. This SIRT3 mediated neuroprotective 1996).
role of viniferin was confirmed by siRNA induced SIRT3 knockdown in Involvement of altered mitochondrial function during ALS patho-
mHtt expressing striatal cell culture (Fu et al., 2012; Herskovits and genesis is evident from the mutant SOD1 accumulation in the ALS pa-
Guarente, 2013). tients (Cozzolino and Carrì, 2012; Tan et al., 2014). The role of SIRT3 in
preserving mitochondrial function was demonstrated against mi-
3.3. SIRT3 and Parkinson’s disease tochondrial fragmentation led neuronal damage in spinal cord motor
neurons transfected with ALS-typical mutant G93A- SOD1 (Song et al.,
PD is an age related movement disorder causing tremor, rigidity, 2013). Another instance of SIRT3 modulation in ALS was recorded
bradykinesia and postural instability resulting from degeneration of the when the primary astrocytes from G93A-SOD1 transgenic mice induced
dopaminergic neurons within the basal ganglia circuitry, specifically motor neuron death in co-culture but this effect was found to be de-
the substantia nigra pars compacta (SNpc) and accumulation of Lewy clined due to the co-culture of the motor neurons with the astrocytes
bodies (inclusions that contain α-synuclein and ubiquitin) in the cyto- which over expressed SIRT3 (Harlan et al., 2016).
plasm (Blesa et al., 2012). The basal ganglion receives two major glu-
tamatergic innervations: from the subthalamic nucleus (STN), the only 3.5. SIRT3 and stroke
excitatory nucleus of the circuit, and the motor cortex. SNpc receives
secondary Glutamatergic projection from amygdala and laterodorsal Stroke is a neurological disorder which occurs due to the neuronal
tegmental nuclei (Misgeld, 2004). loss resulting from interrupted blood supply to brain and ultimately
There are evidences that suggest glutamate excitotoxicity as well depriving brain from oxygen and glucose supply. Ischemic stroke is a
during altered neurotransmitter function in PD, e.g, changes in the complex entity and one of the leading causes of disability and death
glutamate transporters (Raju et al., 2008; Salvatore et al., 2012) as well worldwide (Prabhakaran et al., 2015). Although the exact pathological
as altered subunit composition and phosphorylation pattern of mechanism remains intangible, glutamate mediated excitotoxicity is
NMDARs (Dunah et al., 2000; Xu et al., 2012) in the basal ganglia of believed to play a central role in cerebral hypoxia and ischemia led
different animal models of PD. Importantly, these changes varied ac- neuronal death (Chen et al., 2011). Evidences suggest that cerebral
cording to the degree of striatal denervation and loss of endogenous ischemia causes increased glutamate release in the intracerebral sy-
dopamine (Ambrosi et al., 2014). The vulnerability of dopaminergic napses and thereby elicits prolonged activation of postsynaptic neurons
neurons may be contributed by glutamatergic pathways that associate accompanied with oxidative stress, energy deficit and calcium overload
with the disruption of mitochondrial and bioenergetic homeostasis, leading to mitochondrial dysfunction and induction of apoptotic sig-
compromised antioxidant defense and deranged proteolytic machinery naling cascade (Bosley et al., 1983; Dawson et al., 2000; Kalogeris et al.,
(Ambrosi et al., 2014). 2014).
Since SIRT3 is involved in regulating ROS balance and ATP pro- Mitochondrial involvement in the stroke pathology has been de-
duction and imbalance of these two factors act as key mediators of scribed by various studies; however, information is scanty on the role of
neuronal injury during neurodegenerative diseases, studies on the role SIRT3 in this disease. Moreover, a recent report on the neuroprotective
SIRT3 in PD pathogenesis derive much scientific merit. Some of the effects of SIRT3 in stroke by Yin et al. (2015b) provides a strong support
studies demonstrated that absence of SIRT3 does not elicit anxiety/ in this context. They found that administration of ketones; beta-hy-
depression like behavior and motor dysfunctions, but its deficiency did droxybutyrate (BHB) and acetoacetate (ACA) to mice 30 min after
aggravate dopaminergic neuronal loss in the substantia nigra pars ischemia induced SIRT3-FoxO3a-SOD2 pathway which in turn could
compacta and striatum of MPTP (1-methyl-4-phenyl-1,2,3,6-tetra- reduce oxidative stress and enhanced ETC Complex I activity resulting
hydropyridine) induced PD mice (Liu et al., 2015). MPTP treatment into reduction of infarct volume and improvement of neurological
caused reduced mitochondrial ROS defense capacity with decreased functions. The protective effect of SIRT3 is also demonstrated in oxygen
SOD2 and GPx expression, however, their expression levels were found and glucose deprivation (OGD) model of neuronal ischemia where over
to be further reduced in the absence of SIRT3 (Liu et al., 2015). A si- expression of SIRT3 through lentivirus transfection not only resulted in
milar study demonstrated that SIRT3 plays neuroprotective role in ro- declined oxidative stress associated with OGD but also could maintain
tenone-induced PD cell model wherein, SIRT3 knockout aggravated mitochondrial functions and mitochondrial membrane potential
SOD and GSH reduction, loss of mitochondrial membrane potential and through modulating AMPK-mTOR pathway (Dai et al., 2017). Ad-
worsened rotenone induced cells death. On the other hand, SIRT3 over ditionally, SIRT3 has been found to be involved in suppressing neu-
expression could rescue the cells from α-synuclein accumulation, re- roinflammation in post cardiopulmonary bypass and ischemic re-
duced antioxidant defense by increasing SOD and GSH level and perfusion cerebral injury. Report describes that SIRT3 knockdown is
thereby preventing derangement of mitochondrial membrane potential associated with worsen neurological function due to the increased
(Zhang et al., 2016a). cerebral level of active acetylated form of NLRP3 and NFƙB along with
declined activity of MnSOD during such cerebral injury (Zhang et al.,
3.4. SIRT3 and amyotropic lateral sclerosis (ALS) 2016b). Taking together, these findings strongly argue for the detailed
studies on identification of other key players of such SIRT3 dependent
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease manifestation of stroke pathology.
characterized by progressive loss of upper motor neurons in the cortex
and also lower motor neurons in the brainstem and spinal cord. The 3.6. SIRT3 and hepatic encephalopathy: an evolving concept
selective loss of neurons results in progressive paresis of bulbar, re-
spiratory and limb muscles without dilapidating sensory and cognitive Unlike other neurodegenerative diseases, hepatic encephalopathy
functions (Van Damme et al., 2005). ALS is a complex multi-factorial (HE) is not an age related brain disorder. HE is a metabolic brain dis-
syndrome which is found to be associated with RNA dysregulation, order characterized by variable degree of motor and cognitive deficits
protein misfolding, oxidative damage, defective axonal transport, mi- due to liver dysfunction (Bajaj et al., 2009; Felipo et al., 2012; Singh
tochondrial dysfunction and excitotoxicity (Cozzolino and Carrì, 2012). et al., 2014). The pathology of this neuropsychiatric complication is
The deleterious glutamatergic signaling during ALS is evident from the associated with peripheral inflammation and hyperammonemia led
electrophysiological study demonstrating alterations in the cortical and glutamate excitotoxicity by over activating NMDAR (Monfort et al.,
spinal neurons (Eisen, 2009). Also, Riluzole, the only effective drug in 2002; Cauli et al., 2002; Mondal and Trigun 2014). Over activation of

49
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

the receptor leads to delayed opening of the ion channel and increased associated signaling steps during AD, PD & Hutington’s disease
influx of Ca2+ which allows activation of calcium-dependent signaling (Herskovits and Guarente, 2013) do argue for a crucial role of SIRT3
cascades wherein, nNOS acts as a major mediator of deranged neuronal as well and thus, inviting special scientific focus to delineate SIRT3
functions developed due to NMDAR over activation (Singh and Trigun, signaling associated with neurodegeneration.
2010; Mondal and Trigun, 2015). 3. The cause and effect relationship of SIRT3 vs excitotoxic and neu-
Importantly, at downstream level, NMDAR over activation also re- rodegenerative brain disorders is still in its infancy stage. This needs
sults into mitochondrial dysfunction, bioenergetic deficit, oxidative and to be given due focus, because this is likely to provide end point
nitrosative stresses (Singh et al., 2008; Singh and Trigun, 2010; Felipo evaluation parameters about modulation of SIRT3 activity by small
et al., 2012; Mehrotra and Trigun, 2012; Singh et al., 2014). Though molecule and also for understanding the reverse pharmacology of
information is lacking on implication of Sirtuins in the pathogenesis of plants derived neuroprotectants.
HE, however, a recent report, on the protective effects of resveratrol, a
known sirtuins activator, against the hyperammonemia induced pro- Declaration
inflammatory and pro-apoptotic conditions in brain of the HE rats
(Khanna and Trigun, 2016), does provide strength for a role of Sirtuins The authors declare no conflict of interest.
in HE pathogenesis. Thus, since SIRT3 is known to maintain mi-
tochondrial integrity, it is arguable to speculate a significant role of this Acknowledgments
mitochondrial Sirtuin in the pathogenesis of HE. Accordingly, SIRT3
may also be speculated as a relevant therapeutic target against such This review originated due to research work on SIRT1 financed by
excitotoxic brain disorders. an ICMR project (P-54/38/GFPGER/2011/NCD-II and currently the
research on SIRT3 is being financially supported by a DST project (ER/
4. Concluding remarks 2016/006501/AS). The award of UGC merit fellowship to Anamika and
Lady Tata memorial Trust JRF/SRF to Archita Khanna are also ac-
Sirtuins, by deacylating an array of non-histone proteins, constitute knowledged. The facilities of DST-FIST & UGCeCAS programme in the
an important aspect of epigenetic regulation of metabolic integrity, cell Department of Zoology and from DBT-BHU ISLS have been of great
survival and cellular homeaostasis in normal and in diseased condi- support.
tions. As such it has now invited much attention as a central molecule of
concern for therapeutic management of many crucial human ailments References
like, cancer and cardio vascular diseases. Moreover, implication of
Sirtuins in the pathogenesis of neurological complications is a relatively Ahn, B.H., Kim, H.S., Song, S., Lee, I.H., Liu, J., Vassilopoulos, A., Deng, C.X., Finkel, T.,
less explored area. Nonetheless, the information available so far, from 2008. A role for the mitochondrial deacetylase Sirt3 in regulating energy home-
ostasis. Proc. Natl. Acad. Sci. U. S. A. 105 (38), 14447–14452.
the recent researches, suggest that this class of deacetylases are likely to Ambrosi, G., Cerri, S., Blandini, F., 2014. A further update on the role of excitotoxicity in
play crucial roles in protecting neurons from undergoing neurochemical the pathogenesis of Parkinson’s disease. J. Neural Trans. (Vienna) 121 (8), 849–859.
derangements during most of the brain disorders developed due to Anderson, R.M., Shanmuganayagam, D., Weindruch, R., 2009. Caloric restriction and
aging: studies in mice and monkeys. Toxicol. Pathol. 37, 47–51.
neuro-excitotoxicity led neurodegeneration. Though much of informa- Ansari, A., Rahman, M., Saha, S.K., Saikot, F.K., Deep, A., Kim, K.H., 2017. Function of
tion available so far have been derived from in vitro and/or primary the SIRT3 mitochondrial deacetylase in cellular physiology, cancer, and neurode-
neuronal culture models, but as described in a review from Herskovits generative disease. Aging Cell. 16, 4–16.
Arrasate, M., Finkbeiner, S., 2012. Protein aggregates in Huntington’s disease. Exp.
and Guarente (2013), it is evident that certain specific Sirtuins do Neurol. 238 (1), 1–11.
modulate even some signaling steps of the AD, PD and Hutington’s Bajaj, J.S., Wade, J.B., Sanyal, A.J., 2009. Spectrum of neurocognitive impairment in
disease pathogenesis. Additionally, it is now being realized that there is cirrhosis: implications for the assessment of hepatic encephalopathy. Hepatology 50
(6), 2014–2021.
a need to target organelle specific Sirtuins homologues for under-
Baldelli, S., Aquilano, K., Ciriolo, M.R., 2013. Punctum on two different transcription
standing brain disorder pathogenesis and examining them as relevant factors regulated by PGC-1a: nuclear factor erythroid-derived 2-like 2 and nuclear
therapeutic target. respiratory factor 2. Biochim. Biophys. Acta 1830, 4137–4146.
Out of the 7 Sirtuins identified, SIRT3, due to its location and ac- Bause, A.S., Haigis, M.C., 2013. SIRT3 regulation of mitochondrial oxidative stress. Exp.
Gerontol. 48 (7), 634–639.
tivity in mitochondria, has now become a point of neurological con- Berger, F., Lau, C., Dahlmann, M., Ziegler, M., 2005. Subcellular compartmentation and
cern. This is because, most of the neurodegenerative brain disorders differential catalytic properties of the three human nicotinamide mononucleotide
implicate neuro-excitotoxicity whose pathogenic mechanisms either adenylyltransferase isoforms. J. Biol. Chem. 280, 36334–36341.
Blesa, J., Phani, S., Jackson-Lewis, V., Przedborski, S., 2012. Classic and new animal
originate from and/or converge at mitochondrial derangements in the models of Parkinson’s disease. J. Biomed. Biotechnol. 2012, 845618.
post synaptic neurons challenged with excessive excitatory activity. Bosley, T.M., Woodhams, P.L., Gordon, R.D., Balázs, R., 1983. Effects of anoxia on the
Since, SIRT3 is now considered critical in maintaining mitochondrial stimulated release of amino acid neurotransmitters inthe cerebellum in vitro. J.
Neurochem. 40 (1), 189–201.
integrity; it deserves special focus for understanding pathogenesis of the Braidy, N., Poljak, A., Grant, R., Jayasena, T., Mansour, H., Chan-Ling, T., Smythe, G.,
neurodegenerative brain disorders and hence evaluating it as a target of Sachdev, P., Guillemin, G.J., 2015. Differential expression of sirtuins in the aging rat
therapeutic management of such neurological complications. brain. Front. Cell. Neurosci. 9, 167.
Brown, K., Xie, S., Qiu, X., Mohrin, M., Shin, J., Liu, Y., Zhang, D., Scadden, D.T., Chen,
Some reports, though fragmentary, do emphasize about the roles of
D., 2013. SIRT3 reverses aging-associated degeneration. Cell Rep. 3, 319–327.
SIRT3 in neuroprotection against certain neurodegenerative conditions Butterfield, D.A., 2002. Amyloid beta-peptide(1–42) induced oxidative stress and neu-
and thus, advocate about multimodal research on SIRT3 biochemistry rotoxicity: implications for neurodegeneration in Alzheimer’s disease brain A review.
Free Radical Res. 36, 1307–1313.
in the relevant animal models of neurodegenerative brain disorders.
Cantó, C., Auwerx, J., 2012. Targeting sirtuin 1 to improve metabolism: all you need is
The current research so far has ably recorded identification of meta- NAD+? Pharmacol. Rev. 64 (1), 166–187.
bolic protein targets for SIRT3 and its regulation in maintaining mi- Carafa, V., Nebbioso, A., Altucci, L., 2012. Sirtuins and disease: the road ahead. Front.
tochondrial metabolism and ROS balance at cellular level. As such these Pharmacol. 3.
Cauli, O., Rodrigo, R., Llansola, M., Montoliu, C., Monfort, P., Piedrafita, B., El Mlili, N.,
reprots invite and open at least 3 fundamental aspects of SIRT3 biology Boix, J., Agustí, A., Felipo, V., 2002. Glutamatergic and gabaergic neurotransmission
in the context of brain biochemistry: and neuronal circuits in hepatic encephalopathy. Metab. Brain Dis. 24 (1), 69–80.
Chen, H.S., Lipton, S.A., 2006. The chemical biology of clinically tolerated NMDA re-
ceptor antagonists. J. Neurochem. 97 (6), 1611–1626.
1. Regulation of nuclear transcription of SIRT3 and characterization of Chen, S.D., Yang, D.I., Lin, T.K., Shaw, F.Z., Liou, C.W., Chuang, Y.C., 2011. Roles of
the factors accountable for its targeting to mitochondria in normal oxidative stress, apoptosis, PGC-1α and mitochondrial biogenesis in cerebral
and diseased brain cells ischemia. Int. J. Mol. Sci. 12 (10), 7199–7215. http://dx.doi.org/10.3390/
ijms12107199.
2. The report on the role of SIRT1 in regulating some of the disease

50
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

Chen, Y., Fu, L.L., Wen, X., Wang, X.Y., Liu, J., Cheng, Y., Huang, J., 2014. Sirtuin-3 hemorrhage. BioMed. Res. Int. 2016.
(SIRT3), a therapeutic target with oncogenic and tumor-suppressive function in Imai, S., Armstrong, C.M., Kaeberlein, M., Guarente, L., 2000. Transcriptional silencing
cancer. Cell. Death. Dis. 5 (2), e1047. and longevity protein Sir2 is an NAD- dependent histone deacetylase. Nature 403,
Cooper, H.M., Spelbrink, J.N., 2008. The human SIRT3 protein deacetylase is exclusively 795–800.
mito- chondrial. J. Biochem. 411, 279–285. Jacobs, K.M., Pennington, J.D., Bisht, K.S., Aykin-Burns, N., Kim, H.S., Mishra, M., Sun,
Cozzolino, M., Carrì, M.T., 2012. Mitochondrial dysfunction in ALS. Prog. Neurobiol. 97 L., Nguyen, P., Ahn, B.H., Leclerc, J., Deng, C.X., Spitz, D.R., Gius, D., 2008. SIRT3
(2), 54–66. interacts with the daf-16 homolog FOXO3a in the mitochondria, as well as increases
Dai, S.H., Chen, T., Li, X., Yue, K.Y., Luo, P., Yang, L.K., Zhu, J., Wang, Y.H., Fei, Z., Jiang, FOXO3a dependent gene expression. Int. J. Biol. Sci. 4 (5), 291–299.
X.F., 2017. SIRT3 confers protection against neuronal ischemia by inducing autop- Jiang, H., Khan, S., Wang, Y., Charron, G., He, B., Sebastian, C., Du, J., Kim, R., Ge, E.,
hagy: involvement of the AMPK-mTOR pathway. Free Radic. Biol. Med. 108, Mostoslavsky, R., Hang, H.C., 2013. Sirt6 regulates TNFα secretion via hydrolysis of
345–353. long chain fatty acyl lysine. Nature 496 (7443), 110.
Dawson, L.A., Djali, S., Gonzales, C., Vinegra, M.A., Zaleska, M.M., 2000. Jing, E., O'Neill, B.T., Rardin, M.J., Kleinridders, A., Ilkeyeva, O.R., Ussar, S., Bain, J.R.,
Characterization of transient focal ischemia-induced increases in extra cellular glu- Lee, K.Y., Verdin, E.M., Newgard, C.B., Gibson, B.W., Kahn, C.R., 2013. SIRT3 reg-
tamate and aspartate in spontaneously hypertensive rats. Brain Res. Bull. 253 (6), ulates metabolic flexibility of skeletal muscle through reversible enzymatic deace-
767–776. tylation. Diabetes 62 (10), 3404–3417.
de Oliveira, R.M., Sarkander, J., Kazantsev, A.G., Outeiro, T.F., 2012. SIRT2 as ther- Kalogeris, T., Bao, Y., Korthuis, R.J., 2014. Mitochondrial reactive oxygen species: a
apeutic target for age related diorders. Front. Pharmacol. 3 (82), 1–9. double edged sword in ischemia/reperfusion vs preconditioning. Redox. Biol. 2,
Donmez, G., Outeiro, T.F., 2013. SIRT1 and SIRT2: emerging targets in neurodegenera- 702–714.
tion. EMBO Mol. Med. 5, 344–352. Kaluski, S., Portillo, M., Besnard, A., Stein, D., Einav, M., Zhong, L., Ueberham, U.,
Du, J., Zhou, Y., Su, X., Yu, J.J., Khan, S., Jiang, H., Kim, J., Woo, J., Kim, J.H., Choi, B.H., Arendt, T., Mostoslavsky, R., Sahay, A., Toiber, D., 2017. Neuroprotective functions
He, B., 2011. SIRT5 is a NAD+ dependent protein lysine demalonylase and de- for the histone deacetylase SIRT6. Cell Rep. 18 (13), 3052–3062.
succinylase. Science 334 (6057), 806–809. Khanna, A., Trigun, S.K., 2016. Resveratrol normalizes hyperammonemia induced pro-
Duan, W., 2013. Sirtuins: from metabolic regulation to brain aging. Front. Aging inflammatory and pro-apoptotic conditions in rat brain. Int. J. Compl. Alt. Med. 4 (2),
Neurosci. 23 (5), 36. 00115.
Dunah, A.W., Wang, Y., Yasuda, R.P., et al., 2000. Alterations in subunit expression, Kim, S.H., Lu, H.F., Alano, C.C., 2011. Neuronal SIRT3 protects against excitotoxic injury
composition, and phosphorylation of striatal N-methyl-D-aspartate glutamate re- in mouse cortical neuron culture. PLoS One 6, e14731.
ceptors in a rat 6-hydroxydopamine model of Parkinson’s disease. Mol. Pharmacol. Kincaid, B., Bossy-Wetzel, E., 2013. Forever young: SIRT3 a shield against mitochondrial
57, 342–352. meltdown, aging, and neurodegeneration. Front. Aging Neurosci. 6 (5), 48.
Eisen, A., 2009. Amyotrophic lateral sclerosis-evolutionary and other perspectives. Komlos, D., Mann, K.D., Zhuo, Y., Ricupero, C.L., Hart, R.P., Liu, A.Y., Firestein, B.L.,
Muscle Nerve 40 (2), 297–304. 2013. Glutamate dehydrogenase 1 and SIRT4 regulate glial development. Glia 61 (3),
Farooqui, A.A., 2010. Neurochemical Aspects of Neuro-Traumatic and Neurodegenerative 394–408.
Diseases. Springer, New York. Kong, X., Wang, R., Xue, Y., Liu, X., Zhang, H., Chen, Y., Fang, F., Chang, Y., 2010. Sirtuin
Felipo, V., Urios, A., Montesinos, E., Molina, I., Garcia-Torres, M.L., Civera, M., Olmo, 3, a new target of PGC-1a, plays an important role in the suppression of ROS and
J.A., Ortega, J., Martinez-Valls, J., Serra, M.A., Cassinello, N., Wassel, A., Jorda, E., mitochondrial biogenesis. PLoS One 5, e11707.
Montoliu, C., 2012. Contribution of hyperammonemia and inflammatory factors to Kontush, A., 2001. Amyloid-beta: an antioxidant that becomes a pro-oxidant and criti-
cognitive impairment in minimal hepatic encephalopathy. Metab. Brain Dis. 27, cally contributes to Alzheimer’s disease. Free Radic. Biol. Med. 31, 1120–1131.
51–58. Lacomblez, L., Bensimon, G., Leigh, P.N., Guillet, P., Meininger, V., 1996. Dose-ranging
Flick, F., Lüscher, B., 2012. Regulation of sirtuin function by posttranslational mod- study of riluzole in amyotrophic lateral sclerosis: amyotrophic lateral Sclerosis/
ifications. Front. Pharmacol. 3, 29. Riluzole study group II. Lancet 347, 1425–1431.
Fritz, K.S., Galligan, J.J., Hirschey, M.D., Verdin, E., Petersen, D.R., 2012. Mitochondrial Law, I.K., Liu, L., Xu, A., Lam, K.S., Vanhoutte, P.M., Che, C.M., Leung, P.T., Wang, Y.,
acetylome analysis in a mouse model of alcohol-induced liver injury utilizing SIRT3 2009. Identification and characterization of proteins interacting with SIRT1 and
knockout mice. J. Proteome Res. 11, 1633–1643. SIRT3: implications in the anti-aging and metabolic effects of sirtuins. Proteomics 9
Fu, J., Jin, J., Cichewicz, R.H., Hageman, S.A., Ellis, T.K., Xiang, L., Peng, Q., Jiang, M., (9), 2444–2456.
Arbez, N., Hotaling, K., 2012. trans-(−)-e-Viniferin increases mitochondrial sirtuin 3 Lee, H.G., Zhu, X., Casadesus, G., Pallàs, M., Camins, A., O'Neill, M.J., Nakanishi, S.,
(SIRT3), activates AMP-activated protein kinase (AMPK), and protects cells in models Perry, G., Smith, M.A., 2009. The effect of mGluR2 activation on signal transduction
of Huntington Disease. J. Biol. Chem. 287, 24460–24472. pathways and neuronal cell survival. Brain Res. 244–250.
Giralt, A., Hondares, E., Villena, J.A., Ribas, F., Dıaz-Delfın, J., Giralt, M., Iglesias, R., Li, Y., Xu, W., McBurney, M.W., Longo, V.D., 2008. SIRT1 inhibition reduces IGF-I/IRS-2/
Villarroya, F., 2011. Peroxisome proliferator-activated receptor-c coactivator-1a Ras/ERK1/2 signaling and protects neurons. Cell Metab. 8 (1), 38–48.
controls transcription of the Sirt3 gene, an essential component of the thermogenic Lin, S.J., Defossez, P.A., Guarente, L., 2000. Requirement of NAD and SIR2 for life-span
brown adipocyte phenotype. J. Biol. Chem. 286, 16958–16966. extension by calorie restriction in Saccharomyces cerevisiae. Science 289, 2126–2128.
Haigis, M.C., Guarente, L.P., 2006. Mammalian sirtuins–emerging roles in physiology, Lipton, S.A., 2008. NMDA receptor activity regulates transcription of antioxidant path-
aging, and calorie restriction. Genes Dev. 20 (21), 2913–2921. ways. Nat. Neurosci. 11 (4), 381–382.
Haigis, M.C., Sinclair, D.A., 2010. Mammalian sirtuins: biological insights and disease Liu, L., Peritore, C., Ginsberg, J., Kayhan, M., Donmez, G., 2015. SIRT3 attenuates MPTP-
relevance. Annu. Rev. Pathol. 5, 253–295. induced nigrostriatal degeneration via enhancing mitochondrial antioxidant capa-
Hallows, W.C., Yu, W., Smith, B.C., Devries, M.K., Devires, M.K., Ellinger, J.J., Someya, city. Neurochem. Res. 40, 600–608.
S., Shortreed, M.R., Prolla, T., Markley, J.L., Smith, L.M., Zhao, S., Guan, K.L., Denu, Lombard, D.B., Zwaans, B.M.M., 2014. SIRT3: as simple as it seems? Geronto 60 (1).
J.M., 2011. SIRT3 promotes the urea cycle and fatty acid oxidation during dietary Lombard, D.B., Alt, F.W., Cheng, H.L., Bunkenborg, J., Streeper, R.S., Mostoslavsky, R.,
restriction. Mol. Cell 41, 139–149. Kim, J., Yancopoulos, G., Valenzuela, D., Murphy, A., Yang, Y., Chen, Y., Hirschey,
Han, P., Tang, Z., Yin, J., Maalouf, M., Beach, T.G., Reiman, E.M., Shi, J., 2014. Pituitary M.D., Bronson, R.T., Haigis, M., Guarente, L.P., Weissman Jr, R.V., Weissman, S.,
adenylate cyclase-activating polypeptide protects against beta-amyloid toxicity. Verdin, S., Schwer, B., 2007. Mammalian Sir2 homolog SIRT3 regulates global mi-
Neurobiol. Aging 35 (9), 2064–2071. tochondrial lysine acetylation. Mol. Cell. Biol. 27 (24), 8807–8814.
Harlan, B.A., Pehar, M., Sharma, D.R., Beeson, G., Beeson, C.C., Vargas, M.R., 2016. Lombard, D.B., Tishkoff, D.X., Bao, J., 2011. Mitochondrial sirtuins in the regulation of
Enhancing NAD+ salvage pathway reverts the toxicity of primary astrocytes ex- mitochondrial activity and metabolic adaptation. Handb. Exp. Pharmacol. 206,
pressing amyotrophic lateral sclerosis-linked mutant superoxide dismutase 1 (SOD1). 163–188.
J. Biol. Chem. 291, 10836–10846. Manczak, M., Anekonda, T.S., Henson, E., Park, B.S., Quinn, J., Reddy, P.H., 2006.
Hebert, A.S., Dittenhafer-Reed, K.E., Yu, W., Bailey, D.J., Selen, E.S., Boersma, M.D., Mitochondria are a direct site of A beta accumulation in Alzheimer’s disease neurons:
Carson, J.J., Tonelli, M., Balloon, A.J., Higbee, A.J., Westphall, M.S., 2013. Calorie implications for free radical generation and oxidative damage in disease progression.
restriction and SIRT3 trigger global reprogramming of the mitochondrial protein Hum. Mol. Genet. 15, 1437–1449.
acetylome. Mol. Cell 49, 186–199. McDonnell, E., Peterson, B.S., Bomze, H.M., Hirschey, M.D., 2015. SIRT3 regulates pro-
Herskovits, A.Z., Guarente, L.P., 2013. Sirtuin deacetylases in neurodegenerative diseases gression and development of diseases of aging. Trends Endocrinol. Metab. 26 (9),
of aging. Cell Res. 23, 746–758. 486–492.
Herzig, S., Long, F., Jhala, U.S., Hedrick, S., Quinn, R., Bauer, A., Rudolph, D., Schutz, G., Mehrotra, A., Trigun, S.K., 2012. Moderate grade Hyperammonemia induced concordant
Yoon, C., Puigserver, P., Spiegelman, B., 2001. CREB regulates hepatic gluconeo- activation of antioxidant enzymes is associated with prevention of Oxidative stress in
genesis through the coacti- vator PGC-1. Nature 413, 179–183. brain slices. Neurochem. Res. 37, 171–181.
Hirschey, M.D., Shimazu, T., Goetzman, E., Jing, E., Schwer, B., Lombard, D.B., Grueter, Michán, S., Li, Y., Chou, M.M., Parrella, E., Ge, H., Long, J.M., Allard, J.S., Lewis, K.,
C.A., Harris, C., Biddinger, S., Ilkayeva, O.R., Stevens, R.D., Li, Y., Saha, A.K., Miller, M., Xu, W., Mervis, R.F., 2010. SIRT1 is essential for normal cognitive func-
Ruderman, N.B., Bain, J.R., Newgard, C.B., Farese Jr, R.V., Alt, F.W., Kahn, C.R., tion and synaptic plasticity. J. Neurosci. 30, 9695–9707.
Verdin, E., 2010. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible Michan, S., Sinclair, D., 2007. Sirtuins in mammals: insights into their biological func-
enzyme deacetylation. Nature 464 (7285), 121–125. tions. Biochem. J. 404 (1), 1–3.
Hirschey, M.D., Shimazu, T., Jing, E., Grueter, C.A., Collins, A.M., Aouizerat, B., Min, S.W., Sohn, P.D., Cho, S.H., Swanson, R.A., Gan, L., 2013. Sirtuins in neurodegen-
Stančáková, A., Goetzman, E., Lam, M.M., Schwer, B., Stevens, R.D., 2011. SIRT3 erative diseases: an update on potential mechanisms. Front. Aging Neurosci. 5.
deficiency and mitochondrial protein hyperacetylation accelerate the development of Misgeld, U., 2004. Innervation of the substantia nigra. Cell Tissue Res. 318 (1), 107–114.
the metabolic syndrome. Mol. Cell 44, 177–190. Mondal, P., Trigun, S.K., 2014. Pannexin1 as a novel cerebral target in pathogenesis of
Huang, W., Huang, Y., Huang, R.Q., Huang, C.G., Wang, W.H., Gu, J.M., Dong, Y., 2016. hepatic encephalopathy. Metab. Brain Dis. 29 (4), 1007–1115. http://dx.doi.org/10.
SIRT3 expression decreases with reactive oxygen species generation in rat cortical 1007/s11011-014-9556-x.
neurons during early brain injury induced by experimental subarachnoid Mondal, P., Trigun, S.K., 2015. Bacopa monnieri extract (CDRI-08) modulates the NMDA

51
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

receptor subunits and nNOS-apoptosis axis in cerebellum of hepatic encephalopathy NAMPT gene expression profiles in wild- type adult zebrafish liver. Mol. Bio. Rep. 39
rats. Evid. Based Comp. Altern. Med. 2015. (3), 3281–3289.
Monfort, P., Muñoz, M.D., ElAyadi, A., Kosenko, E., Felipo, V., 2002. Effects of hyper- Schlicker, C., Gertz, M., Papatheodorou, P., Kachholz, B., Becker, C.F., Steegborn, C.,
ammonemia and liver failure on glutamatergic neurotransmission. Metab. Brain Dis. 2008. Substrates and regulation mechanisms for the human mitochondrial sirtuins
17 (4), 237–250. SIRT3 and Sirt5. J. Mol. Biol. 382, 790–801.
Moniot, S., Weyand, M., Steegborn, C., 2012. Structures, substrates, and regulators of Schwer, B., Verdin, E., 2008. Conserved metabolic regulatory functions of sirtuins. Cell
mammalian Sirtuins–opportunities and challenges for drug development. Front. Metab. 7 (2), 104–112.
Pharmacol. 3. Schwer, B., North, B.J., Frye, R.A., Ott, M., Verdin, E., 2002. The human silent in-
Mootha, V.K., Handschin, C., Arlow, D., Xie, X., St Pierre, J., Sihag, S., Yang, W., formation regulator (Sir)2 homologue hSIRT3 is a mitochondrial nicotinamide ade-
Altshuler, D., Puigserver, P., Patterson, N., Willy, P.J., Schulman, I.G., Heyman, R.A., nine dinucleotide- dependent deacetylase. J. Cell Biol. 158, 647–657.
Lander, E.S., Spiegelman, B.M., 2004. Erra and Gabpa/b specify PGC-1a-dependent Shi, T., Wang, F., Stieren, E., Tong, Q., 2005. SIRT3, a mitochondrial sirtuin deacetylase,
oxidative phosphorylation gene expression that is altered in diabetic muscle. Proc. regulates mitochondrial function and thermogenesis in brown adipocytes. J. Biol.
Natl. Acad. Sci. U. S. A. 101, 6570–6575. Chem. 280, 13560–13567.
Mungarro-Menchaca, X., Ferrera, P., Moran, J., Arias, C., 2002. beta-Amyloid peptide Shimazu, T., Hirschey, M.D., Hua, L., Dittenhafer-Reed, K.E., Schwer, B., Lombard, D.B.,
induces ultrastructural changes in synaptosomes and potentiates mitochondrial Li, Y., Bunkenborg, J., Alt, F.W., Denu, J.M., Jacobson, M.P., 2010. SIRT3 deacety-
dysfunction in the presence of ryanodine. J. Neurosci. Res. 68, 89–96. lates mitochondrial 3-hydroxy-3-methylglutaryl CoA synthase 2 and regulates ketone
Nakagawa, T., Guarente, L., 2011. Sirtuins at a glance. J. Cell Sci. 124 (6), 833–838. body production. Cell Metab. 12 (6), 654–661.
Nicholas, J.M., Rothstein, J.D., 2006. Mechanisms of Disease: astrocytes in neurodegen- Shulga, N., Wilson-Smith, R., Pastorino, J.G., 2010. Sirtuin-3 deacetylation of cyclophilin
erative disease. Nat. Clin. Pract. Neurol. 2 (12), 679–689. D induces dissociation of hexokinase II from the mitochondria. J. Cell Sci. 15 (123(Pt
Nicholls, D.G., 2009. Mitochondrial calcium function and dysfunction in the central 6)), 894–902.
nervous system. Biochim. Biophys. Acta 1787, 1416–1424. Sidorova-Darmos, E., Wither, R.G., Shulyakova, N., Fisher, C., Ratnam, M., Aarts, M.,
North, B.J., Verdin, E., 2004. Sirtuins: sir2-related NAD-dependent protein deacetylases. Lilge, L., Monnier, P.P., Eubanks, J.H., 2014. Differential expression of sirtuin family
Genome Biol. 5 (5), 224. members in the developing, adult, and aged rat brain. Front. Aging Neurosci. 6, 33.
North, B.J., Marshall, B.L., Borra, M.T., Denu, J.M., Verdin, E., 2003. The human Sir2 Singh, S., Trigun, S.K., 2010. Activation of Neuronal Nitric Oxide Synthase in cerebellum
ortholog, SIRT2, is an NAD+-dependent tubulin deacetylase. Mol. Cell 11 (2), of chronic Hepatic encephalopathy rats is associated with up-regulation of NADPH
437–444. producing pathway. Cerebellum 9, 384–397.
Olsen, J.V., Vermeulen, M., Santa- maria, A., Kumar, C., Miller, M.L., Jensen, L.J., Gnad, Singh, S., Trigun, S.K., 2014. Low grade cirrhosis induces cognitive impairment and
F., Cox, J., Jensen, T.S., Nigg, E.A., Brunak, S., Mann, M., 2010. Quantitative phos- motor. dysfunction in rats: could be a model for minimal hepatic encephalopathy.
phor proteomics reveals wide spread full phosphorylation site occupancy during Neurosci. Lett. 559, 1 (136–140).
mitosis. Sci. Signal. 3, ra3. Singh, S., Koiri, R.K., Trigun, S.K., 2008. Acute and chronic hyperammonemia modulate
Ong, W.Y., Tanaka, K., Dawe, G.S., Ittner, L.M., Farooqui, A.A., 2013. Slow excitotoxicity antioxidant enzymes differently in cerebral cortex and cerebellum. Neurochem. Res.
in Alzheimer’s disease. J. Alzheimers Dis. 35 (4), 643–668. 33, 103–113.
Onyango, P., Celic, I., McCaffery, J.M., Boeke, J.D., Feinberg, A.P., 2002. SIRT3, a human Singh, S., Mondal, P., Trigun, S.K., 2014. Acute liver failure in rats activates glutamine-
SIR2 hom- ologue, is an NAD-dependent deacetylase localized to mitochondria. Proc. glutamate cycle but declines antioxidant enzymes to induce oxidative stress in cer-
Natl. Acad. Sci. U. S. A. 99, 13653–13658. ebral cortex and cerebellum. PLoS One 9 (4), e95855.
Palacios, O.M., Carmona, J.J., Michan, S., Chen, K.Y., Manabe, Y., Ward Iii, J.L., Someya, S., Yamasoba, T., Weindruch, R., Prolla, T.A., Tanokura, M., 2007. Caloric re-
Goodyear, L.J., Tong, Q., 2009. Diet and exercise signals regulate SIRT3 and activate striction suppresses apoptotic cell death in the mammalian cochlea and leads to
AMPK and PGC-1a in skeletal muscle. Aging (Albany NY) 1, 771. prevention of presbycusis. Neurobiol. Aging 28 (10), 1613–1622.
Park, E.S., Kang, J.C., Jang, Y.C., Park, J.S., Jang, S.Y., Kim, D.E., Kim, B., Shin, H.S., Song, W., Song, Y., Kincaid, B., Bossy, B., Bossy-Wetzel, E., 2013. Mutant SOD1G93A
2014. Cardioprotective effects of rhamnetin in H9c2 cardiomyoblast cells under triggers mitochondrial fragmentation in spinal cord motor neurons: neuroprotection
H2O2 induced apoptosis. J. Ethnopharmacol. 153 (3), 552–560. by SIRT3 and PGC-1α. Neurobiol. Dis. 51, 72–81.
Pillai, V.B., Sundaresan, N.R., Jeevanandam, M.P., Gupta, V., 2010a. Mitochondrial Spandou, E., Soubasi, V., Papoutsopoulou, S., Augoustides-Savvopoulou, P., Loizidis, A.,
SIRT3 and heart disease. Cardiovasc. Res. 88 (2), 250–256. Pazaiti, T., Karkavelas, G., Guiba-Tziampiri, O., 2007. Neuroprotective effect of long-
Pillai, V.B., Sundaresan, N.R., Kim, G., Gupta, M., Rajamohan, S.B., Pillai, J.B., Samant, term MgSO4 administration after cerebral hypoxia-ischemia in newborn rats is re-
S., Ravindra, P.V., Isbatan, A., Gupta, M.P., 2010b. Exogenous NAD blocks cardiac lated to the severity of brain damage. Reprod. Sci. 14 (7), 667–677.
hypertrophic response via activation of the SIRT3-LKB1-AMP-activated kinase Sundaresan, N.R., Samant, S.A., Pillai, V.B., Rajamohan, S.B., Gupta, M.P., 2008. SIRT3 is
pathway. J. Biol. Chem. 285, 3133–3144. a stress- responsive deacetylase in cardiomyocytes that protects cells from stress-
Pillai, V.B., Samant, S., Sundaresan, N.R., Raghuraman, H., Kim, G., Bonner, M.Y., mediated cell death by deacetylation of Ku70. Mol. Cell. Biol. 28, 6384–6401.
Arbiser, J.L., Walker, D.I., Jones, D.P., Gius, D., Gupta, M.P., 2015. Honokiol blocks Sundaresan, N.R., Gupta, M., Kim, G., Rajamohan, S.B., Isbatan, A., Gupta, M.P., 2009.
and reverses cardiac hypertrophy in mice by activating mitochondrial SIRT3. Nat. SIRT3 blocks the cardiac hypertrophic response by augmenting Foxo3a-dependent
Commun. 6, 6656. antioxidant defense mechanisms in mice. J. Clin. Invest. 119, 2758–2771.
Prabhakaran, S., Ruff, I., Bernstein, R.A., 2015. Acute stroke intervention: a systematic Tan, W., Pasinelli, P., Trotti, D., 2014. Role of mitochondria in mutant SOD1 linked
review. JAMA 313 (14), 1451–1462. amyotrophic lateral sclerosis. Biochim. Biophys. Acta 1842, 1295–1301.
Prentice, H., Modi, J.P., Wu, J.Y., 2015. Mechanisms of neuronal protection against ex- Tanno, M., Sakamoto, J., Miura, T., Shimamoto, K., Horio, Y., 2007. Nucleocytoplasmic
citotoxicity, endoplasmic reticulum stress, and mitochondrial dysfunction in stroke shuttling of the NAD+-dependent histone deacetylase SIRT1. J. Biol. Chem. 282 (9),
and neurodegenerative diseases. Oxid. Med. Cell Longev. 964518. http://dx.doi.org/ 6823–6832.
10.1155/2015/964518. Tseng, A.H., Shieh, S.S., Wang, D.L., 2013. SIRT3 deacetylates FOXO3 to protect mi-
Putcha, D., Brickhouse, M., O’Keefe, K., Sullivan, C., Rentz, D., Marshall, G., Dickerson, tochondria against oxidative damage. Free Radic. Biol. Med. 63, 222–234.
B., Sperling, R., 2011. Hip- pocampal hyperactivation associated with cortical thin- Van Damme, P., Dewil, M., Robberecht, W., Van Den Bosch, L., 2005. Excitotoxicity and
ning in Alzheimer’s disease signature regions in non-demented elderly adults. J. amyotrophic lateral sclerosis. Neurodegener Dis. 2 (3-4), 147–159.
Neurosci. 31, 17680–17688. Verdin, E., Hirschey, M.D., Finley, L.W., Haigis, M.C., 2010. Sirtuin regulation of mi-
Qiu, X., Brown, K., Hirschey, M.D., Verdin, E., Chen, D., 2010. Calorie restriction reduces tochondria: energy production, apoptosis, and signaling. Trends Biochem. Sci. 35
oxidative stress by SIRT3-mediated SOD2 activation. Cell Metab. 12 (6), 662–667. (12), 669–675.
Raju, D.V., Ahern, T.H., Shah, D.J., Wright, T.M., Standaert, D.G., Hall, R.A., Smith, Y., Vincent, P., Mulle, C., 2009. Kainate receptors in epilepsy and excitotoxicity.
2008. Differential synaptic plasticity of the corticostriatal and thalamostriatal sys- Neuroscience 158 (1), 309–323.
tems in an MPTP-treated monkey model of parkinsonism. Eur. J. Neurosci. 27, Wang, F., Nguyen, M., Qin, F.X., Tong, Q., 2007. SIRT2 deacetylates FOXO3a in response
1647–1658. to oxidative stress and caloric restriction. Aging Cell. 6 (4), 505–514.
Ramadori, G., Lee, C.E., Bookout, A.L., Lee, S., Williams, K.W., Anderson, J., Elmquist, Wang, W., Esbensen, Y., Kunke, D., Suganthan, R., Rachek, L., Bjørås, M., Eide, L., 2011.
J.K., Coppari, R., 2008. Brain SIRT1: anatomical distribution and regulation by en- Mitochondrial DNA damage level determines neural stem cell differentiation fate. J.
ergy availability. J. Neurosci. 28, 9989–9996. Neurosci. 31 (26), 9746–9751.
Raymond, L.A., André, V.M., Cepeda, C., Gladding, C.M., Milnerwood, A.J., Levine, M.S., Wei, I., Zhou, Y., Dai, Q., Qiao, C., Zhou, I., Hui, H., Lu, N., Guo, Q.L., 2013. Oroxylin A
2011. Pathophysiology of Huntington's disease: time-dependent alterations in sy- induces dissociation of hexokinase II from the mitochondria and inhibits glycolysis by
naptic and receptor function. Neuroscience 198, 252–273. SIRT3 mediated deacetylation of Cyclophilin D in breast carcinoma. Cell. Death. Dis.
Revollo, J.R., Grimm, A.A., Imai, S., 2004. The NAD biosynthesis pathway mediated by 4, e601.
nicotinamide phosphoribosyltransferase regulates Sir2 activity in mammalian cells. J. Weir, H.J., Murray, T.K., Kehoe, P.G., Love, S., Verdin, E.M., O'Neill, M.J., Lane, J.D.,
Biol. Chem. 279, 50754–50763. Balthasar, N., 2012. CNS SIRT3 expression is altered by reactive oxygen species and
Salvatore, M.F., Davis, R.W., Arnold, J.C., Chotibut, T., 2012. Transient striatal GLT-1 in Alzheimer’s disease. PLoS One 7 (11), e48225.
blockade increases EAAC1 expression, glutamate reuptake, and decreases tyrosine Xu, Y., Yan, J., Zhou, P., Li, J., Gao, H., Xia, Y., Wang, Q., 2012. Neurotransmitter re-
hydroxylase phosphorylation at ser(19). Exp. Neurol. 234, 428–436. ceptors and cognitive dysfunction in Alzheimer’s disease and Parkinson’s disease.
Satterstrom, F.K., Swindell, W.R., Laurent, G., Vyas, S., Bulyk, M.L., Haigis, M.C., 2015. Prog. Neurobiol. 97 (1), 1–13. http://dx.doi.org/10.1016/j.pneurobio.2012.02.002.
Nuclear respiratory factor 2 induces SIRT3 expression. Aging Cell. 14, 818–825. Yin, J., Han, P.C., Caselli, R., Beach, T., Serrano, G., Reiman, E., Shi, J., 2015a. Sirtuin 3 is
Scher, M.B., Vaquero, A., Reinberg, D., 2007. SIRT3 is a nuclear NAD+-dependent his- down-regulated in Apolipoprotein E4 carriers with Alzheimer’s disease (P5. 011).
tone dea- cetylase that translocates to the mitochondria upon cellular stress. Genes Neurology 84 P5.011.
Dev. 21, 920–928. Yin, J., Han, P., Tang, Z., Liu, Q., Shi, J., 2015b. Sirtuin 3 mediates neuroprotection of
Schirmer, H., Pereira, T.C., Rico, E.P., Rosemberg, D.B., Bonan, C.D., Bogo, M.R., Souto, ketones against ischemic stroke. J. Cereb. Blood Flow Metab. 35 (11), 1783–1789.
A.A., 2012. Modulatory effect of resveratrol on SIRT1, SIRT3, SIRT4, PGC1α and Yoon, J.C., Puigserver, P., Chen, G., Donovan, J., Wu, Z., Rhee, J., et al., 2001. Control of

52
Anamika et al. Journal of Chemical Neuroanatomy 95 (2019) 43–53

hepatic gluconeo- genesis through the transcriptional coactivator PGC-1. Nature 413, neuroprotective agent in rotenone-induced parkinson cell model. Neurochem. Res. 41
131–138. (7), 1761–1773.
Zakhary, S.M., Ayubcha, D., Dileo, J.N., Jose, R., Leheste, J.R., Horowitz, J.M., Torres, G., Zhang, Z., Ma, Q., Smith, M., Manning, M., Quinones, Q., Podgoreanu, M., 2016b. Sirtuin
2010. Distribution analysis of deacetylase SIRT1 in rodent and human nervous sys- 3 ablation exacerbates cerebral injury after experimental cardiac surgery via in-
tems. Anat. Rec. (Hoboken) 293, 1024–1032. flammasome activation. Circulation 134, A18616.
Zhang, J.Y., Deng, Y.N., Zhang, M., Su, H., Qu, Q.M., 2016a. SIRT3 acts as a

53

You might also like