You are on page 1of 36

Neurochem Res (2010) 35:1880–1915

DOI 10.1007/s11064-010-0307-z

ORIGINAL PAPER

Cellular Stress Responses, Mitostress and Carnitine


Insufficiencies as Critical Determinants in Aging
and Neurodegenerative Disorders: Role of Hormesis
and Vitagenes
Vittorio Calabrese • Carolin Cornelius •
Anna Maria Giuffrida Stella • Edward J. Calabrese

Accepted: 21 October 2010 / Published online: 13 November 2010


 Springer Science+Business Media, LLC 2010

Abstract The widely accepted oxidative stress theory of cancer. A critical key to successful medical intervention is
aging postulates that aging results from accumulation of getting the dose right. Achieving this goal can be extremely
oxidative damage. A prediction of this theory is that, challenging due to human inter-individual variation as
among species, differential rates of aging may be apparent affected by age, gender, diet, exercise, genetic factors and
on the basis of intrinsic differences in oxidative damage health status. The nature of the dose response in and
accrual. Although widely accepted, there is a growing adjacent to the therapeutic zones, over the past decade has
number of exceptions to this theory, most contingently received considerable advances. The hormetic dose–
related to genetic model organism investigations. Proteins response, challenging long-standing beliefs about the nat-
are one of the prime targets for oxidative damage and ure of the dose–response in a lowdose zone, has the
cysteine residues are particularly sensitive to reversible and potential to affect significantly the design of pre-clinical
irreversible oxidation. The adaptation and survival of cells studies and clinical trials as well as strategies for optimal
and organisms requires the ability to sense proteotoxic patient dosing in the treatment of numerous diseases. Given
insults and to coordinate protective cellular stress response the broad cytoprotective properties of the heat shock
pathways and chaperone networks related to protein quality response there is now strong interest in discovering and
control and stability. The toxic effects that stem from the developing pharmacological agents capable of inducing
misassembly or aggregation of proteins or peptides, in any stress responses, including carnitines. This paper describes
cell type, are collectively termed proteotoxicity. Despite in mechanistic detail how hormetic dose responses are
the abundance and apparent capacity of chaperones and mediated for endogenous cellular defense pathways,
other components of homeostasis to restore folding equi- including the possible signaling mechanisms by which the
librium, the cell appears poorly adapted for chronic pro- carnitine system, by interplaying metabolism, mitochon-
teotoxic stress which increases in cancer, metabolic and drial energetics and activation of critical vitagenes,
neurodegenerative diseases. Pharmacological modulation modulates signal transduction cascades that confer cyto-
of cellular stress response pathways has emerging impli- protection against chronic degenerative damage associated
cations for the treatment of human diseases, including to aging and neurodegenerative disorders.
neurodegenerative disorders, cardiovascular disease, and
Keywords Redox state  Cellular stress response 
Mitochondrial bioenergetics  Acetylcarnitine  Hormesis 
Special issue article in honor of Dr. Abel Lajtha Vitagenes
V. Calabrese (&)  C. Cornelius  A. M. G. Stella
Department of Chemistry, University of Catania, Viale Andrea
Doria, 95100 Catania, Italy
e-mail: calabres@unict.it Introduction
E. J. Calabrese
Environmental Health Sciences Division, School of Public In response to fluctuations of their microenvironment,
Health, University of Massachusetts, Amherst, MA, USA cells must continuously activate a series of homeostatic

123
Neurochem Res (2010) 35:1880–1915 1881

mechanisms that aim at maintaining their bioenergetic, and heart disease. In several developed countries, the life
metabolic, ionic, and genomic integrity. The frontier expectancy, especially in females, is now into the 80s, with
between normal fluctuations and potentially pathogenic a maximal lifespan being reported of approximately
perturbations (‘‘stress’’) is not neat, and actually depends 120 years. What dictates or regulates ‘‘why we age’’ and
on the capacity of cells to adapt to distinct levels of stress. ‘‘why the ageing body loses functional capacity’’ has been
Hence, any genetically determined or acquired reduction in a matter of debate for centuries. It seems likely that several
the capacity of cells to adapt to cellular stress is potentially factors work together in the ageing process, or that one
pathogenic because it lowers the threshold at which particular factor is the primary culprit in a given individual.
endogenous or exogenous noxious agents cause irrevers- One of the most studied and accepted hypotheses for the
ible cellular damage, culminating in the irreversible loss molecular basis of aging has been the oxidative stress
of cells by the activation of programmed cell death theory of aging. The origins of the free radical theory of
pathways [1]. aging go back to the mid twentieth century, when it was
Maintenance of optimal long-term health conditions is discovered that oxygen free radicals, traditionally thought
accomplished by a complex network of longevity assur- to be too reactive to exist in biological systems, are formed
ance processes which are controlled by vitagenes, a group in situ in response to radiation and oxygen poisoning are
of genes involved in preserving cellular homeostasis during responsible for the associated toxicities [4, 5] (Table 1).
stressful conditions [2]. Vitagenes encode for heat shock Noting that radiation ‘‘induces mutation, cancer and aging’’
proteins (Hsp) Hsp32, Hsp70, the thioredoxin and the [6, 7] and, drawing on the rate-of-living hypothesis [8],
sirtuin protein systems [2]. Dietary antioxidants, such Denham Harman first conceptualized the free radical the-
as polyphenols and L-carnitine/acetyl-L-carnitine, have ory of aging and suggested that oxygen free radicals
recently been demonstrated to be neuroprotective through (specifically hydroxyl, OH•, and hydroperoxyl, HO•2)
the activation of hormetic pathways, including vitagenes. formed endogenously as by-products from normal oxy-
The hormetic dose–response, challenges long-standing genutilizing metabolic processes can play an essential role
beliefs about the nature of the dose–response in a lowdose in the aging process [6]. Later, Harman added a slight
zone, having the potential to affect significantly the design modification to this theory to bring special attention to role
of pre-clinical studies and clinical trials as well as strate- of the mitochondria in the aging process because these
gies for optimal patient dosing in the treatment of numer- organelles are a major site of reactive oxygen species
ous diseases [3]. Given the broad cytoprotective properties (ROS) production [7]. The interest in the free radical the-
of the heat shock response there is now strong interest in ory was at the beginning limited by the persistent doubt
discovering and developing pharmacological agents capa- about the existence of oxygen free radicals in biological
ble of inducing stress responses. In this review we discuss systems [7]. Then, for more than 50 years, numerous
the most current and up to date understanding of the pos- reports have examined the links between oxidative stress,
sible signaling mechanisms by which acetylcarnitine by longevity, and age-related disease [9]. Continuing to today,
activating vitagenes can enhance defensive systems numerous studies have shown that oxidative damage
involved in bioenergetic and stress resistance homeostasis. increases with age in many organisms and that many forms
of ROS may be the culprits of accumulated oxidative
damage. Thus, Harman’s original hypothesis has been
The ‘‘Free Radical Hypothesis’’ of Aging refined in such a way to address the role of all forms of
ROS in regulating aging and now is generally termed the
Ageing is typically accepted as being a product of one’s oxidative stress theory of aging [10]. The basis of this
genetic composition and the cumulative effects of lifestyle theory is that a chronic state of oxidative stress exists in all
practices and exposures. Humans reach a peak of growth cells of aerobic organisms even under normal physiological
and development around the time of their mid 20s. Starting conditions because of an imbalance between pro-oxidants
at what is commonly referred to as ‘‘middle age’’, the and antioxidants [11]. This imbalance leads to the accu-
operations of the body become more susceptible to wear mulation of oxidative damage to cellular macromolecules
and tear, and there is a general decline in physical and that increases during aging and contributes to a progressive
possibly mental performance. During the latter half of life, decline in the function of cellular processes. Under this
an individual is more prone to experiencing problems with mechanistic framework, the regulation of oxidative stress
a range of bodily functions and to developing various may directly control the aging process [12, 13] (Table 1).
chronic or fatal diseases. The cardiovascular, digestive, During the last few years, cellular oxidant/antioxidant
excretory, nervous, reproductive and urinary systems are balance has become the subject of intense study, particu-
particularly vulnerable, and the most common age-related larly by those interested in brain aging and in neurode-
ailments include Alzheimer’s, cancer, arthritis, diabetes, generative mechanisms [14]. Several lines of evidence

123
1882 Neurochem Res (2010) 35:1880–1915

Table 1 Trends in the free radical theory of aging


1950 1960 1970 1980 1990 2000
O2•--dependent Age-dependent increase Increased life span in transgenic
production of H2O2 of oxidative damage flies
in mitochondria

Free radical theory Identification of Discovery of Age-dependent increase of Age-dependent CuZnSOD overexpression
of oxygen toxicity CuZnSOD MnSOD oxidative damage to increase of oxidative does not extend life span
Gerschman (1954) McCord and Fridovich proteins Stadtman damage to DNA in mouse Epstein (2000)
Fridovich (1969) (1972) (1987) Ames (1990)
Free radical theory Mitochondrial Decreased life span in Mt-catalase
of aging Harman theory of CuZn null flies Phillips overexpression extends
(1956) aging Harman and Hilliker (1989) life span in mouse
(1972) Rabinovitch (2005)
CuZn homozygous null
mice exhibit decreased
life span Huang (2005)

suggest that accumulation of oxidative molecular damage reactive hydroxyl radicals. Thus, SOD2, which directs
is a causal factor in senescence. The direct evidence for this H2O2 out of the mitochondrion, accomplishes two func-
hypothesis is that overexpression of antioxidative genes for tions: the classical scavenging of superoxide, as well as its
Cu, Zn-superoxide dismutase and catalase in transgenic signalization as ROS-dependent signaling pathways. More
Drosophila melanogaster prolongs the life span, retarding than 95% of the O2 taken up by the human body is used by
the age-associated accumulation of oxidative damage [12]. mitochondrial cytochrome oxidase which adds four elec-
Among the correlative evidence supporting the involve- trons to oxygen to generate a molecule of water. Cyto-
ment of oxidative stress are the following: (1) oxidative chrome oxidase normally does not release reactive oxygen
damage to DNA and proteins increases exponentially with species into its surroundings. However, a number of
age, and concomitantly, the rates of mitochondrial O•- 2 and investigations have indicated that brain mitochondria
H2O2 generation as well as the susceptibility of tissues to undergo oxidative stress damage and a decrease of cyto-
experimentally induced oxidative stress are increased; (2) chrome c oxidase activity during aging [17]. It has been
experimental regimens that extend life-span, such as postulated that this complex may act as a bottleneck, cre-
caloric restriction in mammals and reduction of metabolic ating a situation of ‘‘electron traffic jam’’ upstream, which
rate in insect decrease the accumulation rates of oxidative would alter the redox state of oxidoreductases in the
damage; (3) mitochondria make two rather contradictory electron transfer chain, and increase their autoxidizability
contributions to cell survival [15]. The classically recog- and rate of superoxide generation. A finding that lends
nized function is the synthesis of ATP for energizing credibility to this hypothesis is that cytochrome c oxidase
endergonic reactions, the other is generation of reactive activity is directly correlated with the average life-span in
oxygen species which may compromise the long-term different species [17]. Oxidative damage to key intracel-
survival of cells and constitute a major underlying cause of lular targets such as DNA or proteins is an important fea-
the aging process. Indeed, these two rather conflicting ture of the normal cellular aging process in the brain, and
functions are part of the same process, namely mitochon- several studies have shown that oxidative damage to DNA
drial respiration. Physiologically, electron transport chain or protein extracted from brain tissue increases with age
consumes up to 90% of the oxygen taken up by a cell, of [14]. Oxidative damage to DNA has been shown to be
which *1% is transformed in superoxide under normal extensive and could be a major cause of the degenerative
physiological conditions [2, 16]. The rate of mitochondrial diseases related to aging such as cancer [18]. Activation of
electron transfer from semi-reduced Q to molecular O2 to persistent DNA damage response (DDR) signaling is
form superoxide is proportional to the product of the associated with the induction of a permanent proliferative
concentrations of semi-reduced Q and O2. It is estimated arrest known as cellular senescence, a phenomenon
that the under pathophysiological conditions, superoxide intrinsically linked to both tissue aging as well as tumor
production can occur at 2–10% of the uninhibited rates. suppression. The DNA damage observed in senescent cells
Superoxide dismutases, in turn, generate the more stable has been attributed to elevated levels of reactive oxygen
hydrogen peroxide before it is transformed by catalase in species (ROS), failing DNA damage repair processes, and/
water or, depending on the presence of metals, into highly or oncogenic activation. It is not clear how labile molecules

123
Neurochem Res (2010) 35:1880–1915 1883

such as ROS are able to damage chromatin-bound DNA to disappeared [23, 24]; (4) studies that involve exogenous
a sufficient extent to invoke persistent DNA damage and exposure of rodents to a DNA-damaging agent have
DDR signaling. Recent evidence suggests that the nucle- revealed that such treatments result in reduced life span,
otide pool is a significant target for oxidants and that oxi- yet, importantly, without affecting (i.e. accelerating) nor-
dized nucleotides, once incorporated into genomic DNA, mal ageing and (5) potentially most compelling, inherited
can lead to the induction of a DNA strand break-associated disorders originating from defects in DNA damage
DDR that triggers senescence in normal cells and in cells response genes give rise to phenotypes that resemble the
sustaining oncogene activation. Evasion of this DDR and normal ageing process. This connection is perhaps best
resulting senescence is a key step in tumor progression exemplified by the human segmental progerias Werner
[19]. With respect to this, it has been proposed that DNA syndrome and Cockayne syndrome. Finally, recent studies
damage is a major factor underlying neuronal degeneration using genetically defined DNA repair mutant mouse
in normal aging as well as the basis of the neurodegener- models suggest that ‘‘ageing and end-of-life fitness are
ative conditions associated with Alzheimer’s disease (AD) determined both by stochastic damage, which is the cause
[20]. Levels of the oxidized nucleotide 8-hydroxy-deoxy- of functional decline, and genetics, which determines the
guanosine (8-OH-dG) a biomarker of DNA damage, have rates of damage accumulation and decline [25]. Moreover,
also been shown to accumulate with aging. In several experimental evidence indicates that intrinsic DNA repair
tissues, including brain and muscle, levels of 8-OH-dG levels associate with longevity. For instance, EBV-
in mtDNA exceed that of nuclear DNA (nDNA) some immortalized lymphoblastoid cell lines from centenarians
16-fold. possess higher than normal poly(ADP)ribose polymerase 1
Mecocci et al., found that 8-OH-dG significantly cor- (PARP1) activity, presumably reflective of an enhanced
relates with increases in levels of a 7.4-kb deletion in DNA strand break response. On the other hand, the rate of
human brain [21]. Experimental studies using lymphocytes production of pro-oxidants, which are responsible for the
from humans or mice, or employing transgenic mouse majority of intracellular damage, has been found in several
models that harbor a defined reporter gene, demonstrate different species to inversely correlate with maximal life
that mutations increase with age [22]. Considering that expectancy [25]. Consistently, it has been demonstrated
nearly 50% of the human genome is transcribed (with 1% that although DNA repair enzymes are present and active
representing protein coding sequences), only a few random in senescent post-mitotic tissues such as brain, changes in
spontaneous mutational events, such as point mutations, the structure and function of ‘‘old’’ chromatin somehow
deletions/insertions, or chromosomal rearrangements, decreases the capacity of the DNA repair enzymes present
could have profound effects on the entire gene regulatory in the nucleus to repair oxidatively damaged DNA. There
circuitry [22]. are several enzymes systems that have been found to repair
The idea that DNA damage contributes to the ageing damage to DNA caused by oxidising species. These
process and particularly to age-related disease is supported include endonucleases, exonucleases, thymine glycol gly-
by the following considerations: (1) DNA is subject to colases and DNA polymerases. DNA polymerases thus far
continuous modification from both endogenous reactive detected in mammalian brain (alpha, beta, delta and epsi-
chemicals, such as reactive oxygen species, and exogenous lon), undergo age-dependent changes in activity, but it is
environmental factors, such as food agents, industrial not known which cell types contain which polymerases,
genotoxins, and ultraviolet and ionizing radiation; (2) DNA and the ability of nuclei from different brain regions
is intrinsically unstable, experiencing spontaneous base to repair specific types of oxidative DNA damage is
loss and deamination at high frequency. Despite the exis- unknown. In addition, recent evidence indicates that
tence of an array of protective DNA repair systems, there is genetic instability, such as telomere loss, somatic and
evidence that several forms of DNA damage, such as strand mitochondrial DNA mutations, increases with age [23].
breaks, alkali-labile sites (i.e. abasic lesions), base modi- An increase in protein oxidative damage, as indicated by
fications (most notably, 8-oxo-2-deoxyguanosine) and the loss of protein sulfhydryl groups and by a decline in the
cross-links (particularly protein–DNA), accumulate with activity of enzymes, such as glutamine synthetase and
age, although the findings depend on the measurement glucose-6-phosphate dehydrogenase has been demon-
technique employed and the tissue or organ studied; (3) strated to occur in brain during aging [26]. A number of
there is evidence that both antioxidant defense mechanisms experimental evidence indicate that increased rate of free
and DNA repair responses decline with age, mouse species radical generation and decreased efficiency of the repara-
displaying different life expectancies possessed a repair tive/degradative mechanisms, such as proteolysis, both are
potential that correlated with maximal life span. However, factors which primarily contribute to age-related elevation
when the organism’s body size was taken into account, the in the level of oxidative stress and brain damage. With
correlation of DNA repair capacity with life expectancy respect to this, it has been suggested that decreases in

123
1884 Neurochem Res (2010) 35:1880–1915

levels of enzymes which ordinarily protect neuronal cells system to injury and disease [35]. Oxidized protein levels
against oxidative stress with age may be responsible for in the CNS tend to increase with age consistently with
increased levels of free-radical damage in the brain, or that several reports that proteasome activity decreases with age
these enzymes themselves are susceptible to inactivation and in neurodegenerative disorders [36, 37]. Importantly,
by free radical molecules which increase with age in the the accumulation of oxidation products in particular
brain [27]. During aging a number of enzymes accumulate regions of the brain seems to be related to specific cogni-
as catalytically inactive or less active forms. The age- tive defects [38]. In addition, studies on the induction of
related changes in catalytic activity are due in part to HSPs response, a cytoprotective mechanism to counteract
reactions of proteins with oxygen and/or nitrogen free oxidative damage, has showed a regional specificity indi-
radical species produced during exposure to ionizing cating that different brain areas might undergo oxidation
radiation or to metal ion catalyzed oxidation systems. The differently and react to protect themselves based on the
levels of oxidized proteins in brain extracts of rats of dif- strength of the insult [39, 40]. Studies from our laboratory,
ferent ages increase progressively with age, and in old rats using a redox proteomics approach to identify the oxida-
can represent 30–50% of the total cellular protein [26]. tively modified proteins in different brain regions of
These evidence provides strong evidence that there is a senescent compared to adult rats, have demonstrated that
linkage between the age-dependent accumulation of oxi- most of oxidized proteins are involved in energy metabo-
dized proteins and the loss in brain physiological functions. lism pathways, including ATP production, glycolysis and
It has recently been proposed that a primary mechanism Krebs cycle. In addition, protein oxidation also affected
leading to neuronal cell death in ageing and common components of the cell involved in cell structure, signal
neurodegenerative disorders is interference with protea- transduction, as well as cellular stress response, such as
some function [28]. The major neurodegenerative diseases, Hsp70 [41–44].
Alzheimer disease (AD), Parkinson’s Disease (PD), amy- Studies on isoprostanes, end product of lipid peroxida-
trophic lateral sclerosis (ALS), Huntington’s disease (HD) tion that can be measured in CSF and urine in various
and Freidreich’s ataxia (FA) are all associated with the neurodegenerative disorders, suggest that lipid peroxida-
presence of abnormal proteins [16, 29]. The origin of HD tion is an early stage in these disease processes. Similarly,
and FA involve specific genetic defects that lead to pro- another end product of lipid peroxidation, the aldehyde
duction of abnormal proteins [30], whereas AD, ALS, and 4-hydroxy-trans-nonenal (HNE), which is highly neuro-
PD have been described mostly as sporadic, although toxic, avidly binds to proteins, and HNE-protein adducts
familial types of AD, ALS and PD are recognized. are demonstrable in senile plaques and tangles in AD,
Examples are the rare mutations in synuclein or parkin that tissues from ALS patients, and lewy bodies in PD [45, 46].
cause familial PD, and the 2% of ALS cases associated There are two broad outcomes of lipid peroxidation:
with mutation in the gene encoding copper-zinc containing structural damage of membrane integrity and the genera-
superoxide dismutase (CuZnSOD) [16, 30–32]. Even in the tion of aldehydic byproducts, particularly a, b-unsaturated
more common sporadic version of PD, ALS and AD aldehydes, including 4-hydroxynonenal (HNE) and acro-
abnormal proteins are present to a significant extent [33]. lein. These aldehydes react quickly with nucleophilic
Thus the senile plaques typical of AD contain not only sulfhydryl groups and are toxic to primary hippocampal
b-amyloid but a wide range of other proteins. Most of them neuron cultures [45] and are elevated in vulnerable areas of
are oxidized and nitrated. Similar damage has been the brain in LAD [45] and MCI. Current evidence suggests
described for proteins in the Lewy bodies in sporadic PD. that HNE and acrolein are by-products of oxidative damage
Oxidative as well as nitrosative protein damage are also to polyunsaturated fatty acids (PUFAs). HNE and acrolein
elevated in ALS. In non-dividing cells, such as the great are by-products of x-6 PUFAs, whereas 4-hydroxyhexenal
majority of neurones in the adult brain, the protein content is derived from x-3 PUFAs [45]. Docosahexaenoic acid
of cells is approximately constant. Since protein synthesis (DHA), an x-3 PUFA, and arachidonic acid (ARA), an x-6
is continuous, there must be an equilibrium between syn- PUFA, are the predominant PUFAs in gray matter. In
thesis and degradation [34]. A variety of conditions of addition, numerous studies demonstrate that acrolein and
neurodegenerative diseases often lead to post-translational HNE are highly reactive, resulting in the disruption of
modifications of proteins, including oxidation and nitra- normal cellular processes and activities through a Michael
tion, which might be involved in the pathogenesis of addition to key amino acids such as lysine, cysteine, and
neurodegenerative diseases. Redox proteomics, a subset of histidine [46]. Modification of proteins by HNE can result
proteomics, has made possible the identification of spe- in a reduction in the activity of several critical enzymes,
cifically oxidized proteins in neurodegenerative disorders, including lactate dehydrogenase and ATP synthase [45].
providing insight into a multitude of pathways that govern Acrolein can impair metabolic processes by inhibiting
behavior and cognition and the response of the nervous glucose uptake in primary neuronal cultures [45]. In vitro

123
Neurochem Res (2010) 35:1880–1915 1885

studies also support acrolein’s role in the disruption in life span. However, when these transgenic/knockout
of glucose metabolism by demonstrating that acrolein mice are tested using models that develop various types of
when incubated with pyruvate dehydrogenase results in age related pathology, they show alterations in progression
decreased activity [45]. Lovell et al. [47] showed that the and/or severity of pathology as predicted by the oxidative
activity of glutathione transferase, a detoxification enzyme stress theory: increased oxidative stress accelerates
for HNE and acrolein, was significantly decreased in the pathology and reduced oxidative stress retards pathology.
HPG of LAD subjects. These contradictory observations might mean that (1)
In spite of all above mentionated evidence, however, oxidative stress plays a very limited, if any, role in aging
current literature data showing the effects of alterations in but a major role in health span and/or (2) the role that
the antioxidant defense system on the life span of mice (as oxidative stress plays in aging depends on environment. In
well as various invertebrate models) seriously call into environments with minimal stress, as expected under
question the classical interpretation of the oxidative stress optimal husbandry, oxidative damage plays little role in
theory of aging, in particular the role of oxidative damage aging. However, under chronic stress, including patholog-
as a major factor underlying the mechanism of the aging ical phenotypes that diminish optimal health, oxidative
process. The oxidative stress theory of aging proposes a stress/damage plays a major role in aging. Under these
causal relationship between reactive oxygen species and conditions, enhanced antioxidant defenses exert an ‘‘anti-
aging, predicting that manipulations that alter oxidative aging’’ action, leading to changes in life span, age-related
stress/damage will alter aging. While it is clear that oxi- pathology, and physiological function as predicted by the
dative damage increases with age, its role in the aging oxidative stress theory of aging [13].
process is, however, uncertain. Examining the relationship
between ROS and aging in genetic model organisms, such
as Caenorhabditis elegans, summarizing experiments using The Mitochondrial Theory of Aging
long-lived mutants, mutants with altered mitochondrial
function, mutants with decreased antioxidant defenses, Mitochondria are membrane-enclosed organelles found in
worms treated with antioxidant compounds, and worms eukaryotic cells where account for the generation of ATP
exposed to different environmental conditions, there is as a source of chemical energy. The synthesis of ATP
strong indication that, while there is frequently a negative occurs through the respiratory or electron transport chain
correlation between oxidative damage and lifespan, there (ETC.) which is located at the inner mitochondrial mem-
are many examples in which they are uncoupled. Neither is brane and consists of five protein complexes (Complexes
resistance to oxidative stress sufficient for a long life nor I–V). Mitochondrial respiration accounts for approximately
are all long-lived mutants more resistant to oxidative stress. 90% of cellular oxygen consumption. Most of the oxygen
Similarly, sensitivity to oxidative stress does not neces- that is consumed is reduced to water through four con-
sarily shorten lifespan and is in fact compatible with long secutive one-electron reductions. During this process, a
life. Furthermore, recent finding obtained from Clk mutants small proportion of the oxygen molecules (1–2%) are
of Caenorhabditis elegans, characterized by slow physio- converted to superoxide anion radicals. The main sites of
logic rates, delayed development, and increased life span, superoxide production in the respiratory chain have been
challenge the hypothesis that decreased metabolism leads recognized to be complexes I and IV [50] and recent
to increased longevity through a decreased production of studies have identified at least nine submitochondrial
reactive oxygen species. In fact, although mitochondrial reactive oxygen species (ROS) generating sites [51]. The
function is decreased in the Clk mutants, ATP levels are superoxide that is formed then dismutates either sponta-
normal or increased, suggesting decreased energy utiliza- neously or enzymatically through the action of superoxide
tion, with no evidence for systematically increased resis- dismutase to form hydrogen peroxide [52]. Hydrogen
tance to oxidative stress or decreased oxidative damage. peroxide then can diffuse throughout the cell and decom-
This phenotype suggests that increased life span may be pose to form noxious hydroxyl radicals, which can injure
achieved by decreasing energy expenditure, without the cell through interactions with macromolecules. Because
decreased production of ROS [48]. Overall, the data in of these processes, mitochondria are a major source for
C. elegans indicate that oxidative damage can be dissoci- physiological or endogenous production of ROS [2].
ated from aging, at least in experimental situations [49]. If Although much attention has been focused on the harmful
one consider as gold standard for determining whether effects of ROS, it now has become apparent that mitoc-
changes in the aging process occur, alteration in life span, hondrially generated ROS are also involved in the regula-
studies from mice with genetic manipulations in their tion of intracellular signal transduction pathways leading to
antioxidant defense system designed to directly address cellular activities such as proliferation [2]. In addition to
this prediction have, with few exceptions, shown no change supplying ATP, mitochondria are required for numerous

123
1886 Neurochem Res (2010) 35:1880–1915

other cellular functions including biosynthesis of heme, sequence repeats beginning at nucleotides 8470 and 13447,
cholesterol and phospholipids [51], as well as initiation of removing almost a 5-kb region of mtDNA between ATPase
the apoptotic process [2]. Mitochondria are organelles 8 and the ND5 genes. The deletion is thought to occur
which, according to the endosymbiosis theory, evolved during replication of the mtDNA, the absent sequence
from purpurbacteria approximately 1.5 billion years ago. encoding for six essential polypeptides of the respiratory
One of the features of mitochondria is that they have their chain and 5 tRNAs. It has been associated with several
own genome [52–54]. Mitochondria replicate and tran- clinical diseases, such as chronic progressive external
scribe their DNA semiautonomously. Like nuclear DNA, ophthalmoplegia and Kearns Sayre syndrome. Several age-
mitochondrial DNA (mtDNA) is constantly exposed to related disorders have been shown to be linked to higher
DNA damaging agents. Regarding the repair of mtDNA, levels of mtDNA mutations than age-matched controls. In
the prevailing concept for many years was that mtDNA the CNS a 17 times higher levels of the common deletion in
molecules suffering an excess of damage would simply be the striatum of patients with Parkinson’s disease have been
degraded to be replaced by newly generated successors demonstrated, compared to age-matched controls. Evi-
copied from undamaged genomes. However, evidence now dence also exists indicating higher levels of this deletion in
clearly shows that mitochondria contain the machinery to patients with Alzheimer’s disease which parallel increased
repair the damage to their genomes caused by certain levels in the oxidized nucleotide 8-OH-dG [50]. Over 100
endogenous or exogenous damaging agents. Harman [6, 7] mutations of mitochondrial DNA (mtDNA) have been
proposed that free radicals are involved in the aging pro- associated with human disease [55]. The phenotypic
cess and subsequently suggested that mitochondria-derived manifestation of mtDNA mutations is extremely broad,
ROS may contribute to cellular aging [7]. Experimental from oligosymptomatic patients with isolated deafness,
support for mitochondrial involvement in cellular senes- diabetes, ophthalmoplegia, etc., to complex encephalo-
cence was initially provided by treatment of IMR-90 myopathic disorders that may include dementia, seizures,
fibroblasts with N-tertbutyl hydroxylamine, an antioxidant ataxia, stroke-like episodes, etc. The genotype variants are
that is recycled by the mitochondrial electron-transport also wide, with rearrangements (deletions, duplications)
chain. N-tert-butyl hydroxylamine extends fibroblast rep- and point mutations affecting protein coding genes, tRNAs
licative capacity and delays age-related changes in mito- and rRNAs. There are some broad genotype/phenotype
chondrial function, including reducing ROS production, correlations but also substantial overlap. The pathogenetic
preservation of mitochondrial membrane potential, and mechanisms involved in the expression of mtDNA muta-
increasing the cellular GSH/GSSG ratio [51]. More tions are still not yet fully understood. More recently,
recently, it was demonstrated that mitochondria derived mutations of nuclear genes encoding subunits of the
ROS play an important and direct role in the shortening of respiratory chain, particularly those of complex I, have
telomeres and the onset of senescence [51]. It has been been identified. These predominantly, but not exclusively,
proposed that mitochondrial dysfunction induces mito- involve infant onset disease with early death. Recently it
chondrial biogenesis, thus increasing the number of sites in has become clear that the function of the respiratory chain
the cell for the production of ROS, which accelerates may be impaired by mutations affecting other mitochon-
telomere shortening [2, 50]. drial proteins or as a secondary phenomenon to other
It has been proposed that accumulation of mtDNA intracellular biochemical derangements. Examples include
during life is a major cause of age-related disease and this Friedreich ataxia where a mutation of a nuclear encoded
is because of its high mutagenic propensity. The lack of protein (frataxin), probably involved in iron homeostasis in
introns and protective histones, limited nucleotide excision mitochondria, results in severe deficiency of the respiratory
and recombination DNA repair mechanisms, location in chain in a pattern indicative of free radical mediated
proximity of the inner mitochondrial membrane which damage [16, 31, 56, 57].
expose it to an enriched free radical milieu, are all factors Mutations of nuclear encoded proteins involved in
contributing to a 10-fold higher mutation rate occurring in cytochrome oxidase assembly and maintenance have been
the mtDNA than in the nDNA [16]. Moreover, a large body characterised and, as predicted, are associated with severe
of evidence indicates that mtDNA mutations increase as a deficiency of cytochrome oxidase and, most frequently,
function of age reaching the highest levels in brain and Leigh syndrome. Defects of intracellular metabolism, with
muscle. More than twenty different types of deletions have particularly excess-free radical generation including nitric
been documented to accumulate in aging human tissues. oxide or peroxynitrite, may cause secondary damage to the
The first report on an age-related increase in a mtDNA respiratory chain. This is probably of relevance in Hun-
deletion, was called the ‘‘common deletion’’ and was found tington disease, motor neuron disease (amyotrophic lateral
in elderly brain and in Parkinson’s disease [16]. This sclerosis) and Wilson disease. These disorders seem to
deletion has been described to occur between 13-bp have defective oxidative phosphorylation as a common

123
Neurochem Res (2010) 35:1880–1915 1887

pathway in their pathogenesis and it may be that treatments retrograde response has been named mitochondrial hor-
designed to improve respiratory chain function may ame- mesis or mitohormesis, and may in addition be applicable
liorate the progression of these disorders [55]. These to the health-promoting effects of physical exercise in
finding establishes the relationship between age-associated humans and, hypothetically, impaired insulin/IGF-1-
accumulation of mtDNA mutations and bioenergy dys- signaling in model organisms. Consistently, abrogation of
function as a key feature of the aging process, at least in this mitochondrial ROS signal by antioxidants impairs the
tissues predominantly composed by postmitotic cells, such lifespan-extending and health-promoting capabilities of
as CNS and skeletal muscle. Relevant to mitochondrial glucose restriction and physical exercise, respectively. In
bioenergetics, in fact, is the finding of a significant summary, ROS are essential signaling molecules (Fig. 1)
decrease in state 3/state 4 ratio, which has been observed to which are required to promote health and longevity. Hence,
occur in brain during aging [57]. Since this ratio relates to the concept of mitohormesis provides a common mecha-
the coupling efficiency between electron flux through the nistic denominator for the physiological effects of physical
electron transport chain and ATP production, an increase in exercise, reduced calorie uptake, glucose restriction, and
state 4 would results in a more reductive state of mito- possibly beyond [62].
chondrial complexes and, consequently, to an increase in
free radical species production. A decrease in state 3/state 4
respiration during aging has been found associated with a Energy Thresholds in Brain Mitochondria: Implication
significant decrease in cardiolipin content in brain mito- for Age-Related Disorders
chondria [58]. This loss could play a critically important
role in the age-related decrements in mitochondrial func- Mitochondria contains essential mechanisms of energy
tion, and appears to be associated with both quantitative production, signalization, biosyntheses, and apoptosis and
and qualitative region-specific protein changes which are the coordinated orchestration and control of these specific
parallel to structural changes, such as decrease of the inner events is a critical determinant in cell physiology [50].
membrane surface, smaller as well as sparser cristae, However significant morphological and functional differ-
decreased fluidity and increased fragility. Modifications in ences exist in mitochondria isolated from different tissues
cardiolipin composition it is recognized to accompany associated with different energy demands as well as pref-
functional changes in brain mitochondria which include all erences in the type of energy substrates, especially in
proteins of the inner mitochondrial membrane that gener- organs that present with highly specialized functions.
ally require interaction with cardiolipin for optimal cata- Consistent with this notion, in human heart, mouse and rat
lytic activity. Acetylcarnitine fed to old rats increased mitochondria, have been identified approximately 600–700
cardiolipin levels to that of young rats and also restored proteins with a strong diversity in their expression levels
protein synthesis in the inner mitochondrial membrane, as among tissues; this suggests quite a large regulatory and
well as cellular oxidant/antioxidant balance, suggesting composition diversity of the mitochondrial proteome,
that administration of this compounds may improve cel- which could explain the observed structural and functional
lular bioenergetics in aged rats [59]. Interestingly, caloric heterogeneity along with the consequent diversity in the
restriction, a dietary regimen that extends life-span in control of mitochondrial respiration. These differences in
rodents, maintains the levels of 18:2 acyl side chains and the control of oxidative phosphorylation might reflect
inhibits the cardiolipin composition changes [60] In addi- physiological variations in the OXPHOS machinery, which
tion, caloric restriction showed to retards the aging asso- could allow the efficiency and the rate of ATP production
ciated changes in oxidative damage, mitochondrial oxidant to adapt to the tissue-specific energy demand. This, oxi-
generation and antioxidant defenses observed during aging dative phosphorylation capacity presents an high degree of
[2, 61]. variability depending on (1) the mitochondrial content, (2)
Recent evidence suggests that calorie restriction and the amount of respiratory chain complexes, and (3) their
specifically reduced glucose metabolism induces mito- intrinsic activity: these factors combine together to result in
chondrial metabolism to extend life span in various model a greater level of functional diversity [62]. Human cells
organisms, including Saccharomyces cerevisiae, Dro- contain from a few hundred to more than a thousand
sophila melanogaster, Caenorhabditis elegans and possi- mitochondria; each mitochondrion in turn has 2–10 copies
bly mice. In conflict with Harman’s free radical theory of mtDNA, thus, several thousands copies of the mito-
of aging, these effects may be due to increased formation chondrial genome can be present within a single cell.
of reactive oxygen species within the mitochondria causing Importantly, unique to mtDNA is that it is inherited
an adaptive response that culminates in subsequently exclusively through the mother, and may exist in many
increased stress resistance assumed to ultimately cause a different copies in the oocyte cytoplasm. This implies that
long-term reduction of oxidative stress. This type of no mtDNA recombination occurs at fertilization and only a

123
1888 Neurochem Res (2010) 35:1880–1915

Fig. 1 Major sites for mitochondrial ROS production and the superoxide at complex I, II, and III. Complex I and II generate
principle of preconditioning. The ubiquinone (Q) cycle is initiated superoxide within the mitochondrial matrix. Complex III can generate
when one electron from ubiquinol (QH2) is donated to the Rieske- superoxide in both the intermembrane space and the matrix. Release
iron sulfur (R-FeS) protein and the second electron is donated to of superoxide from complex III into the cytosol is followed by
cytochrome b. The intermediate moiety is the free radical ubisem- conversion to H2O2 and subsequent activation of oxidant-dependent
iquinone (Qo), which can donate electrons to molecular oxygen to (redox) signaling pathways which results in precondition
generate superoxide. Mitochondrial electron transport chain generates

sequential accumulation of mutations from the maternal normal, very low weight (37 kg) and basal temperature
lineage account for mtDNA variations. Moreover, mtDNA reaching 38C. Moreover, she had a partially uncoupled
is particularly prone to mutation, being estimated as 10 respiration which accounted for her generation of excessive
times greater then nuclear DNA [52], owing to the absence heat and high calorie consumption. Although the primary
of protective proteins (such as histones) and of a high- etiologic event in Luft’s disease remains to be identified,
efficiency repair system. Thus, mutant and wild-type the disease is associated with release of mitochondrial
(normal) mtDNA can coexist within a cell in any propor- calcium stores, abnormal calcium cycling and sustained
tion and this situation is termed heteroplasmy. It is stimulation of loosely coupled respiration. The discovery
becoming increasingly clear that the mitochondrial genome of Luft’s disease cleared the path for fertile investigations
may play an essential role in the pathogenesis of neuro- and since 1960s, over 120 human mitochondrial diseases
degenerative diseases, and evidence for mitochondria being have been discovered, many of which involve selected
a site of damage in neurodegenerative disorders is based in populations in the central nervous system consisting of
part on observed decreases in the respiratory chain com- postmitotic, highly energy-dependent cells. Many of these
plex activities in Parkinson’s, Alzheimer’s, and Hunting- diseases have been associated with specific inherited
ton’s disease. Such defects in respiratory complex mitochondrial DNA mutations and respiratory chain defi-
activities, possibly associated with oxidant/antioxidant ciencies. Point mutations may involve either the RNA or
imbalance, are thought to underlie defects in energy protein-encoding genes, and rearrangements may take the
metabolism and induce cellular degeneration [53]. The first form of deletions or duplications. In the presence of het-
hint that mitochondria play a role in human disease did not eroplasmy there is a critical ratio of mutant to wild-type
emerge until 1958, when a Swedish patient was identified mitochondrial genomes that is necessary before the disease
who had symptoms of severe perspiration, polydipsia, becomes both biochemically and clinically apparent. As
polyphagia, weight loss and weakness [63]. Biochemical might be expected mtDNA disorders are phenotypically
investigation showed a basal metabolic rate 200% above diverse given the ubiquitous presence of mitochondria and

123
Neurochem Res (2010) 35:1880–1915 1889

the variation in the levels of heteroplasmy in the body. Yet on different types of mitochondria, which leads to the
many have predominant neurologic and muscular symp- observation that the threshold value for a given complex
toms, including dementia, seizures, ataxic syndromes, of the electron transport chain can vary according to the
peripheral neuropathies, and progressive myopathy. Post- threshold in the energy demand of different tissues.
mitotic tissues typically show increased levels of mutant According to the metabolic control theory [69–71] the
mitochondria due to the inability of these tissues to select importance of the control of a given isolated step on
against cells containing mutant mtDNA genomes. CNS the global flux depends on several parameters, including
imaging of patients with mtDNA disorders often reveals the absolute content of enzyme, its intrinsic kinetic
moderate degrees of cerebral or cerebellar atrophy that are parameters and associated regulations, the architecture of
consistent with neurodegeneration, often comparable with the metabolic network, the concentration of intermediate
the pathologies associated with the brain in senescence or substrates, and the respective steady state [70]. Further-
in dementia [64]. more, at each level, single threshold effect will reinforce
Several mechanisms have been proposed to explain the the others, as interpreted by the Double threshold
variability of the phenotypic expression of a mtDNA hypothesis [50, 72], whose predictions are now beginning
mutation, such as sporadic mutation or mitotic segrega- to be documented. Consistent with this, metabolic control
tion. However all these hypotheses incorporate a unique analysis (MCA) of complex I activities in mitochondria
feature of mitochondrial genetics and pathologies; that of has revealed some interesting findings concerning mito-
the heteroplasmic concept of mtDNA mutations. Levels chondrial heterogeneity in the brain. When the relative
of mutation can vary considerably between mitochondria, contribution of individual mitochondrial respiratory chain
cells, and even tissues within the same individual. Con- complexes to the control of NAD-linked substrate (glu-
sequently, the expression of a mutation in the mtDNA can tamate and malate) oxidative phosphorylation in mito-
be thought of as a function of the degree of the hetero- chondria from various rat brain regions was investigated,
plasmy. In general, whether or not a metabolic defect it was found that complex I possessed a higher control
expresses itself as a recognizable clinical disease will over oxidative phosphorylation in synaptic mitochondria
depend upon the extent to which it affects the metabolic than in non-synaptic mitochondria. Using rotenone or
pathway in question and this can lead to a threshold myxothiazol and KCN to titrate out complex I, III and IV
expression of the disease state [65]. One of the most activities, respectively, while measuring oxidative phos-
important features recognized in mitochondrial diseases is phorylation parameters, the highest flux control coeffi-
the existence of a threshold in the degree of a mito- cients were found with complex I in mitochondria of
chondrial deficit for the expression of the disease, and synaptic origin. This demonstrates that respiratory chain
these were shown by Wallace [66, 67] to be related to the complexes are involved in the control of mitochondrial
balance between normal and mutant mtDNA. Consistent respiration. However, the distribution of the control
to this, only 10% of wild type DNA is enough to maintain indices of these complexes may be different, depending
a normal respiratory rate, whereas it must be achieved on the tissue from which the mitochondria were isolated.
80–90% deletion of mtDNA before complex IV activity is Brain mitochondria show lowest oxidative phosphoryla-
compromised [50, 68]. This indicates a threshold in the tion efficiency with complex I substrates, measured as
heteroplasmy of the mutation around 90% exists before a P/O ratios, compared to mitochondria from heart and liver
pathological consequence become manifest. Below this [72]. Data from studies of rat brain mitochondria of non
threshold the flux of respiration and of ATP synthesis are synaptic origin have shown that thresholds exist whereby
at a level which does not compromise normal metabolism. complex activities need to be reduced by at least 60%
As consequence, there are at least four levels at which before major changes in ATP synthesis and oxygen con-
threshold effects occur in mitochondrial metabolism, with sumption occur. Interestingly, in synaptic mitochondria,
respect to their possible involvement in the pathogenesis titration of various complexes with specific inhibitors
of neurodegeneration. The first is the expression of the generated threshold curves showing that complex I, III
heteroplasmy of mtDNA at the level of a given enzymatic and IV activities had to be decreased 25, 80 and 70%,
step, whereby mitochondria from patients might exhibit a respectively before major changes in rates of oxygen
particular ratio of defective DNA compared to normal consumption and ATP synthesis were observed [70–72].
DNA. The second is the threshold effect observed in the Thus mitochondria of synaptic origin complex I activity
mitochondrial metabolism as a result of a decrease in a has a major control of oxidative phoshorylation, such that
given mitochondrial activity. The third may occur in the when a threshold of 25% inhibition is exceeded, energy
expression of defective mitochondria with respect to the metabolism is compromised, and reduction in ATP syn-
whole cellular metabolism. The fourth, is the fact that thesis ensues. Moreover, glutathione depletion abolishes
the control coefficient of a given step may vary depending the threshold for complex I providing experimental

123
1890 Neurochem Res (2010) 35:1880–1915

evidence that antioxidant status, which is critically action potentials. This is corroborated by the finding that
involved in neurodegenerative disorders and in particular reduction of complex I activities in synaptosomes from
in PD, maintain energy thresholds in mitochondria aged rats resulted in a faster release of glutamate, com-
[71, 72]. pared to synaptosomes from adult rats, suggesting that
According to the metabolic control theory, at molecular nerve terminals from aged animals, as opposed to younger
level, a critical step in the expression of a threshold for a rats, are more vulnerable to mitochondrial dysfunction and
clinical disease is the impact that a localized defect in a damage [78]. Further refinements of this notion point to the
given point on the global flux of a metabolic network nature of complex I assembly in brain mitochondria.
[50, 73–75]. Mammalian complex I is, in fact, composed of 45 different
Mitochondrial dysfunction is characteristic of several subunits that assemble together at the inner mitochondrial
neurodegenerative disorders [50] and also of the aging membrane [50]. The assembly of this complex is compli-
process [55]. Parkinson’s disease is characterized by a cated partly because of its large size and partly because its
selective decrease in dopamine in the striatum caused by a regulation by two genomes. Seven of the complex I sub-
degeneration of dopaminergic neurons in the zona com- units are encoded by the mitochondrial (mt) DNA and the
pacta of the substantia nigra [54, 59, 66]. Complex I remaining by the nuclear genes. The nuclear DNA-encoded
(NADH:ubiquinone oxidoreductase, EC 1.6.5.3) activity is subunits are synthesized in the cytosol and are imported
reduced by 35–40% in substantia nigra homogenates in into the organelle. The mtDNA-encoded subunits must
postmortem studies of Parkinson’s disease patients [64]. assemble with the nuclear encoded subunits to generate the
However, recent evidence has suggested that a complex I functional holoenzyme [50]. The multimeric holoenzyme is
deficiency may be widespread in the parkinsonian brain, as L-shaped with a hydrophobic membrane arm embedded in
the occurrence of a mutation in mitochondrial DNA in the mitochondrial inner membrane and perpendicular to it
idiopathic Parkinson’s disease [71], and an associated is a hydrophilic peripheral arm which protrudes into the
reduction in complex I activity in mitochondria from the mitochondrial matrix [72]. The 14 most conserved subunits
frontal cortex [39, 71] have been reported. This evidence (which form the fully functional complex I in bacteria)
supports the hypothesis that complex I deficiency plays a form the ‘core’ structure. The ‘core’ structure is also
central role in the initial etiology of the disease [16, 31]. considered to be the minimal structure required for the
The cause of the complex I deficiency is unknown, how- functionality of the enzyme [72]. The function of most of
ever, followup studies on Parkinson’s disease patients the additional 31 supernumerary subunits is not yet clear
injected with fetal tissue enriched in dopaminergic cells but some are hypothesized to stabilize or to protect the
shows that an environment exists in the parkinsonian brain complex from ROS damage. Several of these subunits may
that induces the classical disease pathology in the trans- have an additional function. For example, the subunits
planted cells [76]. It is not known if complex I activity in NDUFA13 [also known as GRIM-19] and NDUFS1 have
these cells is reduced following transplantation or if a been implicated to have a role in apoptosis [72].
complex I defect in surrounding cells induces the neuro- Assembly of complex I subunits is an intricately elab-
degenerative condition. However, synaptosomes show that orate process, however emerging evidence indicates that
complex I deficiencies can result in large amounts of glu- complex I assembly is not sequential but is modular which
tamate being released that may be potentially excitotoxic to means that the subunits are not incorporated one by one
surrounding cells [77]. Dopamine neurons are known to into the complex but instead several discrete assembly
possess glutamate receptors, therefore, existing complex I intermediates are assembled independently and are joined
deficiencies that lead to increased glutamate release may in several steps to form a peripheral arm and a membrane
increase excitotoxic events in the parkinsonian brain [77]. arm, with peripheral arm assembled independently of the
The significance of energy threshold theory applied membrane arm; sub-assemblies of the nuclear DNA-
to mitochondrial dysfunction is, is relevant to described encoded subunits and the mtDNA-encoded subunits can be
decrease of 40% in complex I activity in Parkinson’s dis- formed independently. In addition to this, complex I
ease patients [72], which would correspond to a 35–40% assembly is a highly dynamic process in which existing and
inhibition of ATP synthesis and respiration in the synaptic newly synthesized subunits can be exchanged, so that a
mitochondria model. Titration of complex I activity in complex I transition between assembled and intermediate
synaptosomes, followed by depolarization with KCl or forms may constitute a mechanism which promotes the
4-aminopyridine, reduces ATP concentrations and leads to turnover of subunits, some of which may be oxidatively
significant amounts of Ca2?-independent glutamate damaged [72]. Considering that an increased number of
release [77]. These results indicates that a 40% decrease in complex I subunits become oxidatively damaged in the
complex I activity in the substantia nigra in PD patients can brains of patients with Parkinson’s disease [79], it is con-
induce excess release of excitotoxic glutamate following ceivable to hypothesize that a decreased rate of protein

123
Neurochem Res (2010) 35:1880–1915 1891

import and/or assembly which slows the exchange rate of organization of the complexes. Organization into super-
pre-existing complex I subunits for new subunit import complexes has the advantage of improving stabilization of
may underlie a pathogenic basis of the disease. This may individual complexes, but at the same time mutations
lead to increased oxidative damage, complex I deficiency leading to a loss of complex III prevents supercomplex
and subsequent overt disease. The complex I assembly formation, thus leading to a secondary loss of complex I.
process is even more complicated by the fact that in Finally this model is sustained by the finding that the
addition to a perfect assembly, the nuclear transcription, different complexes are present in vivo at defined stoi-
translation, processing, export of the subunits and then chiometries, and single particle electron microscopy has
their import into the mitochondria, insertion into the provided the 3D maps of the supercomplexes. The su-
membrane, stabilization and activation of the enzyme, percomplexes have been identified and characterized from
every step requires a precise coordination. In light of these a broad range of organisms using BN-PAGE and sucrose
events it is reasonable that chaperons, which assist the gradient centrifugation. The supercomplex I–III, charac-
correct assembly of other polypeptide containing structures terized in Arabidopsis, consists of complex I and dimeric
in vivo, although are not a component of the final func- complex III. The supercomplex III–IV consisting of
tional structure, may contribute to pathogenesis of the dimeric complex III and one to two copies of monomeric
disease. This possibility is corroborated by the evidence complex IV, has been characterized in yeast. The largest
that mutations in the assembly chaperones that lead to the supercomplex from bovine heart, consists of complex I
inability to assemble a properly functioning complex I and plus complex III2 plus complex IV1–4 and is termed the
thus lead to complex I deficiency diseases, have been ‘respirasome’. The supramolecular organization of the
reported [55, 59]. Contrarily to the numerous complex III oxidative phosphorylation complexes into respirasomes
and complex IV assembly chaperones known, so far only may be of functional importance as they may enhance
three candidate complex I assembly factors have been electron transfer rates, reduce diffusion distances of sub-
found. Two of these, the NDUFAF1 (the human homo- strates by substrate channeling and increase the stability
logue of fungal protein CIA30) and a paralogue of complex of the complexes. Although little is known about super-
I subunit B17.2 known as B17.2-like (B17.2L) are well complexes in the brain, the different energy thresholds
established and well studied complex I assembly chaper- and flux control coefficients described above may be
ones [80, 81]. An emerging area of investigation is the related to a range of supercomplex formations in brain
supramolecular organization into ‘respirasomes’ of indi- mitochondria [82]. Formation of supercomplexes I–III has
vidual electron transport chain complexes. The five com- been demonstrated to be facilitated by lipid component in
plexes (complexes I–V) of the oxidative phosphorylation the inner membrane, in particular cardiolipin, which
(OXPHOS) system of mitochondria can be extracted in the appears critical for the stability of supercomplexes [83].
form of active supercomplexes. Single-particle electron As a consequence, exposure of mitochondria to reactive
microscopy has provided 2D and 3D data describing the oxygen species can oxidatively damage cardiolipin with
interaction between complexes I and III, among I, III and an impact on supercomplexes stability. This may be an
IV and in a dimeric form of complex V, between two ATP underlying cause of aging and age-related disorders like
synthase monomers. The stable interactions are called su- Parkinson’s disease, where oxidative damage can induce
percomplexes which also form higher-ordered oligomers. reduction in mitochondrial energetic capacity due to dis-
Cryo-electron tomography provides new insights on how sociation of supercomplexes, with pathological implica-
these supercomplexes are arranged within intact mito- tions [72]. Consistent to this, progress by structural
chondria. In the past, two models have been proposed for investigations, paralleled by insights into the functional
the organization of the respiratory chain. The random roles of respirasomes including substrate channelling and
collision model and the solid state model. According to the stabilization of individual complexes have revealed car-
first model, complex I–IV moves freely by lateral diffusion diolipin an important factor involved in the structural
in the inner mitochondrial membrane and electron transfer stability of respirasomes which in turn is required to
is based on random collisions of the involved components. maintain cells and tissues in a healthy state. Defects in
Ubiquinone and cytochrome c have a diffusion rate that is cardiolipin remodeling cause devastating diseases like
faster than the movements of the membrane embedded Barth syndrome, a disease associated with a mutation in
complexes [72]. In spite of this elaboration, the solid model human TAZ gene, which results in production of an
of the electron transport chain complex organization is altered gene product, Tafazzin, a putative phospholipid
more widely accepted now, which promotes the preferen- acyltransferase. Patients suffering from Barth syndrome
tial associations and specific aggregation of the complexes. exhibit defects in cardiolipin, associated with mitochon-
This stems from immunoprecipitation as well as flux con- drial morphological abnormalities and respiratory chain
trol experiments, which point to the supramolecular dysfunction [83].

123
1892 Neurochem Res (2010) 35:1880–1915

Redox Regulation of Mitochondrial Dynamics Fusion ‘‘Q-cycle’’ mechanism (Fig. 1). Conceivably, a key reac-
and Fission: Role in Cell Survival tion in the Q-cycle is the ‘‘bifurcated’’ electron transfer at
the Qo site, although the exact mechanism is still contro-
Mitochondria are the major source of energy (ATP and versial [84]. In the bifurcated reaction, QH2 is oxidized at
NAD?) production in eukaryotic cells, and produce cor- the Qo site, with one electron being transferred through the
respondingly large amounts of superoxide anion radical high potential chain to reduce cyt c, and the other electron
through abstraction of an electron from oxygen in the so- is transferred through the low potential chain, to reduce a
called electron transport chain at the inner mitochondrial quinoid species (Q or SQ, depending on the state of the
membrane [50]. Mitochondrial function is essential to two-electron gate) at the Qi site. Two turnovers of the Qo
neuronal processes such as energy production, Ca2? regu- site are required to reduce a Qi site Q to a QH2 (Fig. 13).
lation, maintenance of plasma membrane potential, protein Under some conditions, the Q-cycle can be short-circuited
folding by chaperones, axonal and dendritic transport and by various ‘‘bypass’’ reactions, some of which yield the
the release and re-uptake of neurotransmitters at synapses. physiologically deleterious superoxide [2]. The bypass
Mitochondria help neurons to satisfy the high energy reactions are typically observed in vitro under ‘‘partially
demand which is required for proper neuronal function and, inhibited’’ conditions, e.g. in the presence of antimycin A,
unlike other cell types, cannot switch to glycolysis when or under high proton motive force, where it is possible that
oxidative phosphorylation becomes limited. Furthermore, the [2Fe2S] cluster can oxidize QH2 to a semiquinone
cells in response to changing energy demands continually (SQ), but processing of electrons by the low potential chain
adjust the rate of mitochondrial fission and fusion, to is hindered, resulting in the accumulation of a SQ inter-
facilitate the distribution of mitochondria [2, 61]. mediate, which in turn can reduce O2 to superoxide [2].
Mitochondria may produce superoxide in relatively Physiologically, electron transport chain consumes up to
constant amounts, or may elicit spontaneous or environ- 90% of the oxygen taken up by a cell, of which *1% is
mentally-induced ‘‘superoxide flashes’’ [2]. This is con- transformed in superoxide under normal physiological
sistent with the general notion that in the mitochondrial conditions [85]. The rate of mitochondrial electron transfer
respiratory chain complexes I and III are the main sites of from semi-reduced Q to molecular O2 to form superoxide
superoxide production [2]. In this context, any impairment is proportional to the product of the concentrations of semi-
in the electron transfer process determines that upstream reduced Q and O2. It is estimated that the under patho-
carriers become more reduced, thus filling Q cycle electron physiological conditions, superoxide production can occur
pool. The cytochrome (cyt)2 bc1 complex (EC 1.10.2.2) at 2–10% of the uninhibited rates. Superoxide dismutases,
(cyt bc1 complex), localized in the inner membrane of in turn, generate the more stable hydrogen peroxide before
mitochondria, couples the oxidation of a substrate quinol it is transformed by catalase in water or, depending on the
(QH2) with the generating proton motive force across the presence of metals, into highly reactive hydroxyl radicals.
energy transducing membrane system, where the energy is Thus, SOD2, which directs H2O2 out of the mitochondrion,
ultimately stored in the form of ATP (Fig. 1). Cytochrome accomplishes two functions: the classical scavenging of
bc1 complexes contain four redox-active metal centers, superoxide, as well as its signalization as ROS-dependent
arranged in two separate chains. The ‘‘high potential signaling pathways. The binding of CO to cytochromes
chain’’ consists of the Rieske iron-sulfur [2Fe2S] cluster localized within complex IV (i.e., cytochrome a3) pro-
(Fig. 1) and cyt c1. The ‘‘low potential chain,’’ which binds motes reduction in the respiratory carriers in the cyto-
to the cyt b subunit of ubiquinol oxidizing complexes, chrome bc1 region of complex III, associated with
consists of two b-type hemes, cyt bL and bH labeled for increased leakage of superoxide and H2O2 production
their relatively lower and higher electrochemical potentials (Fig. 1).
[84]. Three enzymatic binding sites participate in catalysis Much of the superoxide produced is efficiently con-
on the cyt bc1 complex, the quinol oxidase (Qo) site, verted to hydrogen peroxide via the activity of superoxide
quinol reductase (Qi) site, and a docking site for soluble cyt dismutases including cytoplasmic (SOD1) and mitochon-
c on cyt c1. The Qo site is located toward the positively drial (SOD2) forms of the enzyme. Being a highly reactive
charged side of the membrane, where protons are released free radical, superoxide can damage molecules (DNA,
during turnover of the enzyme (Fig. 1). The Qi site is proteins and lipids), and so its conversion to hydrogen
located toward the negatively charged surface of the peroxide protects cells. However, in the presence of even
membrane, where protons are taken up during catalysis. very low concentrations of Fe2? or Cu?, hydrogen perox-
The water-soluble cyt c docking site is located on the ide can generate hydroxyl radical which is a potent inducer
c-type cyt representing the terminal electron carrier within of membrane lipid peroxidation [33]. In addition, nitric
the cyt bc1 complex on the positive side of the membrane. oxide (which is generated in response to activation of the
Cyt bc1 complex catalysis is thought to occur by a enzyme nitric oxide synthase by Ca2?/calmodulin) can

123
Neurochem Res (2010) 35:1880–1915 1893

interact with mitochondrial superoxide to generate the GTPases optic atrophy-1 (OPA1) and mitofusin-2 (Mfn2)
highly reactive free radical peroxynitrite [41, 86]. Methods occur in Charcot-Marie-Tooth Disease (CMT), a peripheral
have been developed to detect and quantify oxidative neuropathy characterized by muscle weakness and axonal
damage to proteins, lipids and DNA, with antibodies that degradation of sensory and motor neurons, and hereditary
selectively recognize proteins modified by the lipid per- motor and sensory neuropathy type VI, clinically similar to
oxidation product 4-hydroxynonenal [86] and by nitration CMT with the addition of optic atrophy and visual
[41, 86] being particularly useful. In addition, fluorescent impairment, as well as in Autosomal Dominant Optic
probes for imaging relative levels of overall mitochondrial Atrophy, a disease characterized by progressive loss of
redox status and superoxide have been used to elucidate vision and degeneration of the optic nerve and retinal
roles for mitochondrial ROS in a range of physiological ganglion cells. In addition, there is evidence that increased
and pathological processes [87]. The damaging effects of mitochondrial fission occur in Parkinson’s disease (PD)
excessive production of ROS are believed to contribute to a models through two proteins, PTEN-induced kinase 1
wide range of diseases including cancers, cardiovascular (PINK1) and Parkin, which are mutant in familial forms of
disease and inflammatory conditions such as arthritis [88]. PD. Furthermore, mutant huntingtin, the disease-causing
Neurons may be particularly vulnerable to mitochondrial protein in Huntington’s disease (HD), alters mitochondrial
ROS because of their high energy demands, their excit- morphology and dynamics [91]. Rotenone, a pesticide and
ability, and the fact that they are postmitotic and are inducer of PD symptoms, and amyloid-b (Ab) peptide,
therefore in most cases not replaceable [88]. However, which is causally linked to Alzheimer’s disease (AD), are
lower subtoxic levels of mitochondrial ROS can activate able to initiate mitochondrial fission [92].
signaling pathways that protect cells against injury and Posttranslational modifications caused by nitrosative/
disease [2, 86]. oxidative stress may sustain the pathogenesis of the more
Recent evidence suggests that changes in mitochondrial common sporadic cases in neurodegeneration, which
activity can trigger morphological adaptations of the appear to result from complex interactions of multiple
mitochondrial network, and clinical studies further indicate genes and/or their products with environmental factors.
that molecular defects affecting its dynamics lead to Recent studies have identified three post-translational
pathology. This suggests the existence of a link between modifications that seem to regulate mitochondrial fission
energy status and organellar network configuration. With and fusion proteins: phosphorylation, sumoylation and
respect to this, a balance between fusion and fission events, ubiquitination [92]. Protein Kinase A (PKA, cAMP-
mediated by specific proteins that could participate in the dependent protein kinase) phosphorylates Drp1, which
modulation of energy production, occurs controlling results in a significant decrease in GTPase activity and
mitochondrial configuration [89]. In physiological settings, inhibition mitochondrial fission. Interestingly, mitosis-
mitochondria are known to continuously undergo fission promoting factor (MPF, Cdk1/cyclin B) phosphorylates
and fusion (known as mitochondrial dynamics) to generate Drp1 at Ser585 in rats during mitosis. Unlike phosphory-
smaller organelles or rather elongated structures, respec- lation by PKA, Cdk1/cyclin B phosphorylation seems to
tively. Through this process, termed mitochondrial bio- stimulate mitochondrial fission during mitosis. Through
genesis, formation of new mitochondria occur, a process this versatile mechanism, phosphorylation of Drp1 at
associated to repair of defective mitochondrial DNA, different amino acids causes opposite effects [93, 94].
through redistribution of mitochondria to sites with high Sumoylation is another means of Drp1 regulation. SUMO1
energy demand. In neurons, the fission/fusion machinery (small ubiquitin-related modifier-1) interacts with Drp1
proteins, regulated by a machinery involving large dyn- and associates with mitochondria at fission sites before and
amin-related GTPases, maintain mitochondrial integrity after fission [94]. Overexpression of SUMO1 leads to
and insure their presence at critical sites that demand high mitochondrial fragmentation and apoptosis, probably
concentrations of ATP, especially the synapse [90]. These because SUMO1 protects Drp1 from degradation, stabi-
proteins includes Drp1 and Fis1, acting as fission proteins, lizing Drp1 and enhancing its binding to mitochondria.
and Mitofusin (Mfn) and Opa1, operating as fusion pro- Ubiquitination has also been documentated to be related
teins. Given the critical role of mitochondria in neurons, with mitochondrial dynamics [94]. There is evidence that
impaired mitochondrial dynamics is increasingly impli- MARCH-V or MARCH5, a mitochondrial outer membrane
cated in neurodegenerative diseases including Parkinson, protein, ubiquitinates Drp1, thus promoting fission. Finally,
Alzheimer and Huntington diseases [91]. the finding that Bax and Bak, two pro-apoptotic regulators,
Dysfunction in mitochondrial dynamics can result from interact with mitochondrial fission and fusion GTPases,
either genetic mutations in fission- or fusion related genes lends support to the functional link between mitochondrial
or from posttranslational changes to the fission or fusion dynamics and apoptosis. Bax associates with Drp1 and
proteins. Hereditary mutations in the mitochondrial fusion Mfn2 on mitochondria during apoptosis. Furthermore,

123
1894 Neurochem Res (2010) 35:1880–1915

neurons under nitrosative stress exhibit Bax foci on mito- inducing a parallel increase in the formation of superoxide
chondria undergoing mitochondrial fission. Inhibition of from complex III. Superoxide, in turn, induces MnSOD
Drp1 function delays mitochondrial fission, which results overexpression with formation of hydrogen peroxide and
to be associated with Bax foci formation and neuronal cell consequent signalization of the so called preconditioning
death [94]. (Fig. 1). As signaling molecule H2O2 activates protein
An additional potentially deleterious effect of increased kinase B (Akt), which deactivates glycogen synthase
ROS levels is chronic mitochondrial fission. Nitrosative kinase-3b, allowing the nuclear translocation of Nrf2.
stress causes profound mitochondrial fission in neurons Nuclear Nrf2 binds to antioxidant response elements in the
prior to the onset of neuronal loss in an animal model of HO-1, MnSOD, and NFR-1 gene promoters, thus amplify-
stroke and treatment with antioxidants has been shown to ing the initial signal and driving the transcription of TFAM
reduce mitochondrial fission [94]. Amyloid-beta peptide, and other genes that have promoter binding sites for NRF-1
thought to be a key mediator of AD pathogenesis, engenders [2, 3, 41, 86, 99]. These genes are involved in the control of
S-nitrosylation and thus hyperactivation of the mitochon- mitochondrial transcription and protein synthesis, mito-
drial fission protein Drp1. This activation leads to excessive chondrial protein import, and oxidative phosphorylation
mitochondrial fragmentation, bioenergetic compromise, [99]. Consistent with this notion, acetylcarnitine was found
and synaptic damage in models of AD [90]. Expression of to reverses the age-related decrease in the activity of com-
Mfn or dominant-negative Drp1 partially prevented mito- plex III and oxidative phosphorylation through complexes
chondrial fission and neuronal cell death by nitric oxide, III and IV, and increases the amount of cytochrome b and
which is known to be largely caspase independent. Another aa3 hemes in mitochondria isolated from old rats [97]. Of
study found that oxidative stress promoted mitochondrial interest, both cytochrome b and aa3 proteins are encoded by
fission in cerebellar granule neurons and that Mfn2 the mitochondrial genome, suggesting that acetylcarni-
expression was protective [94]. Whether oxidative stress tine enhances either mtDNA transcription, the stability of
directly regulates the mitochondrial fission and fusion mitochondrial mRNA, or mitochondrial protein synthesis
GTPases is currently unclear. However, given that mito- [97].
chondrial integrity is essential to cellular homeostasis, when
the energetic demand is low, excess mitochondria are
unnecessary and can generate excessive reactive oxygen Hormesis
species [95]. In addition, if uncoupled, they can consume
ATP, thus their elimination by autophagy is an efficient Hormesis is a dose response phenomenon characterized by
cytoprotective response. Furthermore, uncoupled or unsta- a low dose stimulation and a high dose inhibition (Fig. 2).
ble mitochondria may release ROS, and various apoptosis- It may be graphically represented by either an inverted
promoting factors, including cytochrome c, AIF, SMAC/ U-shaped dose response or by a J- or U-shaped dose
DIABLO, thus promoting damage to neighboring mito- response. The term hormesis was first presented in the
chondria [96]. published literature in 1943 by Southam and Ehrlich who
Another important event in mitochondrial biology is reported that low doses of extracts from the Red Cider tree
mitochondrial biogenesis, which represents a phenomenon enhanced the proliferation of fungi with the overall shape
relying on a spatiotemporally coordinated synthesis and of the dose response being biphasic. However, credit for
import of approximately 1000 nuclear-encoded proteins, experimentally demonstrating the occurrence of hormesis
some of which are assembled with mitochondrial-encoded goes to Hugo Schulz (1888) [100] who reported biphasic
proteins within newly synthesized inner and outer mito- dose responses in yeast following exposure to a large
chondrial phospholipid membranes. The replication of number of toxic agents. The work of Schulz inspired a
mitochondrial DNA, as well as the mitochondrial fusion and large number of investigators in diverse fields to assess
fission mechanisms, also must be synchronized with these whether such low dose effects may be a general feature of
processes [97]. Oral acetylcarnitine supplementation biological systems. In fact, similar types of dose response
increases mitochondrial content, as well as nuclear tran- observations were subsequently reported by numerous
scripts of factors involved in mitochondrial biogenesis researchers assessing chemicals [101] and radiation
(PGC-1a, NRF-1, TFAM). This event results associated to [102–113] with investigators adopting different names such
increased levels of mitochondrial transcripts for COX I, as the Arndt-Schulz Law, Huppe’s Rule, and other terms to
ATP6 and ND6, as well as 16S rRNA. In the same condi- describe these similar dose response phenomena. Despite
tions, stimulation of vitagenes, in particular HO-1, lead to the rather substantial historical literature concerning hor-
production of carbon monoxide which, according to the metic dose responses, this concept had a difficult time
model proposed by Piantadosi [98], is able to bind to the- being incorporated into routine safety assessment and
reduced heme a3 in the cytochrome c oxidase complex, pharmacological investigations, principally because it

123
Neurochem Res (2010) 35:1880–1915 1895

Fig. 2 Dose-response curve. Quantitative features of hormesis Fig. 3 Citations of Hormesis/Hormetic in Web of Science Database

(1) required more rigorous evaluation in the low dose zone, into all leading textbooks of toxicology [124] encyclope-
(2) failure of investigators to understand its clinical sig- dias [115, 125] and other leading monographs. In fact,
nificance (3) failure to appreciate the quantitative features while the terms hormetic and hormesis were cited only
of the hormetic dose response (4) failure to understand the about 160 times during the entire decade of the 1980’s
limitations of its implications for commercial applications within the Web of Science database, in 2009 alone these
in agriculture as well as medicine, (5) because of the pre- terms were cited nearly 2,500 times (Fig. 3).
dominant interest in responses at relatively high doses Of further significance were observations that these
during most of the twentieth century as well as (6) the broad ranging dose response relationships also shared the
continuing, yet inappropriate, tendency to associate the same general quantitative features. More specifically, the
concept of hormesis with the medical practice of home- low dose stimulation which becomes manifested immedi-
opathy [114–116]. However, from the late 1970’s ately below the pharmacological and toxicological thresh-
[117, 118] there has been a growing interest in hormetic- olds is modest in magnitude being at most only about
like biphasic dose responses across the broad spectrum of 30–60% greater than the control group response. The width
biomedical sciences. This resurgence of interest resulted of the hormetic stimulation is usually about 10–20 fold
from a variety of factors, including the capacity to measure starting immediately from the zero equivalent dose (i.e.
progressively lower doses of drugs and chemicals, the estimated threshold) (Fig. 2). The hormetic dose response
adoption of cell culture methods which has permitted more may result from either a direct stimulation or via an
efficient testing of numerous doses and the need to overcompensation stimulatory response following disrup-
re-examine the validity of linearity at low dose modelling tion in homeostasis [110, 126]. Regardless of the mode of
of cancer risks due to their enormous cost implications for action by which the stimulation occurs the quantitative
regulations, as well the astute observations of independent features of hormetic dose responses are similar. These
investigators and their capacity to generalize their findings observations are based on copious data derived from the
across biological systems [117, 119]. published literature ranging from plants to humans [121,
What has emerged from these research initiatives from 127], involving numerous receptor systems [114, 128–
highly diverse biomedical areas is the recognition that 130]. These findings have recently led to nearly 60 bio-
hormetic dose responses were common and highly gener- medical scientists recommending that biological stress
alizable, being independent of biological model, endpoints responses, including those of pre- and post-conditioning, be
measured and chemical class and/or physical agent studied integrated within an hormetic context, along with the
[110–113, 120–123]. This was an unexpected finding as adoption of a terminology that would be based within an
hormetic responses were often considered by many in the interdisciplinary framework [131].
so-called mainstream branches of toxicology and pharma- The hormetic dose response confers a new set of inter-
cology to be paradoxical, not commonly expected and pretations for the dose response. At high doses within a
being of questionable reliability with a lack of capacity for toxicological setting, the typical endpoints measured indi-
replication. The casual dismissal of the hormesis concept cate cellular damage. However, as the dose decreases
during the mid decades of the last century is reflected in the below the threshold the low dose stimulation more likely
general absence of the hormesis concept from the leading represents a manifestation of an adaptive response that
toxicological and biomedical textbooks. This situation has conforms to a measure of biological performance as may
radically changed such that hormesis is now incorporated be seen in the cases of modest increases in cognition,

123
1896 Neurochem Res (2010) 35:1880–1915

growth, longevity, bone density and other biomedical of considerable human variation in response to a drug, it is
endpoints of interest. possible that the test population may have their respon-
The consistency of the vast array of hormetic findings siveness distributed over a range of responses that includes
suggests strongly that this dose response may be a mani- toxicity, optimal response and a group in which the dose is
festation of the plasticity of biological systems. That ineffective. The data from all subjects in such studies
essentially all biological models respond to imposed stress would normally be averaged together leading to a marked
with the same quantitative features of the dose response is a dilution of an overall positive treatment effect in the
central finding within the biological sciences that has not optimal response zone subgroup. This suggests a possible
been previously recognized. These findings suggest that the reason why drugs that were very successfully tested in
hormetic dose response would have been broadly selected preclinical studies with highly inbred strains of animals
for and highly conserved. This adaptive response not only could and often have failed during the clinical trial. Of
enhances survival by conferring resistance to environ- particular note is that investigators may have to modify
mental stress but it represents a way to regulate the allo- doses based upon the sensitivity or susceptibility of the
cation of biological resources in a manner that ensures subjects. Calabrese and Baldwin [137] have shown that the
cellular and organismal stability. The consistency of the hormetic dose response is often expressed in the broad
hormetic dose response across the vast array of biological range of subjects independent of their susceptibility. As
models, at different levels biological organization, expected, those individuals that are very resistant to the
regardless of the type of stress and health status of the drug or chemical treatment would have their hormetic
individual, indicate that hormesis provides a quantitative response shifted to the right on the dose response graph
index of biological plasticity across multiple levels of whereas those individuals with greater than normal sus-
biological organization, making it a central biological ceptibility would have their hormetic response shifted to
element for enhancing survival. the left. The hormetic dose response therefore imposes
These quantitative features of the hormetic dose response considerable challenges to the biomedical community that
have important medical implications. Most significantly, is interested in the development of drugs that are concerned
the hormetic dose response imposes constraints upon the with improvements in human performance.
magnitude of a drug to induce a desired effect. For example, The hormetic dose response can also have undesirable
if a drug increased cognitive performance in an elderly effects. This may be most readily seen in the case of drugs
patient by approximately 25–30%, the hormetic model that are designed to suppress growth or kill cells or
suggests that this level of performance could not be further organisms at higher doses. For example, there is now
increased using a new drug combination. This concept has substantial evidence that low doses of many antitumor
been supported in a variety of studies on hormesis and drug drugs can stimulate the proliferation of such cells at lower
interaction. Flood [132–135] has demonstrated that the concentrations [138]. This also been shown to be the case
hormetic response for memory was bounded by the 30-60% with antibiotics, including penicillin [139] and streptomy-
increase even when several drugs were used in combination cin [139, 140]. This phenomenon has also been reported
which were designed to maximize memory outcome. This with selected cardiac glycosides that have effects on non-
response magnitude constraint has been reported for target tissues such as the prostate where it is able to
immune stimulation, bacterial growth, increases in hair enhance the proliferation of smooth muscle cells by about
growth, plant growth, decrease in anxiety, decreases in 30% with clinically relevant doses [141, 142]. Such a 30%
tumor incidence and numerous other endpoints [136]. increase in prostate smooth muscle was considered likely
This limitation in the magnitude of the stimulatory to impede urination in males. The failure to consider the
response is a critical implication of the hormesis dose possibility of the hormetic response not only can lead to a
response concept. It is an observation which is based on lack of recognition of a desirable drug induced response
extensive findings and it is a controlling feature which but it can also result in failure to prevent an adverse effect
defines what pharmaceutical companies can expect to of drug treatment.
achieve with drugs that are designed to enhance perfor- Since hormetic dose responses have now been widely
mance. However, the limitation in the magnitude of reported in biological systems, there is the desire to
response is also potentially important with respect to the develop a new subfield of hormetic mimetics. These mi-
capacity to detect a desirable response. This may not be a metics are agents that can activate hormetic pathways with
particularly important issue when using highly inbred the intention of producing a desirable clinical effect. For
animal models or cell cultures where experimental condi- example, it would be desirable to develop agents that can
tions can be highly controlled. However, attempting to induce the desirable adaptive hormetic response without
measure a low dose hormetic stimulation within the context having risks associated with the exposures as in the case of
of a clinical trial can be problematic. Given the likelihood certain chemical agents and radiation.

123
Neurochem Res (2010) 35:1880–1915 1897

Mitostress Versus Mithormesis and Neuroprotection up-regulates the expression of Mn-SOD via an NF-kB-
mediated mechanism, thereby protecting neurons against
Recent findings have overturned the long-held belief that excitotoxic, ischemic and oxidative injuries [148]. While
mitochondrial ROS have only a negative impact on cell mitochondrial H2O2 may activate adaptive stress response
function and survival. It is now evident that mitochondrial pathways in neurons, they may also play neuroprotective
superoxide and hydrogen peroxide play important roles in roles by acting on other cell types in the nervous system.
a range of cellular functions, and can also activate sig- For example, microglial activation, which occurs in
naling pathways that promote cell survival and disease response to increased oxidative stress, can have either
resistance. Exposure of hippocampal neurons to subtoxic beneficial or detrimental effects on neurons and neural
levels of hydrogen peroxide triggers the release of Ca2? progenitor cells depending upon the type and amounts of
from the endoplasmic reticulum by causing the opening of cytokines and growth factors secreted by the microglia
both IP3 and ryanodine receptor channels [143]. Interest- [149]. In addition, oxidative stress can stimulate angio-
ingly, superoxide enhances long-term potentiation of genesis in the brain [2], which is critical to restore normal
synaptic transmission in hippocampal CA1 neurons by a neuronal perfusion during the days and weeks after a
mechanism requiring activation of ryanodine receptors stroke. This last effect represents an example of ‘‘trans-
and extracellular regulated kinases [144]. Mitochondrial cellular’’ hormesis mediated by ROS [150].
ROS may also play roles in recovery from injury. For
example, superoxide can stimulate neurite outgrowth by
directly activating protein kinase C [145]. Moreover, Redox-Dependent Control of the Proteostasis Network:
peroxynitrite can promote the phosphorylation and nitra- Relevance to Antidegenerative Strategies
tion of regulatory sites within receptor tyrosine kinases
thereby activating cell survival signaling pathways Neurodegenerative disorders are often characterized by the
including those involving PI3 kinase–Akt and mitogen- aggregation and accumulation of misfolded proteins (e.g.
activated protein (MAP) kinases [2]. Nitration of proteins Alzheimer’s disease, Parkinson’s disease, Amyotrophic
involved in synaptic vesicle trafficking can enhance glu- lateral sclerosis, Huntington’s disease (HD) and fronto-
tamate release, suggesting a potential role for superoxide temporal dementia). Aggregated proteins are very toxic to
and peroxynitrite in regulating neurotransmission [2]. cells in culture and both in vitro and in vivo there is
Mitochondrial superoxide production, it is known, con- overwhelming evidence that these aberrant proteins are key
tributes to damage of neurons in pathological conditions players in neurodegeneration. Protein quality control is a
such as chronic cerebral hypoxia or Alzheimer’s disease cellular defense mechanism against misfolded proteins that
[2]. However, it has been widely reported that transient prevents aggregate formation under physiological condi-
exposure of neurons to low levels of superoxide which are tions. The presence of accumulated aggregates of mis-
converted into hydrogen peroxide can protect the neurons folded proteins in many neurodegenerative disorders,
against a subsequent exposure to what would have suggests that protein quality control failed to restore
otherwise been a lethal level of stress. This neuroprotec- homeostasis in these pathological conditions. In fact, evi-
tive effect of a subtoxic increase in cellular oxidative dence from observations in cellular disease models, mouse
stress has been termed ‘‘preconditioning’’ and clearly falls models, as well as from post mortem patient material
under the paradigm of hormesis [2]. Although the indicates activation of the quality control machinery in
involvement of oxidants in many signaling pathways is response to the pathological process. In addition, interfer-
well documented, the cellular strategies for conferring ence with protein quality control by genetic or chemical
pathway specificity to such reactive molecules is still manipulation often results in aggregate formation and
elusive. Recent studies suggest that cells may spatially neurodegeneration. This stresses the importance of proper
restrict oxidant production to allow microdomain-specific quality control in neurodegenerative disorders and indi-
signaling [146]. The specific molecular mechanisms by cates that it may provide a target for therapeutic inter-
which mitochondrial ROS elicit hormetic responses in vention [1–3, 15, 31, 86, 151–153]. A wide range of
neurons are poorly understood, but emerging evidence cellular defence mechanisms operate as a proteostasis
suggests important roles for certain transcription factors network, to counteract this effect, including antioxidant
with regulatory effects. For example, NF-kB is activated proteins [superoxide dismutase 1and 2 (SOD1, SOD2),
in neurons in response to oxidative stress and plays a catalase, glutathione peroxidase, thioredoxin reductase,
pivotal role in the adaptive response that protects the heme oxygenase (HO-1) and cystationine-b synthase
neurons against more severe oxidative stress, by induc- (CBS)], glutathione, molecular chaperones such as the heat
ing the expression of protective genes against oxidative shock proteins (HSPs) that reduce aggregation and promote
stress including SOD2 and Bcl-2 [147], as well TNF folding of misfolded proteins [154].

123
1898 Neurochem Res (2010) 35:1880–1915

The proteostasis network regulates protein function [1, performed by ribosomes on the cytosolic surface of the ER,
155–158], as an integrated biological system evolved to and the unfolded polypeptide chains are translocated into
protect protein fold. As such, proteostasis controls con- the ER lumen via the Sec61 complex [161]. In the ER
centration, conformation, binding interactions (quaternary lumen these chains are often N-glycosylated and folded
structure), and location of individual proteins making up into secondary and tertiary structures that are stabilized by
the proteome by readapting the innate biology of the cell, disulfide bonds [162]. Most glycolpolypeptides thus elab-
often through transcriptional and translational changes [1]. orated are released from calnexin/calreticulin/ERp57 in a
Proteostasis thus influences specific cellular functions and native, transport competent state, are rapidly deglycosy-
enables differentiated cells to change their physiology for lated and sequestered in transport vesicles that leave the
successful organismal development and aging in the face of ER. Glycoproteins presenting not complete folding after a
constant intrinsic and environmental challenges to prevent single step of association with calnexin/calreticulin/ERp57
disease onset. Proteostasis is influenced by the chemistry of and polypeptides, undergo an additional folding attempt
protein folding/misfolding and by numerous regulated characterized by disulfide rearrangement or reshuffling.
networks of interacting and competing biological pathways Glycopolypeptides released from calnexin which display
that influence protein synthesis, folding, trafficking, dis- major folding defects are finally targeted by BiP/Grp78 and
aggregation, and degradation. The composition of the dislocated across the ER membrane for proteasome-medi-
proteostasis network is regulated by signaling pathways ated degradation by ER-associated degradation (ERAD)
including (1) the unfolded protein response (UPR) that [163].
increases the production of endoplasmic reticulum (ER) ERAD is a mechanism employed by ER protein quality
chaperones and suppresses translation, (2) the heat shock control system and the calnexin cycle to eliminate mis-
response (HSR), (3) the ubiquitin proteasome (UPS) for folded or unassembled proteins generated during the fold-
removal of proteins and (4) the authophagy degradation ing process [163]. ERAD impairment is implicated in some
pathway that targets long lived proteins and organelles for neurodegenerative diseases associated to protein misfold-
removal, together with epigenetic programs [159]. At this ing, such as Huntington’s, Parkinson’s diseases and ALS
level, regulation of cellular redox state becomes a crucial [160]. Chaperones, transmembrane proteins and ubiquitin-
mediator of multiple metabolic, signalling and transcrip- associated enzymes selecting target misfolded proteins to
tional processes operating within this integrated network the cytoplasm for degradation by the proteasome system
machinery [2, 41, 152, 153, 156]. comprise the ERAD machinery [163], which includes
the ER degradation-enhancing mannosidase-like lectins
ER Pathway (EDEMs), consisting of mannosidase-like proteins directly
involved in recognition and targeting of unfolded proteins
The ER is a highly dynamic organelle with a central role in for degradation [163]. Three EDEM homologues, EDEM-
lipid and protein biosynthesis. The ER produces the 1, -2 and -3 have been identified, and probably their
transmembrane proteins and lipids for most cell organelles function is similar, delivering ERAD substrates to the
and is responsible for the synthesis of almost all secreted retrotranslocation channel [160]. EDEM1 associates with
proteins. Synthesized and correctly folded proteins that the DERLIN family of proteins [161], which are ER-
pass the quality control are transported through the secre- membrane proteins components of the retrotranslocation
tory pathway to reach their final destination including cell channel, besides to Sec61p. It is estimated that the protein
surface, lysosomes, GA, extracellular space, and the ER. concentration within the ER lumen reaches 100 mg/ml, a
After translocation to the ER through the Sec61p translo- concentration highly prone to protein aggregation that is
con complex channel, nascent proteins are assisted in their avoided by an effective folding quality control system
folding by a complex family of chaperones, foldases and [164]. Conditions that interfere with the ER function con-
co-factors [160]. The ER also has an important role in Ca2? sequently lead to abnormal protein folding and to the
storage and signaling. The resting intra-ER Ca2? concen- accumulation of unfolded/misfolded proteins on its lumen,
tration is three to four orders of magnitude higher than a cellular condition referred as ‘ER stress’ [160]. ER stress
cytosolic Ca2?. This gradient is generated by the can be originated by various cellular alterations, such as
sarco(endo)plasmic reticulum Ca2? ATPase (SERCA) calcium homeostasis disruption, redox changes, reduction
proteins, which pump Ca2? into the ER, and the of disulfide bonds, glucose/energy deprivation, viral
Ins(1,4,5)P3 and ryanodine receptors that release Ca2? infections, accumulation of folding-incompetent products
from the ER [2]. Due to its ability to store and secrete of mutated genes, and high secretory activity, among others
Ca2?, the ER controls a wide range of cellular processes [2]. To manage protein folding stress, cells activate adap-
such as organogenesis, transcriptional activity, stress tive pathways known as the Unfolded Protein Response
responses, and apoptosis [2]. The translation of proteins is (UPR), which aims to re-establish cell homeostasis [2]. The

123
Neurochem Res (2010) 35:1880–1915 1899

unique oxidizing environment of the ER and the numerous in signal transduction processes or cellular stress responses.
protein chaperones present in the organelle are crucial for We now know that the ER plays pivotal roles in regulating
the proper folding of proteins and protein complexes [118]. cellular Ca2? homeostasis and transduces Ca2?-mediated
The disulfide bond formation is catalyzed by protein signals from extracellular signals (excitatory neurotrans-
disulfide isomerase (PDI) [165]. PDI is oxidized by Ero1-a mitters and growth factors) and signals emanating from
and -b into a disulfide donor, whereas reduced PDI can internal organelles including mitochondria and the nucleus
isomerize disulfide bonds in client proteins. Other ER- [166]. When stimulated by IP3 or Ca2? itself, Ca2? is
resident folding factors include amino acid cis–trans released from the ER through opened IP3 receptor and
isomerases, the chaperones GRP94 and Ig heavy chain ryanodine receptor channels, respectively. An ER mem-
binding protein (BiP), N-glycosylation enzymes and the brane Ca2? ATPase is responsible for transporting Ca2?
lectins calnexin and calreticulin that specifically chaperone into the ER. Excessive ER stress and dysregulation of ER
N-glycans [165], all of which operate in complex multi- Ca2? homeostasis is believed to contribute to the dys-
protein structures [162]. While assisting with folding, these function and death of neurons in ischemic stroke, Parkin-
chaperones and foldases bound client proteins in the ER son’s disease and AD [162]. The ER is also an important
until the maturely folded proteins meet all quality control sensor of cellular stress. In response to energetic, oxidative
standards and exit the ER [165]. Thus, the ER is exquisitely and ionic (particularly Ca2?) stress, protein synthesis may
sensitive to alterations in homeostasis, and proteins formed be decreased, and signals are sent from the ER to the
in the ER may fail to attain correct conformation due to: (1) nucleus that result in the up-regulation of the expression of
lack of chaperones or cellular energy to promote chaper- genes encoding ER protein chaperones such as GRP78 and
one-protein interactions; (2) Ca2? depletion; (3) disruption GRP94, and antioxidant enzymes [163]. The ER plays
of redox state; (4) protein mutations that hamper adequate important roles in hormetic responses to stress in neurons.
folding; and (5) reduction of disulfide bonds [2]. Bcl-2 can decrease ER Ca2? uptake, resulting in enhanced
Eukaryotic cells respond to unfolded proteins in their Ca2? signaling and activation of cyclic AMP response
endoplasmic reticulum (ER stress), amino acid starvation, element binding protein, thereby promoting the regenera-
or oxidants by phosphorylating the alpha subunit of tion of injured axons [167]. Moreover, ER-mediated sig-
translation initiation factor 2 (eIF2a). This adaptation nals associated with the UPR enhance antioxidant defenses
inhibits general protein synthesis while promoting trans- and inhibits caspase-dependent cell death. Thus, mild ER
lation and expression of the transcription factor ATF4 [2]. stress can elicit multiple hormetic responses in neurons. A
Atf4(-/-) cells are impaired in expressing genes involved better understanding of adaptive responses to ER stress
in amino acid import, glutathione biosynthesis, and resis- may lead to novel therapeutic approaches for both acute
tance to oxidative stress [2]. Under low stress conditions, and chronic neurodegenerative conditions. As evidence, it
GRP78 is associated with several different ER proteins has been reported that, by activating ryanodine receptors in
including PERK (PKR-like ER kinase), IRE-1 (inositol the ER, paraxanthine (the major metabolite of caffeine) can
requiring element-1) and ATF6 (activating transcription prevent the degeneration of dopaminergic neurons in a
factor 6). When cells are under oxidative and metabolic model of Parkinson’s disease [2].
stress, GRP78 is recruited to oxidatively damaged and Induction of ER protein chaperones is another thera-
misfolded proteins, and PERK, IRE-1 and ATF-6 are peutic approach that is supported by experimental data
mobilized to serve the complementary roles in the UPR. showing that GRP78, GRP94 and ORP150 [168] can pro-
PERK suppresses protein translation by phosphorylating tect neurons against excitotoxic and ischemic death. Mild
eIR2a, and also phosphorylates and thereby activates the ER stress responses may be elicited by factors that are
transcription factor Nrf2 resulting in the production of known to be neuroprotective including mild energetic
antioxidant and phase 2 enzymes. IRE-1 activation results stress (dietary energy restriction and 2-deoxyglucose
in the production and nuclear translocation of the tran- treatment and exercise [169]. Adaptation to ER stress is
scriptional regulator XBP-1 which, in turn, induces the mediated by the activation of an integrated signal trans-
expression of GRP78 and GRP94. During the UPR, ATF-6 duction pathway known as the unfolded protein response
is cleaved to generate an active transcription factor which (UPR). In neurodegeneration, either in mouse models or in
translocates into the nucleus and induces the expression of post-mortem brain from ND patients, activation of the UPR
several cytoprotective proteins. Examples of stimuli that correlates with the amount of aggregated proteins [170].
activate the UPR include nutrient deprivation, altered Moreover, the UPR has generally been viewed as a
protein glycosylation, oxidative stress and increased in- homeostatic mechanism designed to avoid the accumula-
tralumenal Ca2? levels. tion of toxic protein aggregates. However, evidence exists
Until recently, the endoplasmic reticulum (ER) was that deletion of X Box binding protein-1 (XBP-1), a key
viewed as a protein synthesis factory that was not involved UPR transcription factor that regulates genes involved in

123
1900 Neurochem Res (2010) 35:1880–1915

protein folding and quality control, by disrupting UPR, can neurological disorders [160]. In contrast, lysosomal-related
prevent neurodegeneration [171]. Although counterintui- compartments are able to degrade substrates such as pro-
tive, this finding can be explained in several integrated tein complexes and organelles. Increasing evidence indi-
ways: (1) mild, UPR stimulates the adaptive up-regulation cates that the clearance of proteins through autophagy
of chaperones, thus resulting cytoprotection, whereas pro- correlates with their propensity to aggregate. In light of the
longed UPR activation results in Bax-mediated activation evidence that many aggregate-prone proteins cause many
of pro-apoptotic pathways depending on c-Jun N-terminal distinct diseases possibly by gain of function mechanisms,
kinase (JNK) stimulation [172]. In addition, ablation of efforts have been made to explore therapeutic strategies
CHOP, a transcription factor involved in the UPR, restores that reduce the levels of such proteins, at either the syn-
motor function in a mouse model of Charcot Marie Tooth thesis or degradation stage [160]. Recent studies indicate a
1B neuropathy, possibly via inhibition of ER stress- crucial role of autophagy in protein quality control, with a
induced apoptosis [173]; (2) inhibition of the ER-associ- demonstrated function in the maintenance of the homeo-
ated degradation (ERAD) pathway consequent to the stasis of the nervous system [172]. Notably, p62 (also
ablation of XBP-1 might explain the compensatory stim- termed sequestosome 1) interplays between polyubiquiti-
ulation of autophagy; (3) accumulation of unfolded or nation and the autophagy-degradative mechanisms, thus
misfolded proteins due to the absent UPR leads to shuttling substrates of the proteasome system. Compared to
exhaustive ER stress, which in turn induces autophagy via the latter, autophagy is less selective, but emerging evi-
activation of protein kinase C h (PKC h) [174]; (4) dis- dence indicates that autophagy is highly regulated and
turbance of the UPR induces oxidative stress and depletion specific for the different target organelles, such as mito-
of glutathione, which activate UPR-dependent cell death chondria (mitophagy) endoplasmic reticulum (reticulo-
[172], (5) oxidative stress can also stimulate autophagy by phagy), peroxisomes (pexophagy), ribosomes (ribophagy),
activating ATG4, a cysteine redox-sensitive protease. granules (crinophagy), and pathogens (xenophagy). Cyto-
Thus, at sublethal levels, ROS can act as signaling mole- sol, cytoskeleton, nuclei (nucleophagy), and protein
cules for the induction of cytoprotection and can even aggregates (aggrephagy) can also be removed by
prolong life span, as described in C. elegans under glucose autophagy.
restriction [175]. Several upstream regulators of autophagy have been
characterized [176]. The class III PI3 kinase complex,
Autophagy Pathway including Beclin-1/Atg6, is required for generation of pre-
autophagosome structures. The mammalian target of rap-
Autophagy is the cellular pathway of ‘‘self digestion,’’ a amycin (mTOR), a nutrient-sensing kinase complex that
regulated lysosomal pathway for the degradation and regulates cell growth and survival, blocks autophagy dur-
recycling of long-lived proteins and organelles. During ing nutrient-rich conditions by inhibiting the Atg1 com-
autophagy, cytoplasmic constituents are sequestered into plex, which is involved in the initiation stages of
double-membraned autophagosomes, which are delivered autophagic activity. Upstream activators of mTOR (NF-kB,
to lysosomes and degraded. This process generates nucle- class I PI3 kinase, Akt and NF-kB) suppress autophagy,
otides, amino acids, and fatty acids, which are recycled for whereas inhibitors of this pathway such as PTEN (phos-
ATP generation and macromolecular synthesis [176]. phatase and tensin homologue) induce autophagy.
While the UPR controls the degradation of smaller units Accordingly, rapamycin, a classical mTOR inhibitor, has
of unfolded or misfolded proteins via transcription of been shown, in vivo, to alleviate neurodegeneration in
chaperones, larger aggregates are detoxified via autophagy. models of Huntington’s disease, associated with reduced
Autophagy involves the encapsulation of cargo on a dou- neuronal loss, improved survival and degradation of toxic
ble-membrane vesicle to form the autophagosome. Auto- protein aggregates [172]. Atg proteins regulate steps which
phagosomes fuse with lysosomes to form the autolysosome are sequential in the autophagy process, whose core is
where the cargo is degraded. In addition, hybrid interme- organized around two ubiquitin-like conjugation systems,
diates called amphisomes are formed when autophago- such as the ubiquitin-like protein Atg12 and the ubiquitin-
somes fuse with the endosomes or MVEs, before fusing like protein LC3-I. When Atg12 is activated, it forms a
with lysosomes. The ubiquitin-proteasome and autophagy- covalent bond with Atg5, which then interacts with other
lysosome pathways are the two main routes of protein components to form a multimeric complex that translocates
clearance in eukaryotic cells [176]. Proteasomes predomi- to membranes of early autophagosomes. Alternatively,
nantly degrade short-lived nuclear and cytosolic proteins. LC3 is cleaved and then conjugated to phosphatidyletha-
However, substrates need to be unfolded to pass through nolamine (PE) via Atg7 and Atg3 (another ubiquitin carrier
the narrow pore of the proteasome barrel, which excludes protein (E2)-like protein), generating LC3-II. The uncon-
the clearance of aggregated proteins observed in many jugated LC3-I remains in the cytosol while the conjugated

123
Neurochem Res (2010) 35:1880–1915 1901

LC3-II form targets to the autophagosomal membrane after proteins are converted into toxic or benign forms. The
the formation of the active Atg12–Atg5 complex [177]. As endomembrane neuronal network divides brain cells into
elongation of the autophagosomal membrane occurs, different subcellular compartments that possess distinct
cytoplasmic proteins and organelles are sequestered into sets of molecular chaperones and protein interaction
double-membraned autophagosomes, which are delivered networks [2]. Cells buffer proteotoxic events related to
to lysosomes and degraded [176]. Autophagy is critical for intracellular protein misfolding via chaperone-mediated
the maintenance of neuronal homeostasis and contributes partitioning of nonnative conformers between pathways for
to basal elimination of misfolded proteins in the nervous proper folding, inclusion body formation, and degradation.
system. Brain specific ablation of the essential autophagy- Chaperones, in fact, act as agonists and antagonists of
related genes atg5 and atg7 leads to spontaneous neuro- disease protein aggregation to prevent the accumulation of
degeneration with pathological features closely resembling toxic intermediates in the aggregation pathway. Interacting
Alzheimer’s and Parkinson’s disease consisting of neuro- partners can also modulate the conformation and locali-
logical/motor dysfunction, accumulation of poly-ubiquiti- zation of disease proteins and thereby influence proteo-
nated protein aggregates, neuronal loss and premature toxicity [152]. Thus, interplay between these protein
death [160]. Similar to UPR, a dual role in cell survival or homeostasis network components can modulate the self-
cell death has been attributed to autophagy. Autophagic association of disease proteins and determine whether they
degradation of cellular materials generates amino acids and elicit a toxic or benign outcome [2]. Polypeptides that fail
fatty acids, which can be used for protein synthesis and to fold properly, along with damaged and oxidized mature
ATP generation during stressful conditions such as star- proteins, are targeted for degradation by specialized cel-
vation, as well as removal of protein aggregates (which can lular degradation machineries. A failure to prevent the
trigger apoptosis) and damaged mitochondria (source of misfolding and aggregation of one protein can destabilize
apoptotic proteins and toxic ROS) result to be protective the proteome, resulting in uncontrolled aggregation of
mechanisms. Conversely, prolonged autophagy can lead other polypeptides. When conformationally challenged
to cell death through excessive self-digestion or activation aggregation-prone proteins are expressed, the resulting
of apoptosis. Consistent with this notion, prosurvival unfolded or misfolded proteins are rapidly degraded via the
autophagy in response to starvation, growth factor with- ubiquitin–proteasome pathway. However, in some cases,
drawal, ischemia/reperfusion injury, and various chemo- protein aggregation leads to the development of so-called
therapeutic drugs [178, 179] has been proven, as well as ‘conformational diseases’. Among the conformational
autophagic cell death has been observed in response to diseases are the human neurodegenerative diseases, such as
hypoxia, oxidative stress, radiation, GF withdrawal, lipo- Alzheimer’s (AD), Parkinson’s (PD) and Huntington’s
polysaccharide, overexpression of smARF, and various (HD). In AD, dual digestion of amyloid precursor protein
chemotherapeutic drugs [178]. (APP) releases the aggregation-prone peptides that are
collectively termed amyloid-b (Ab). The accumulation and
aggregation of Ab (particularly the highly aggregative
Cellular Stress Response and The Vitagene Network Ab1–42) is associated with AD. Similarly, aberrant
aggregation of a-synuclein is associated with the emer-
Protein thiols and, more in general, protein quality control gence of PD, whereas Huntington and other CAG triplet
processes, play a critical role in cellular homeostasis which diseases are typified by the accumulation of polyglutamine-
is maintained by a highly complex network of molecular containing aggregates. This also includes prion diseases
interactions that balances protein biosynthesis, folding, such as Creutzfeldt-Jakob disease with accumulation of
translocation, assembly/disassembly, and clearance [151]. misfolded prion protein, type II diabetes with accumulation
The ability to ensure proper protein folding is critical for of islet amyloid polypeptide, and amyotrophic lateral
cellular function and organismal viability. In cells under- sclerosis with aggregated superoxide dismutase-1 [153]. As
going division, damaged and oxidized proteins can be one of the most characteristic feature present in most brain
sequestered and retained in mother cells, enabling daughter oxidant disorders is the occurrence of extra- or intracellular
cells to have a pristine, undamaged proteome. However, in fibrillar aggregates containing misfolded proteins with
post-mitotic cells, such as most neurons, protein quality beta-sheet conformation. These aggregates are composed
control must be maintained by other, possibly more com- of distinct proteins in each neurodegenerative disease. As
plex, mechanisms [152]. Furthermore, particular neuronal already mentioned, toxicity deriving from misfolded pro-
populations are more vulnerable to proteotoxicity while teins or peptides are collectively termed proteotoxicity, and
others are more able to tolerate the misfolding and aggre- it has become evident that protein aggregation is tightly
gation of disease proteins. Thus, the cellular environment linked to the emergence and development of neurodegen-
must play a significant role in determining whether disease erative diseases. Moreover, the mechanisms that have been

123
1902 Neurochem Res (2010) 35:1880–1915

found to counter toxic protein aggregation in different cell oxidative stress (MAOS), thereby impairing mitochondrial
types and different organisms are highly conserved. Thus, function and thus causing degenerative cell death [155].
it is likely that proteotoxicity that is associated with the Cellular stress response is the ability of a cell to coun-
expression of neurodegeneration-linked aggregation-prone teract stressful conditions (Fig. 4). This phenomenon,
proteins in any tissue can provide insights into the neuronal which includes heat shock response (HSR), represents an
defence mechanisms. Recent data suggest that small olig- ancient and highly conserved cytoprotective mechanism
omeric aggregating structures, termed also protofibrils, are [2, 3, 14]. Production of heat shock proteins, including
the underlying cause, rather than high-molecular-mass protein chaperones, is essential for the folding and repair of
aggregates as previously assumed. In vitro studies on damaged proteins, serving thus to promote cell survival
fragments of amyloidogenic proteins and synthetic pep- conditions that would otherwise result in apoptosis
tides have established that the tendency for a protein to [15, 16]. The term ‘molecular chaperone’ denotes a large
form amyloid is often limited to a short sequence of the full family of ubiquitous proteins that function as part of an
protein, known as an SRE (selfrecognition element). SREs ancient defense system in our cells. Chaperones promote
form the core of amyloid fibrils. These amyloidogenic cell survival by sequestering damaged proteins and pre-
sequences constitute ‘hotspots’ for aggregation of the venting their aggregation. During stressful conditions, such
native protein into amyloid fibrils and are often the sites of as elevated temperature, they prevent protein aggregation
mutations leading to early-onset amyloidosis. In the case of by facilitating the refolding or elimination of misfolded
tau-paired helical filaments, which accumulate in the neu- proteins. The stress-induced response to damaged proteins
rofibrillary tangles characteristic of AD and other neuro- is helped by a sophisticated regulatory system, which shuts
degenerative diseases, it has been shown that only three down most cellular functions and, in parallel, induces the
residues (Val-Tyr-Lys), are sufficient for fibril formation. synthesis of several chaperones and other survival-pro-
Similarly, short sequences forming the core domain of moting proteins. Therefore, many of the chaperones are
various amyloid fibrils have been identified for Ab (amy- also called stress or ‘heat shock’ proteins in reference to the
loid b-peptide), calcitonin, IAPP (islet amyloid polypep- archetype of cellular stress, heat shock. Besides their role
tide), insulin and a-synuclein. It may be possible to delete during stress, chaperones have multiple roles under normal
residues freely on either side of an SRE while retaining the conditions. They promote the transport of macromole-
ability to form amyloid [154]. Genetic and age-related cules (e.g. proteins or RNA) and participate in remodel-
factors, as it is known, act as vicious cycle increasing the ling events involving larger protein complexes, including
amounts of pathogenic proteins in AD where, the increase signaling, transcription, cell division, migration and
in Ab42 levels is caused by: (1) mutations in amyloid differentiation.
precursor protein or presenilins (for example, c-secretase), Molecular chaperones both in the cytosol (heat-shock
(2) by reactive oxygen species and (3) by reductions in proteins, crystallins, prefoldin, Hsc70,) in the endoplasmic
Ab-degrading enzymes (AbDE), such as neprilysin and reticulum (Bip, Grp94, calnexin, calreticulin) form large
insulin-degrading enzyme, as well as increases in tau complexes and have a large number of co-chaperones to
concentrations are influenced by oxidant damage, phos- regulate their activity, binding properties and function
phorylation and calcium; in PD, where increased levels of [151]. These chaperone complexes regulate local protein
a-synuclein caused by triplication of its gene or mutations networks, such as the mitochondrial protein transport
in parkin, DJ1, ubiquitin carboxy-terminal hydrolase 1 apparatus and the assembly and substrate specificity of the
(UCHL1), phosphatase and tensin homologue induced major cytoplasmic proteolytic system, the proteasome
kinase 1 (PINK1) or leucine-rich repeat kinase 2 (LRKK2), [156]. Chaperones has been demonstrated to protect the
are associated with proteasome impairment and oxidative cell nucleus after stress and to promote the transport of
stress; in HD, with polyglutamine expansions in huntingtin ribosomal subunits and the mobility of steroid receptors
(HTT). The protein aggregation process itself is enhanced inside the nucleus and, in addiction, regulate both the
by: increasing protein concentration; the action of trans- activation and the disassembly of numerous transcrip-
glutaminases; protein chaperone insufficiency; mutations in tional complexes, emerging thus as regulators critical
a-synuclein (PD) and polyglutamine expansions in hun- transcriptional networks [157]. Stress-induced nuclear
tingtin (HD); and/or post-translational modifications, such translocation of chaperones may preserve also nuclear
as oxidations induced by, for example, hydrogen peroxide remodelling capacity during environmental damage, and
(H2O2), Fe2? and Cu?, and phosphorylation. Although thus protect the integrity of DNA. Consistently, there is
the proteins involved can differ, there is considerable significant interest in the discovery and development of
overlap in the mechanisms by which they damage and kill small molecules that modulate heat shock responses and
neurons. Oligomers of Ab, a-synuclein and HTT might parallel stress response pathways for therapeutic purposes
damage and kill neurons by inducing membrane-associated [2, 3, 14, 16].

123
Neurochem Res (2010) 35:1880–1915 1903

Fig. 4 Cell stress responses. Expression of HS genes is induced in repressed state before stress, are titrate away by damaged or
response to physiological and environmental stress conditions misfolded proteins with resulting HSF-1 activation. Multi-step
including longevity stimuli, such as fasting, caloric restriction or activation of HSF1 involves post-translational modifications, such
acetylcarnitine, and protein conformational diseases. HSF1 can also as hyperphosphorylation and deacetylation, which allow HSF1 to
be directly stimulated by longevity stimuli such as the histone trimerize, translocate into the nucleus, and bind to heat-shock
deacetylase SIRT1 that directly activates HSF1 by deacetylation, thus elements (HSEs) in the promoter regions of its target hsp genes.
fostering longevity. Cumulating misfolded proteins in response to During aging, a gradual decline in potency of the heat shock response
proteotoxic environmental stress conditions triggers the cellular stress occur and this may prevent repair of protein damage, leading to
response. HSPs that are normally bound to HSF1, maintaining it in a degeneration and cell death

Cellular stress response (Fig. 5) requires the activation aggregation when the concentration of the aggregating
of pro-survival pathways which, under control of protective protein is high. Consistent with this notion, although pro-
genes called vitagenes [2, 3] produce molecules (heat tein aggregation is hazardous under certain circumstances,
shock proteins, glutathione, bilirubin) endowed with anti- the creation of apparently less-toxic large aggregates is
oxidant and anti-apoptotic activities. Generally, molecular protective. This hypothesis is the basis of the therapeutic
chaperones help hundreds of signaling molecules to keep potential of heat shock proteins (HSPs), which prevent
their activation-competent state, and regulate various sig- protein misfolding and aggregation. Transgenic animal
naling processes ranging from signaling at the plasma models of the diseases have demonstrated that induction or
membrane to transcription. In addition to these specific overexpression of HSPs can suppress neuronal dysfunction
regulatory roles, recent studies have revealed that chaper- and degeneration. Hsp70, in fact it has can reduce the
ones act as genetic buffers stabilizing the phenotypes of amount of misfolded and aggregated a-Syn species in vivo
various cells and organisms [82, 158]. Among the cellular and in vitro, and protect neuronal cells from a-Syn-
pathways conferring protection against oxidative stress, a dependent neurotoxicity [180], highlighting some of the
key role is played by the products of vitagenes [2, 3, 14–16, intriguing aspects of HSP-based therapy.
27, 41]. These include members of the heat shock protein
(Hsp) family, such as heme oxygenase-1 and Hsp72, sir-
tuins and the thioredoxin/thioredoxin reductase system [42, The Carnitine System: Physiological Roles,
43, 57, 86]. Recent studies have shown that the heat shock Insufficiency and Interplay With Mitochondrial
response contributes to establishing a cytoprotective state Bioenergetics in Aging and Neurodegenerative
in a wide variety of human diseases, including inflamma- Disorders
tion, cancer, aging and neurodegenerative disorders [35].
Given the broad cytoprotective properties of the heat shock Carnitine is present in mammalian cells as free carnitine
response there is now strong interest in discovering and acylcarnitines. While carnitine’s most widely known
and developing pharmacological agents capable of induc- function is its involvement in b-oxidation of fatty acids, it
ing the heat shock response [2]. Molecular chaperones are may also have other roles in metabolism. The importance
known to disrupt aggregates but also to promote active of acylcarnitines in tissues with high rates of b-oxidation

123
1904 Neurochem Res (2010) 35:1880–1915

biological role is to facilitate the transport of fatty acids to


mitochondria for b-oxidation, it facilitates oxidative
metabolism of carbohydrates, increases phosphorylation
oxidative reactions, promotes the excretion of accumulat-
ing organic acids [184–186]. Thus, carnitine functions as
an acyl group acceptor that facilitates mitochondrial export
of excess carbons in the form of acylcarnitines. Whole
body carnitine homeostasis is regulated at multiple levels,
including dietary intake, intestinal absorption, de novo
biosynthesis, and renal reabsorption. Humans obtain car-
nitine from their diet and through endogenous biosynthesis.
Dietary LC derives from the intake of red meats and dairy,
the endogenous synthesis of LC from the aminoacid pre-
cursors L-lysine and L-methionine, mostly in liver and
Fig. 5 Chemical structures of carnitine and acetylcarnitine molecules
kidney, trough different metabolic stages for wich ascorbic
acid, niacina, piridossin and iron are necessary [187, 188].
such as heart and muscle is intuitive. However, acylcarni- The dietary intake of LC in humans ranges from 1 to
tine and carnitine supplementation have resulted in bene- 15 lmol/kg body weight/day, whereas the rate of biosyn-
ficial effects in the treatment of various neurological thesis is about 1–2 lmol/kg body weight/day [189]. In
diseases, in spite of the fact that fat are not the major fuel omnivorous humans, animal products provide an estimated
for brain [181]. The acylcarnitine profile has been shown to 75% of whole body carnitine reserves [190]. Conversely,
be useful in identifying inborn errors of metabolism and to vegetarians meet most of their carnitine needs via endog-
be altered under different metabolic conditions [182]. enous production [190]. Importantly, typical vegetarian
Moreover, recent data suggest multifactorial roles for a- diets, high in fruits and vegetables and low in fat and
cylcarnitines in neuroprotection. Brain acylcarnitines can calories, naturally lessens carnitine requirements. By con-
function in synthesizing lipids, altering and stabilizing trast, Western diets comprised highly processed foods
membrane composition, modulating genes and proteins, which are low in carnitine content and rich in fat and sugar.
improving mitochondrial function, increasing antioxidant Consequently, heavy consumption of these foods can pro-
activity, and enhancing cholinergic neurotransmission voke an imbalance between carnitine supply and demand
[183]. The importance of carnitine in brain is emphasized [97, 191]. Carnitine endogenous biosynthesis is limited by
by carnitine deficiency symptoms, many of which involve availability of trimethyllysine, a byproduct of lysosomal
major deleterious effects in brain. Because the brain is protein degradation. Carnitine reserves can be compro-
highly reliant on oxidative metabolism, impairment of fatty mised by depletion of essential micronutrients and/or
acid metabolism and energy production due to lack of metabolites, such as iron, vitamin C, and a-ketoglutarate,
carnitine leads to metabolic encephalopathy [183]. Struc- all of which are required cofactors for carnitine biosyn-
turally, the astrocyte swells and mitochondria are expanded thetic enzymes [192].
in nerve cells under carnitine deprivation [183]. Fatty acids In the plasma, neither LC nor ALC are bound to proteins
become key energy substrates for brain under metaboli- [182]. Concentrations of carnitine and acylcarnitines
cally compromised conditions such as fasting or starvation. change under altered dietary conditions. During starvation
Therefore functions of carnitine and acylcarnitines in fatty and after eating a high-fat diet, the proportion of carnitine
acid metabolism, ketosis and buffering of the concentration that is acetylated in liver and kidney significantly increases
ratio of acyl-CoA to free CoA, are significant in brain associated with an accumulation of several short, medium
metabolism, particularly metabolic disturbances present in and long chain acylcarnitine (LCAC) in skeletal muscle
neurological disease [97]. Currently, only a relatively small and a lowering of acylcarnitine levels in the liver and
subset of acylcarnitines is usually investigated and more kidney. Oppositely, a high carbohydrate diet causes very
research is needed to exploit the use of acylcarnitines in the low levels of ALC in liver. Plasma levels do not directly
treatment of neurological diseases using a list of acylcar- correlate with cerebral levels, however starvation causes a
nitines encompassing a wide range of these molecules. significant increase in mean brain acylcarnitine concen-
L-carnitine (LC) (trimethylamino-b-hydroxybutyrate) tration, with almost all of the increase beeing short-chain
(Fig. 5) is a quaternary amine, naturally present in mam- acylcarnitines [97]. Carnitine and its relative esters may be
malian cells and tissues as both free carnitine and acyl- redistributed to the brain during fasting and the brain may
carnitines, including acetyl-L-carnitine (ALC). Only LC is use them for energy production or, possibly, for the
synthesized in the tissue and have biological activity. Its delivery of acetyl groups [193–196]. Plasma concentration

123
Neurochem Res (2010) 35:1880–1915 1905

Table 2 Changes in carnitine and acylcarnitine concentrations under uptake were similar to those of carnitine transport [97].
dietary manipulations and alterations in metabolism Exogenously administered acetylcarnitine is transported
Model Diet Carnitine Acylcarnitine into mitochondria where the acetyl group favzors the pro-
duction of acetyl-CoA both directly and by activation of
Human Starvation/fasting ; :
carnitine acetyltransferase (CAT), which is decreased in
High-fat/fat load ; : aging. The acetyl-CoA acts on the acetylation status of
Diabetic ketosis ; : mitochondrial proteins that increase mitochondrial tran-
scription and protein synthesis. As a result, cytochrome b
content increases, leading to increased activity of electron
and urinary excretion of acetylcarnitine vary with physio- transport chain (ETC.) complexes, and stimulating the
logical and pathological states, reflecting mainly the degree oxidative phosphorylation (OXPHOS). This sequence of
of acetyl-CoA synthesis. In subjects under fasting condi- events leads to the restoration of the aging-related mito-
tions, acetylcarnitine excretion in the urine increases to chondrial defect [186].
78% of the excreted acylcarnitines [97]. In contrast, in Biological effects of acetylcarnitine relate to (1) the
obese human subjects under fasting conditions, the increase acetyl unit as acetyl-CoA, (2) the carnitine derived from
in urinary excretion of acetylcarnitine is markedly slower the compound, or (3) the intact acetylcarnitine molecule
and at a lower level, suggesting a slower formation of [97]. Administered acetylcarnitine is transported via the
acetyl-CoA derived from fat oxidation (Table 2). In dia- translocase of the inner membrane to the mitochondrial
betic patients with ketosis, acetylcarnitine represented 61% matrix where CAT generates acetyl-CoA leaving carnitine.
of the total urine acylcarnitines, and decreased dramatically CAT uses substrate in one direction of the reaction that is
upon insulin treatment. The equilibrium between acetyl- competitive inhibitor respect to the reaction products
CoA (plus carnitine) and CoA (plus acetylcarnitine) obtained in that direction [195]. Therefore, considering the
(acetyl-CoA/CoA ratio) is crucial for mitochondrial Km of CAT be 350 lM, if the intramitochondrial con-
metabolism. The mitochondrial content of endogenous centration of acetylcarnitine is higher than this value likely
acetylcarnitine is an indicator of mitochondrial metabolism exogenous acetylcarnitine facilitates the continuous gen-
of acetyl-CoA (Fig. 6). Acetyl-CoA, derived from pyru- eration of acetyl-CoA. This, in turn results in an increase of
vate, amino acids, and fatty acids, is reversibly converted acetyl-CoA/CoA ratio and in a consequent decrease of the
to acetylcarnitine and CoA in the presence of carnitine by activity of pyruvate dehydrogenase associated with inhi-
the carnitine acetyltransferase (CAT), a mitochondrial bition of mitochondrial fatty acid b-oxidation and the TCA
matrix enzyme attached to the inner membrane. This pro- cycle. Thus increases in mitochondrial acetyl-CoA may
cess regenerates free CoASH, which allows fatty acid impact negatively on mitochondrial metabolism. However
oxidation and the tricarboxylic acid (TCA) cycle to pro- consumption of acetyl-CoA in irreversible metabolic
ceed. Acetylcarnitine is transported to the cytosol through pathways, i.e., lipoic acid, and citrate synthesis within
the mitochondrial inner membrane in exchange with car- mitochondria paralleled by corresponding stimulation of
nitine by the antiport carnitine acylcarnitine translocase biosynthetic processes within the cytosol, drives the pro-
(CACT), thus providing acetyl groups for the synthesis of cess maintaining the mitochondrial content of acetyl-CoA
sterols, fatty acids, and ketone bodies [182]. Esters of the within a safe range [97]. Acetylcarnitine provides acetyl-
fatty acid of CoA are almost exclusively formed in the CoA for the synthesis of acetylcholine in the brain and fatty
cytosol and they are not transported trough the membranes. acids in lipogenic tissues that contain fatty acid synthase,
Consequently, under normal conditions, carnitine palmi- i.e., brain, liver, and adipose tissue. Citrate generated by
toyltransferase (CPT) 1 and 2, localized, respectively on the condensation of acetyl-CoA and oxaloacetate in the
the extern and inner surface of inner mitochondria mem- TCA cycle is transported into the cytosol where it is
brane, catalyzes the transfer of acyl groups from acyl-Co- cleaved by citrate lyase to re-generate cytosolic acetyl-
enzymeA to carnitine to produce acylcarnitines and free CoA. One-third of the acetyl-CoA that reaches the cytosol
coenzyme A in the cytosol (Fig. 6). Acylcarnitine, trough a via citrate in the brain is used for acetylcholine synthesis
specific carrier protein, then moves in the mitochondria, [97]. Intravenous administration of acetylcarnitine leads
where are reconstituted and metabolized as esters of ace- the acetyl moiety enter the the TCA cycle, as investigated
tilCoA, while the carnitine is transported back in the by 13C NMR spectroscopy, with 13C found in the gluta-
cytosol. Ohashi et al. found that acetylcarnitine is trans- mate, glutamine, and glutathione molecules [97]. Further-
ported by the organic cation transporter 2 (OCTN2), a more, carnitine stimulates oxidative metabolism of
homolog of the OCTN1, with high affinity (Km 8.50 lM) pyruvate and branched-chain amino acids, and is involved
in a saturable manner [188]. The pH profile and Hill in a host of other cellular functions [182]. An increased
coefficient (0.989) of the OCTN2-mediated acetylcarnitine acetyl-CoA/CoA ratio decreases the activity of pyruvate

123
1906 Neurochem Res (2010) 35:1880–1915

Fig. 6 The carnitine system and its interplay with mitochondrial exceeds consumption, these molecules can be converted, through
energetics and metabolism. Carnitine system modulates the metabolic CAT back to acylcarnitine derivatives and exported from the
fate of acetyl-CoA amd hence mitochondrial energetics. Long chain mitochondria via CrTrl. Supplemented acetylcarnitine is transported
acyls (LC), traverse the outer mitochondrial membrane via CPT1, into the mitochondria via CarTrl. Acetyl-CoA formed through the
before being internalized through the inner mitochondrial membrane mass-action of CAT, or synthesized de novo from acetate, becomes
(IMM) by carnitine translocase (CrTrl) and processed by CPT2, to available for tricarboxylic acid cycle (TCA), lipoic acid synthesis,
AcylCoA in the matrix side of the IMM. B-oxidation process mitochondrial protein acetylation and energy-dependent processes
degrades LC-AcylCoA to one two-carbon fragments of AcetylCoA in modulation
each successive cycle. In conditions where AcylCoA generation

dehydrogenase and inhibits mitochondrial fatty acid residues, and in addition to this, site-specific acetylation
b-oxidation and the TCA cycle. Nuclear acetyl CoA is of non-histone proteins plays an important role in tran-
either directly imported from the cytosol or synthesized scriptional regulation. In particular, highmobility group
from acetate by acetyl-CoA synthetase or via the potential (HMG)-box proteins are acetylated, and TFAMcontain
nuclear CAT using acetylcarnitine. The free passage of twoHMGbox-like domains [97]. Acetylated histone tails
acetyl-CoA through the nuclear pore complex facilitates are associated with active chromatin, whereas histone
the traffic of cytosolic acetyl-CoA to the nucleus [97]. deacetylation is associated with transcriptional repression
Consistent with this notion protein lysine acetylation is of genes, because the removal of acetyl groups from lysine
emerging as a key posttranslational modification in cellular residues limits accessibility of the DNA for transcription
regulation, particularly through the modification of his- [97]. Therefore, histone acetyltransferases and deacetylases
tones and nuclear transcription regulators [196]. It is are transcriptional co-regulators. All known acetyltrans-
already established that transcription of specific nuclear ferases use acetyl-CoA as a donor for acetylation. Although
genes controls the replication and transcription of the the existence of distinct mitochondrial and nucleo-cyto-
mitochondrial genome. Nuclear respiratory factor-1 (NRF-1) solic acetyl-CoA pools has been not univocally demon-
and mitochondrial transcription factor A (TFAM) are strated, nuclear concentration of acetyl-CoA seems to be
required for mitochondrial DNA (mtDNA) replication; the limiting factor for histone acetylation [198]. This
together with mitochondrial transcription factors B (TFB1 complex level of regulation of mitochondrial biology is
and TFB2), they stimulate the transcription of both light integrated to the intrinsic function of SIRTs (sirtuins,
and heavy chains of mtDNA [197]. Furthermore, tran- SIRT3, 4, and 5) family enzymes, which are nicotin-
scription of nuclear DNA recently has been linked to amide adenine nucleotide (NAD?)-dependent deacetylase
acetylation and deacetylation of core histone tails at lysine silent information regulators [2]. Accordingly, proteomic

123
Neurochem Res (2010) 35:1880–1915 1907

investigations on protein acetylation have identified 388 Virtually every enzyme in glycolysis, gluconeogenesis, the
acetylation sites on 195 proteins in mitochondria from tricarboxylic acid (TCA) cycle, the urea cycle, fatty acid
HeLa cells and mouse liver, representing 20% of mito- metabolism, and glycogen metabolism was found to be
chondrial proteins such as proteins involved in the TCA acetylated in human liver tissue [196]. The importance of
cycle, fatty acid b-oxidation, amino acid and carbohydrate lysine acetylation in the regulation of chromatin dynamics
metabolism, membrane transport, and ETC. [199]. Along and gene expression is well recognized as an emerging
with this line of evidence, long-chain acyl-CoA synthetase areas of interest. Recent finding extends the scope of cell
1 was found to be acetylated at the lysine residue 633 in rat regulation by lysine acetylation to an extent comparable
liver mitochondria [199]. Lysine acetylation neutralizes the to that of other major posttranslational modifications such
positive charge and increases the hydrophobicity of the as phosphorylation and ubiquitination. Most intermediate
lysine side chain. While the functional relevance of this metabolic enzymes in fact are acetylated and it has been
postranslational modification is not completely defined, it demonstrated that acetylation can directly affect the
is conceivable that these modifications can modify protein enzyme activity or stability. Acetylation of metabolic
conformation and, consequently, function. This is the case enzymes was found to change in response to the alterations
for mitochondrial matrix acetyl-CoA synthetase, which is of extracellular nutrient availability, providing evidence for
reversibly acetylated at a lysine residue in the active site of a physiological role of dynamic acetylation in metabolic
the enzyme; thus, SIRT3-induced deacetylation activates regulation. Lysine acetylation is an evolutionarily con-
the enzyme. Another mitochondrial target for both SIRT3 served mechanism involved in regulation of metabolism in
and SIRT4, is glutamate dehydrogenase which is inhibited response to nutrient availability and cellular metabolic
upon deacetylation. Remarkably, SIRT3 by reversibly status. Acetylation may play a key role in the coordination
binding to mitochondrial complex I and by decreasing the of different metabolic pathways in response to extracellular
acetylation status, leads to an increase of its activity [97]. conditions [196]. The carnitine system has a key role in in
As a result, cytochrome b content increases, leading to the deacylation–reacylation processes of membrane phos-
increased activity of electron transport chain (ETC.) com- pholipids. Accordingly evidence exist which indicates that
plexes, associated with stimulation of oxidative phos- carnitine and acetylcarnitine increase fluidity in rat brain
phorylation (OXPHOS). Thus, reversible acetylation of the microsomes and liposomes [183]. Due to their amphiphilic
mitochondrial proteome, which represents an efficient structure, these molecules may directly interact with the
mechanism to regulate the activity of mitochondrial pro- surface charges on cell membranes. The carboxylic group,
teins, may be relevant to aging process, where this in particular can interact with charges on membrane
sequence of events, conceivably, can revert the mitochon- phospholipids, glycolipids, and proteins. Recent evidence
drial dysfunction associated to age-related pathology [97]. demonstrate that acetylcarnitine is a critical factor for the
In addition to regulation of mitochondrial enzyme activi- elongation–desaturation of the n-3 polyunsaturated fatty
ties, acetylation possibly modulates de novo synthesis of acids to form 22:6n-3, DHA, in mitochondria [200]. ALC
mitochondrial acetyl-CoA. Since acetyl-CoA is the acety- is thought to provide an intra-mitochondrial source of
lation donor for all known acetyltransferases and the Km acetyl groups and donate them in the elongation pathway.
for acetyl-CoA is high (330 lM), reasonably, in conditions Changes in DHA content of membranes can influence
of carnitine deficiency the concentration of mitochondrial synaptic plasticity, inflammatory response, gene expres-
acetyl-CoA could be a limiting factor in the acetylation sion, ion channels, membrane-bound proteins and neuro-
reactions, such as during aging. In fact, carnitine acetyl- transmission. Likewise, in mitochondria phospholipids
transferase is decreased in aging and, in these conditions, may be important for assembly of the respiratory chain
exogenous acetylcarnitine transported into the mitochon- complexes. Consequently, acetylcarnitine prevents damage
dria by generating the necessary acetyl group for building associated with inhibition of these complexes, whereby
up of acetyl-CoA, modulates the acetylation status of preserving mitochondrial membrane integrity or improving
mitochondrial proteins. Likewise, following acetylcarnitine overall metabolic functions through promoting acetylation
supplementation it is plausible that a nuclear CAT might processes of critical protein and lipid targets.
contribute to nuclear acetyl-CoA. Consistent to this, global Recent evidence suggests that L-carnitine and acetyl-
transcription in yeast, has been recently evidenced to be carnitine supplementation is important to attenuate oxida-
regulated by the steady-state level of acetyl-CoA supplied tive stress, and mitochondrial dysfunction associayed to
by the nuclear acetyl-CoA synthetase [198], which is under various physiopathological conditions such as aging,
the control of the histone acetylase activity rather than the stress-related disease states of brain, and in metabolic
histone deacetylase activity. Notably, recent evidence diseases where one common denominator is a reduction
indicates that lysine acetylation is a prevalent modification in tissue lactic acidosis and consequent formation of
in enzymes that catalyze intermediate metabolism [196]. ROS. One major mechanism of action of ALC is the

123
1908 Neurochem Res (2010) 35:1880–1915

improvement of mitochondrial bioenergetics which allows normal GSH content and increase in the amount of cyto-
the neuron to produce ATP necessary to maintain the chrome b and aa3 hemes mitochondria isolated from old
normal membrane potential and restore altered redox bal- rats [97, 208], enhancing mtDNA transcription, the stabil-
ance (i.e., synthesis of glutathione) [41, 201], through a ity of mitochondrial mRNA, and mitochondrial protein
shift in both the mitochondrial and cytosolic redox state synthesis. Further to this, in rodents ALC up-regulates the
and/or through the induction of antioxidant genes [35, 142, expression of metabotropic glutamate receptor 2 (mGlu2)
202]. As a consequence, intracellular reducing power in dorsal root ganglia (DRG) of the spinal cord with a
increases being available for detoxification through the mechanism demonstrated to be dependent on activation of
glutathione system. Thus, due to their capacity in increas- the NF-kappaB pathway through acetylation of p65/RelA.
ing mitochondrial energetics and function, both carnitine On the basis of this action ALC is suggested for clinical
and acetylcarnitine molecules are critical for the prevention management of neuropathic pain [209–211].
of neurodegeneration and the regulation of neurotrans- Due to its central role in fatty acid utilization, the car-
mission [97, 182, 183]. nitine system is a determinant in insulin regulation of fat
Acetylcarnitine supplementation can be of therapeutic and glucose metabolic rate. Insulin resistance has been
benefit for chronic degenerative diseases, Alzheimer’s described as a ‘marker of early mitochondrial carnitine-
disease (AD) slowing the progression of mental deterio- dependent dysfunction’ [212]. For example, increased lipid
ration in this disease, multiple sclerosis, chronic fatigue accumulation in muscle leads to prolonged inhibition of
syndrome, depression in the elderly, HIV infection, dia- muscle isoform of carnitine palmitoyltransferase I enzyme
betic neuropathies, it provides to protection during meta- (M-CPT I). This insulin-regulated enzyme facilitates bidi-
bolic stress such as hypoxia, ischemia and reperfusion of rectional glucose and fatty-acid carbon flux, which conse-
the brain and of the heart, cognitive impairment resulting quently promotes insulin resistance [212]. Abnormalities in
from alcoholism, and ageing, brain injury, autism, Par- M-CPT I expression have also been reported in metabolic
kinson disease, Down syndrome, Huntington’s disease, disorders (e.g., metabolic syndrome, diabetes mellitus
cerebellar ataxia, age-associated mental decline, hepatic [186, 213]. In healthy subjects, L-carnitine infusion in the
encephalopathy and ammonia neurotoxicity [183]. By presence of hyperinsulinemia increases OCTN2 mRNA
restoring tissue carnitine in the elderly, acetylcarnitine expression and total skeletal muscle carnitine content
supplementation facilitates the elimination of potentially [214]. In streptozotocin (STZ)-induced diabetic rats, car-
toxic acyl-CoA metabolites derived from fatty acid oxi- nitines show to modulate insulin-like growth factor axis
dation. Besides the afore mentioned role on mitochondrial [215]. Importantly, tissue export of acetylcarnitine lower
bioenergetics, which allows brain cells to restore altered local concentration of acetylCoA, which in turn, stimulates
redox balance [3, 45], ALC is neuroprotective through a PDH activity and this basic mechanism contributes to
variety of other effects such as the increase in PKC activity enhance glucose oxidation [186]. Consistently, carnitine
[41], and modulation of synaptic plasticity via NMDA administration to obese rats not only restores tissue carni-
receptor expression and increase in the production of tine content but also enhances glucose disposal and mito-
neurotrophins [202]. Importantly, treatment with ALC chondrial performance [186]. This highlights the crucial
induces up-regulation of the gene coding for Hsp72 role of carnitine system in regulating mitochondrial
explaining its protective effects in inflammation, neurode- emergetics, in particular CAT enzyme whose overexpres-
generative disorders, and aging [203–205]. Another gene sion has been proven to promote glucose uptake and mit-
identified that is positively modulated by acetylcarnitine in igate lipid-induced suppression of glucose oxidation,
the brain is the mitochondrial voltage-dependent anion counteracting the Randle effects ‘‘in vivo’’ (i.e., lipid-
channel (VDAC) protein [206]. VDAC, also known as induced lowering of glucose oxidation) [216].
porin, is a small pore-forming protein of the mitochondrial Carnitine, exogenously administered, attenuates organic
outer membrane. It is the primary pathway for diffusion of aciduria induced by alcohol given chronically to rats as
ions and metabolites across the outer membrane and plays well s prevents neuronal injury in the hippocampus by
a key role in apoptosis by forming the outer pore compo- inhibiting the increase in oxidized glutathione and mito-
nent of the mitochondrial permeability transition (MPT) chondrial dysfunction [184, 185]. Studies in our laboratory
complex. Defects in the function of complexes II and III have shown that ALC reduces Ab toxicity in primary
have been observed in Huntington’s disease patients [207], cortical neuronal cultures by increasing both HO-1 and
while complex IV is involved in the mitochondrial dys- Hsp70 expression [217]. In rats chronic ALC treatment
function underlying the pathogenesis of AD. Acetylcarni- increases life-span, improves cognitive behaviour and
tine has been shown to reverse age-related decreases in the long-term memory performance [202]. Furthermore,
activity of complex III and oxidative phosphorylation and chronic ALC treatment has been proven to prevent age-
the inhibition of complex IV, resulting in restoration of related changes in mitochondrial respiration and decrease

123
Neurochem Res (2010) 35:1880–1915 1909

oxidative stress biomarkers through the up-regulation of subsequent alternative arrangements of genomic reorgani-
vitagene products [2, 3, 35, 41, 201, 204]. Consistently, we zation and biochemical traduction. An ultimative arrange-
have demonstrated that acetyl-L-carnitine induces HO-1 in ment was then reached in which mitochondria became
a dose and time dependent manner, an effect associated specialized in energy production and the nucleus–cytosol
with up-regulation of other Hsps and high expression of the became specialized in structure. This final design prompted
redox-sensitive transcription factor Nrf2, a mechanism the development of multicellularity and the evolution of
following the hormetic law-dose response [3, 201]. These higher plants and animals, including humans [220]. In this
evidence, intriguingly related with modulation of mito- reorganization, the transfer of virtually all of the genes of
chondrial biogenesis via the transcriptional control of the the mitochondrial genome into the chromosomal nDNA
nuclear respiratory factor-1 (NRF-1) [97, 99], highlight the was finalized. In spite of this, mtDNA persisted, today still
crucial role of the carnitine system and the clinical rele- retaining 13 polypeptide-encoding genes plus a small and
vance of its insufficiency in mitochondrial energetics and large rRNA gene and 22 tRNA genes. All of the mtD-
metabolic disruptions associated with aging, overnutrition NAencoded polypeptides are core subunits of the enzyme
and neurodegenerative disorders. complexes of the mitochondrial energy-generating appa-
ratus, oxidative phosphorylation (OXPHOS). The 13
polypeptides of the mtDNA include 7 of the 45 polypep-
Conclusions and Future Directions tides of complex I (ND1, -2, -3, -4L, -4, -5, -6), 1 of the
11 polypeptides of complex III (cytochrome b), 3 of the 13
While the anatomical paradigm of medicine and the polypeptides of complex IV (COI, -II, -III), and 2 of the 15
Mendelian paradigm of genetics have been powerful pre- polypeptides of complex V (ATP6 and -8). All of the other
dictors of medical relationships for the past century, they genes of the mitochondrial genome are dispersed across the
are failing to direct us toward solutions for the common chromosomes and include the mitochondrial DNA poly-
age-related diseases. Continually increasing resources are merase g (POLG), RNA polymerase, ribosomal proteins or
being expended to combat the age-related diseases that metabolic enzymes. Why it was beneficial for the first
include diabetes and metabolic syndrome, Alzheimer’s 1,500 mitochondrial genes to be transferred to the nucleus,
disease, Parkinson’s disease, cardiovascular disease, and and not for the last 13, still remains an intriguing unraveled
cancer. Yet the causes of these diseases remain an open question [67, 220, 221].
question, while their incidence and morbidity either remain Mitochondria are major cellular sources (and targets) of
constant or increase [218]. Huge investments in biomedical free radicals, play key roles in the regulation of calcium
research in the recent past have resulted in some important homeostasis, and are effectors of the intrinsic apoptotic
accomplishments, such as sequencing of the human gen- pathway due to the release of signaling molecules that
ome, identification of thousands of human chromosomal activate and trigger specific caspase cascades. Mitochon-
single nucleotide polymorphisms (SNPs), or regional drial dysfunction is inherent in a variety of human disor-
clusters of chromosomal SNPs. However, these accom- ders from the classical mitochondrial diseases arising from
plishments have failed to reveal the genetic causes for the mitochondrial DNA mutations (encephalomyopathies) to
common age-related diseases [67]. When a current para- those involving mitochondrial signaling pathways to the
digm fails to make productive predictions, then hypothesis- rest of the cell, modulated by organellar dynamics and
based research begins to fail. To resolve the crisis and culminating in programmed cell death. The role of mito-
return to productive ‘‘normal science,’’ a new paradigm chondria in normal aging has been the focus of extensive
must be generated that comprise the potential of the pre- research in the last decades and being complementary to
vious founding paradigm whereby adding new elements and maturing from the free radical theory of aging, to the
that address the current problems being compared and oxidative stress theory of aging, to the mitochondrial oxi-
possibly overcome [219]. dative stress theory of aging, and today addressing the tight
Life involves the dynamic interplay between structure co-regulation of mitochondrial energy and redox signaling.
and energy. For the eukaryotic cell, this duality was In this scenario, it appears conceivably the function of the
solidified approximatelyt 2 billion years ago by the sym- carnitine system as a prototypical vitagene operating at the
biosis of what appears to have been a glycolytic motile cell, functional interface of energy distribution between ances-
which gave rise to the nucleus–cytosol, and an oxidative a- tral mechanisms of cell proliferation and differentiation
proteobacterium, which evolved into the mitochondrion and homeostatic mitochondrial dependent processes of cell
[67]. Initially, each organism was free living and contained survival which require energy for cellular stress response
all of the genes for an independent life form. However, and redox homeostasis [41]. Very importantly, this new
over the subsequent 1.2 billion years, the single-cell envisioned role exploited for the carnitine system, partic-
descendants of the initial symbiosis went through ularly, acetylcarnitine as a molecule endowed with the

123
1910 Neurochem Res (2010) 35:1880–1915

capability of potentiating the cellular stress response 15. Calabrese V, Butterfield DA, Scapagnini G, Stella AM, Maines
pathways appear to be a promising alternative therapeutic MD (2006) Redox regulation of heat shock protein expression
by signaling involving nitric oxide and carbon monoxide: rele-
approach for those pathophysiological conditions, such as vance to brain aging, neurodegenerative disorders, and longev-
neurodegeneration or cancer, where hormetic stimulation ity. Antioxid Redox Signal 8:444–477
of the vitagene pathway is strongly warranted. 16. Calabrese V, Guagliano E, Sapienza M, Panebianco M, Calafato
S, Puleo E, Pennisi G, Mancuso C, Butterfield AD, Giuffrida
Acknowledgments Work from the authors’ laboratories was sup- Stella AM (2007) Redox regulation of cellular stress response in
ported by grants from MIUR, FIRB RBRN07BMCT, I.N.B.B., and by aging and neurodegenerative disorders: role of vitagenes. Neu-
‘‘Fondi Ateneo’’ 2008 and 2009. We are very much honoured to rochem Res 32:757–773
contribute to this Special Issue in honour of Abel Lajtha. Everybody 17. Sohal RS, Toroser D, Brégère C, Mockett RJ, Orr WC (2008)
in the field of Neurochemistry knows Abel Lajtha and his great Age-related decrease in expression of mitochondrial DNA
contribution to the advancement of research in Neurochemistry. He encoded subunits of cytochrome c oxidase in Drosophila mel-
has been the Editor in Chief of the Journal ‘‘Neurochemical anogaster. Mech Ageing Dev 129:558–561
Research’’ for so many years holding very stimulating and interesting 18. Tudek B, Winczura A, Janik J, Siomek A, Foksinski M, Oliński
Meetings of the Editorial Board to which we are very proud to have R (2010) Involvement of oxidatively damaged DNA and repair
participated. It is difficult to imagine future Editorial Board Meetings in cancer development and aging. Am J Transl Res 2:254–284
without him. We would like to thank him very much for his great 19. Rai P (2010) Oxidation in the nucleotide pool, the DNA Damage
contribution to the advancement and success of Neurochemical response and cellular senescence: defective bricks build a
Research and ask him to continue helping us with its precious efforts defective house. Mutat Res. [Epub ahead of print]
and advices. 20. Mangialasche F, Polidori MC, Monastero R, Ercolani S, Cam-
arda C, Cecchetti R, Mecocci P (2009) Biomarkers of oxidative
and nitrosative damage in Alzheimer’s disease and mild cog-
nitive impairment. Ageing Res Rev 8:285–305
References 21. Lezza AM, Mecocci P, Cormio A, Beal MF, Cherubini A,
Cantatore P, Senin U, Gadaleta MN (1999) Mitochondrial DNA
1. Balch WE, Morimoto RI, Dillin A, Kelly JW (2008) Adapting 4977 bp deletion and OH8dG levels correlate in the brain of
proteostasis for disease intervention. Science 319:916–919 aged subjects but not Alzheimer’s disease patients. FASEB J
2. Calabrese V, Cornelius C, Dinkova-Kostova AT, Calabrese EJ, 13:1083–1088
Mattson MP (2010) Cellular stress responses, the hormesis 22. Wilson DM 3rd, Bohr VA, McKinnon PJ (2008) DNA damage,
paradigm and vitagenes: novel targets for therapeutic interven- DNA repair, ageing and age-related disease. Mech Ageing Dev
tion in neurodegenerative disorders. Antioxid Redox Signal 129:349–352
610:285–308 23. Passos JF, Saretzki G, von Zglinicki T (2007) DNA damage in
3. Calabrese V, Cornelius C, Dinkova-Kostova AT, Calabrese EJ telomeres and mitochondria during cellular senescence: is there
(2009) Vitagenes, cellular stress response and acetylcarnitine: a connection? Nucleic Acids Res 35:7505–7513
relevance to hormesis. Biofactors 35:146–160 24. Niedernhofer LJ, Garinis GA, Raams A, Lalai AS, Robinson
4. Gerschman R, Gilbert DL, Nye SW, Dwyer P, Fenn WO (1954) AR, Appeldoorn E, Odijk H, Oostendorp R, Ahmad A, van
Oxygen poisoning and x-irradiation: a mechanism in common. Leeuwen W, Theil AF, Vermeulen W, van der Horst GT,
Science 119:623–626 Meinecke P, Kleijer WJ, Vijg J, Jaspers NG, Hoeijmakers JH
5. Gerschman R, Gilbert DL, Nye SW, Fenn WO (1954) Influence (2006) A new progeroid syndrome reveals that genotoxic stress
of x-irradiation on oxygen poisoning in mice. Proc Soc Exp Biol suppresses the somatotroph axis. Nature 444:1038–1043
Med 86:27–29 25. Sohal RS, Orr WC (1992) Relationship between antioxidants,
6. Harman D (1956) Aging: a theory based on free radical and prooxidants, and the aging process. Ann N Y Acad Sci 663:
radiation chemistry. J Gerontol 11:298–300 74–84
7. Harman D (1972) The biologic clock: the mitochondria? J Am 26. Stadtman ER (2006) Protein oxidation and aging. Free Radic
Geriatr Soc 20:145–147 Res 40:1250–1258
8. Speakman JR (2005) Body size, energy metabolism and life- 27. Wang Q, Zhao X, He S, Liu Y, An M, Ji J (2010) Differential
span. J Exp Biol 208:1717–1730 proteomics analysis of specific carbonylated proteins in the
9. Gilbert DL, Colton CA (1999) Reactive oxygen species in temporal cortex of aged rats: the deterioration of antioxidant
biological systems: an interdisciplinary approach. Kluwer, New system. Neurochem Res 35:13–21
York 28. Chen JJ, Lin F, Qin ZH (2008) The roles of the proteasome
10. Bokov A, Chaudhuri A, Richardson A (2004) The role of oxi- pathway in signal transduction and neurodegenerative diseases.
dative damage and stress in aging. Mech Ageing Dev 125: Neurosci Bull 24:183–194
811–826 29. Grimm S, Hoehn A, Davies KJ, Grune T (2010) Protein oxi-
11. Sohal R, Weindruch R (1996) Oxidative stress, caloric restric- dative modifications in the ageing brain: Consequence for the
tion, and aging. Science 273:59–63 onset of neurodegenerative disease. Free Radic Res. [Epub
12. Muller FL, Lustgarten MS, Jang Y, Richardson A, Van Remmen ahead of print]
H (2007) Trends in oxidative aging theories. Free Radic Biol 30. Lodi R, Tonon C, Calabrese V, Schapira AH (2006) Friedreich’s
Med 43:477–503 ataxia: from disease mechanisms to therapeutic interventions.
13. Salmon AB, Richardson A, Pérez VI (2010) Update on the Antioxid Redox Signal 8:438–443
oxidative stress theory of aging: does oxidative stress play a role 31. Calabrese V, Cornelius C, Mancuso C, Pennisi G, Calafato S,
in aging or healthy aging? Free Radic Biol Med 48:642–655 Bellia F, Bates TE, Giuffrida Stella AM, Schapira T, Dinkova
14. Calabrese V, Giuffrida Stella AM, Butterfield DA, Scapagnini G Kostova AT, Rizzarelli E (2008) Cellular stress response: a
(2004) Redox regulation in neurodegeneration and longevity: novel target for chemoprevention and nutritional neuroprotec-
role of the heme oxygenase and HSP70 systems in brain stress tion in aging, neurodegenerative disorders and longevity. Neu-
tolerance. Antioxid Redox Signal 6:895–913 rochem Res 33:2444–2471

123
Neurochem Res (2010) 35:1880–1915 1911

32. Calabrese V (2007) Highlight commentary on ‘‘redox proteo- 48. Van Raamsdonk JM, Meng Y, Camp D, Yang W, Jia X, Bénard
mics analysis of oxidatively 3 modified proteins in G93A–SOD1 C, Hekimi S (2010) Decreased energy metabolism extends life
transgenic mice—a model of 4 familial amyotrophic lateral span in Caenorhabditis elegans without reducing oxidative
sclerosis’’. Free Radic Biol Med 43:160–162 damage. Genetics 185:559–571
33. Halliwell B (2009) The wanderings of a free radical. Free Radic 49. Van Raamsdonk JM, Hekimi S (2010) Reactive oxygen species
Biol Med 46:531–542 and aging in caenorhabditis elegans: causal or casual relation-
34. Seet RC, Lee CY, Lim EC, Tan JJ, Quek AM, Chong WL, Looi ship? Antioxid Redox Signal
WF, Huang SH, Wang H, Chan YH, Halliwell B (2010) Oxi- 50. Calabrese V, Scapagnini G, Giuffrida Stella AM, Bates TE,
dative damage in Parkinson disease: measurement using accu- Clark JB (2001) Mitocondrial involvement in brain function and
rate biomarkers. Free Radic Biol Med 48:560–566 dysfunction: relevance to aging, neurodegenerative disordes and
35. Poon HF, Calabrese V, Calvani M, Butterfield DA (2006) Pro- longevity. Neurochem Res 26:739–764
teomics analyses of specific protein oxidation and protein 51. Druzhyna NM, Wilson GL, LeDoux SP (2008) Mitochondrial
expression in aged rat brain and its modulation by L-acetylcar- DNA repair in aging and disease. Mech Age Dev 129:383–390
nitine: insights into the mechanisms of action of this proposed 52. Hamm-Alvarez S, Cadenas E (2009) Mitochondrial medicine
therapeutic agent for CNS disorders associated with oxidative and mitochondrion-based therapeutics. Adv Drug Deliv Rev
stress. Antioxid Redox Signal 8:381–394 60:1437–1438
36. Bayot A, Gareil M, Rogowska-Wrzesinska A, Roepstorff P, 53. Hamm-Alvarez S, Cadenas E (2009) Mitochondrial medicine
Friguet B, Bulteau AL (2010) Identification of novel oxidized and therapeutics, Part II. Adv Drug Deliv Rev 61:1233
protein substrates and physiological partners of the mitochon- 54. Yap LP, Garcia JV, Han D, Cadenas E (2009) The energy-redox
drial ATP-dependent Lon-like protease Pim1. J Biol Chem axis in aging and age-related neurodegeneration. Adv Drug
285:11445–11457 Deliv Rev 61:1283–1298
37. Ugarte N, Petropoulos I, Friguet B (2010) Oxidized mitochon- 55. Schapira AH (2002) Primary and secondary defects of the
drial protein degradation and repair in aging and oxidative mitochondrial respiratory chain. J Inherit Metab Dis 25:207–214
stress. Antioxid Redox Signal 13:539–549 56. Tonon C, Lodi R (2008) Idebenone in Friedreich’s ataxia.
38. Sultana R, Perluigi M, Newman SF, Pierce WM, Cini C, Coccia Expert Opin Pharmacother 9:2327–2337
R, Butterfield DA (2010) Redox proteomic analysis of car- 57. Calabrese V, Scapagnini G, Colombrita C, Ravagna A, Pennisi
bonylated brain proteins in mild cognitive impairment and early G, Giuffrida Stella AM, Galli F, Butterfield DA (2003) Redox
Alzheimer’s disease. Antioxid Redox Signal 12:327–336 regulation of heat shock protein expression in aging and neu-
39. Calabrese V, Mancuso C, Ravagna A, Perluigi M, Cini C, De rodegenerative disorders associated with oxidative stress: a
Marco C, Butterfield DA, Giuffrida Stella AM (2007) In vivo nutritional approach. Amino Acids 25:437–444
induction of heat shock proteins in the substantia nigra follow- 58. Petrosillo G, Matera M, Casanova G, Ruggiero FM, Paradies G
ing L-DOPA administration is associated with increased activity (2008) Mitochondrial dysfunction in rat brain with aging
of mitochondrial complex I and nitrosative stress in rats: regu- Involvement of complex I, reactive oxygen species and cardio-
lation by glutathione redox state. J Neurochem 101:709–717 lipin. Neurochem Int 53:126–131
40. Bellia F, Calabrese V, Guarino F, Cavallaro M, Cornelius C, De 59. Ames BN, Liu J (2004) Delaying the mitochondrial decay of
Pinto V, Rizzarelli E (2009) Carnosinase levels in aging brain: aging with acetylcarnitine. Ann N Y Acad Sci 1033:108–116
redox state induction and cellular stress response. Antioxid 60. Hempenstall S, Picchio L, Mitchell SE, Speakman JR, Selman C
Redox Signal 11:2759–2775 (2010) The impact of acute caloric restriction on the metabolic
41. Calabrese V, Mancuso C, Calvani M, Rizzarelli E, Butterfield phenotype in male C57BL/6 and DBA/2 mice. Mech Ageing
DA, Giuffrida Stella AM (2007) Nitric oxide in the CNS: neu- Dev 131:111–118
roprotection versus neurotoxicity. Nature Neurosi 8:766–775 61. Mattson MP (2008) Dietary factors, hormesis and health. Age-
42. Calabrese V, Signorile A, Cornelius C, Mancuso C, Scapagnini ing Res Rev 7:43–48
G, Ventimiglia B, Ragusa N, Dinkova-Kostova A (2008) Prac- 62. Ristow M, Zarse K (2010) How increased oxidative stress
tical approaches to investigate redox regulation of heat shock promotes longevity and metabolic health: the concept of
protein expression and intracellular glutathione redox state. mitochondrial hormesis (mitohormesis). Exp Gerontol 45:
Methods Enzymol 441:83–110 410–418
43. Calabrese V, Colombrita C, Sultana R, Scapagnini G, Calvani 63. Benard G, Faustin B, Passerieux E, Galinier A, Rocher C,
M, Butterfield DA, Stella AM (2006) Redox modulation of heat Bellance N, Delage JP, Casteilla L, Letellier T, Rossignol R
shock protein expression by acetylcarnitine in aging brain: (2006) Physiological diversity of mitochondrial oxidative
relationship to antioxidant status and mitochondrial function. phosphorylation. Am J Physiol Cell Physiol 291:1172–1182
Antioxid Redox Signal 8:404–416 64. Luft R, Ikkos D, Palmieri G, Ernster L, Afzelius A (1962) A
44. Perluigi M, Di Domenico F, Butterfield DA, Giorgi A, Schininà case of severe hypermetabolism of nonthyroid origin with a
ME, Coccia R, Cini C, Bellia F, Cambria MT, Cormelius C, defect in the maintenance of mitochondrial respiratory control: a
Calabrese V (2010) Redox proteomics in aging rat brain: correlated clinical, biochemical and morphological study. J Clin
involvement of mitochondrial GSH status and mitochondrial Invest 41:1776–1804
protein oxidation in the aging process. J Neurosci Res (in press) 65. Schapira AH (2008) Mitochondrial dysfunction in neurodegen-
45. Bradley MA, Markesbery WR, Lovell MA (2010) Increased erative diseases. Neurochem Res 33:2502–2509
levels of 4-hydroxynonenal and acrolein in the brain in pre- 66. Wallace DC (1999) Mitochondrial diseases in man and mouse.
clinical Alzheimer disease. Free Radic Biol Med 48:1570–1576 Science 283:1482–1488
46. Sultana R, Butterfield DA (2010) Proteomics identification of 67. Wallace DC (2008) Mitochondria as Chi. Genetics 179:727–735
carbonylated and HNE-bound brain proteins in Alzheimer’s 68. Wang Y, Michikawa Y, Mallidis C, Bai Y, Woodhouse L,
disease. Methods Mol Biol 566:123–135 Yarasheski KE, Miller CA, Askanas V, Engel WK, Bhasin S,
47. Lovell MA, Xie C, Markesbery WR (1998) Decreased gluta- Attardi G (2001) Muscle-specific mutations accumulate with
thione transferase activity in brain and ventricular fluid in Alz- aging in critical human mtDNA control sites for replication.
heimer’s disease. Neurology 51:1562–1566 Proc Natl Acad Sci U S A 98:4022–4027

123
1912 Neurochem Res (2010) 35:1880–1915

69. Mazat J, Rossignol R, Malgat M, Rocher C, Faustin B, Letellier Lakatta EG, Sheu SS, Ouyang K, Chen J, Dirksen RT, Cheng H
T (2001) What do mitochondrial diseases teach us about normal (2008) Superoxide flashes in single mitochondria. Cell 134:
mitochondrial functions that we already knew: threshold 279–290
expression of mitochondrial defects. Biochim Biophys Acta 88. McCord JM (2002) Superoxide dismutase in aging and disease:
1504:20–30 an overview. Methods Enzymol 349:331–341
70. Letellier T, Malgat M, Rossignol R, Mazat JP (1998) Metabolic 89. Cho DH, Nakamura T, Lipton SA (2010) Mitochondrial
control analysis and mitochondrial pathologies. Mol Cell Bio- dynamics in cell death and neurodegeneration. Cell Mol Life
chem 184:409–417 Sci. 2010 Jun 25. [Epub ahead of print]
71. Davey GP, Peuchen S, Clark JB (1998) Energy thresholds in 90. Nakamura T, Cieplak P, Cho DH, Godzik A, Lipton SA (2010)
brain mitochondria. J Biol Chem 273:12753–12757 S-Nitrosylation of Drp1 links excessive mitochondrial fission
72. Pathak RU, Davey GP (2008) Complex I and energy thresholds to neuronal injury in neurodegeneration. Mitochondrion 10:
in the brain. Biochim Biophys Acta 1777:777–782 573–578
73. Benard G, Bellance N, James D, Parrone P, Fernandez H, 91. Gu Z, Nakamura T, Lipton SA (2010) Redox reactions induced
Letellier T, Rossignol R (2007) Mitochondrial bioenergetics and by nitrosative stress mediate protein misfolding and mitochon-
structural network organization. J Cell Sci 120:838–848 drial dysfunction in neurodegenerative diseases. Mol Neurobiol
74. Rocher C, Taanman JW, Pierron D, Faustin B, Benard G, 41:55–72
Rossignol R, Malgat M, Pedespan L, Letellier T (2008) Influ- 92. Nakamura T, Lipton SA (2010) Redox regulation of mito-
ence of mitochondrial DNA level on cellular energy metabo- chondrial fission, protein misfolding, synaptic damage, and
lism: implications for mitochondrial diseases. J Bioenerg neuronal cell death: potential implications for Alzheimer’s and
Biomembr 40:59–67 Parkinson’s diseases. Apoptosis [Epub ahead of print]
75. Benard G, Faustin B, Galinier A, Rocher C, Bellance N, 93. Büeler H (2010) Mitochondrial dynamics, cell death and the
Smolkova K, Casteilla L, Rossignol R, Letellier T (2008) pathogenesis of Parkinson’s disease. Apoptosis [Epub ahead of
Functional dynamic compartmentalization of respiratory chain print]
intermediate substrates: implications for the control of energy 94. Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E
production and mitochondrial diseases. Int J Biochem Cell Biol (2008) Mitochondrial fragmentation in neurodegeneration. Nat
40:1543–1554 Rev Neurosci 9:505–518
76. Kordower JH, Chu Y, Hauser RA, Freeman TB, Olanow CW 95. Bonda DJ, Wang X, Perry G, Smith MA, Zhu X (2010) Mito-
(2008) Lewy body-like pathology in long-term embryonic nigral chondrial dynamics in Alzheimer’s disease: opportunities for
transplants in Parkinson’s disease. Nat Med 14:504–506 future treatment strategies. Drugs Aging 27:181–192
77. Kilbride SM, Telford JE, Tipton KF, Davey GP (2008) Partial 96. Gottlieb RA, Carreira RS (2010) Autophagy in health and dis-
inhibition of complex I activity increases Ca(2?)-independent ease. 5. Mitophagy as a way of life. Am J Physiol Cell Physiol
glutamate release rates from depolarized synaptosomes. J Neu- 299:C203–210
rochem 106:826–834 97. Rosca MG, Lemieux H, Hoppel CL (2009) Mitochondria in the
78. Kilbride SM, Telford JE, Davey GP (2008) Age-related changes elderly: is acetylcarnitine a rejuvenator? Adv Drug Deliv Rev
in H2O2 production and bioenergetics in rat brain synaptosomes. 61:1332–1342
Biochim Biophys Acta 1777:783–788 98. Piantadosi CA (2008) Carbon monoxide, reactive oxygen sig-
79. Keeney PM, Xie J, Capaldi RA, Bennett JP Jr (2006) Parkin- naling, and oxidative stress. Free Radic Biol Med 45:562–569
son’s disease brain mitochondrial complex I has oxidatively 99. Piantadosi CA, Carraway MS, Babiker A, Suliman HB (2008)
damaged subunits and is functionally impaired and misassem- Heme oxygenase-1 regulates cardiac mitochondrial biogenesis
bled. J Neurosci 26:5256–5264 via Nrf2-mediated transcriptional control of nuclear respiratory
80. Vogel RO, Smeitink JA, Nijtmans LG (2007) Human mito- factor-1. Circ Res 103:1232–1240
chondrial complex I assembly: a dynamic and versatile process. 100. Schulz H (1888) Uber Hefegifte. Pfluger’s Arch Ges Physiol
Biochim Biophys Acta 1767:1215–1227 42:517–541
81. Vogel RO, van den Brand MA, Rodenburg RJ, van den Heuvel 101. Calabrese EJ (1999) Evidence that hormesis represents an
LP, Tsuneoka M, Smeitink JA, Nijtmans LG (2007) Investiga- ‘‘overcompensation’’ response to a disruption in homeostasis.
tion of the complex I assembly chaperones B17.2L and NDU- Ecotoxicol Environ Safety 42:135–137
FAF1 in a cohort of CI deficient patients. Mol Genet Metab 102. Mitchel REJ (2007) Low doses of radiation reduce risk in vivo.
91:176–182 Dose Response 5:1–10
82. Dudkina NV, Kouřil R, Peters K, Braun HP, Boekema EJ (2010) 103. Lacassagne A, Buu-Hoi NP, Rudali G (1945) Induction of the
Structure and function of mitochondrial supercomplexes. Bio- carcinogenic action produced by a weakly carcinogenic hydro-
chim Biophys Acta 1797:664–670 carbon on a highly active carcinogenic hydrocarbon. Br J Exp
83. Wittig I, Schägger H (2009) Supramolecular organization of Pathol 26:5–12
ATP synthase and respiratory chain in mitochondrial mem- 104. Sykes PJ, Morley AA, Hooker AM (2006) The pKZ1 recom-
branes. Biochim Biophys Acta 1787:672–680 bination mutation assay: a sensitive assay for low dose studies.
84. Forquer I, Covian R, Bowman MK, Trumpower BL, Kramer Dose Response 4:91–105
DM (2006) Similar transition states mediate the Q-cycle and 105. Boreham DR, Dolling J-A, Somers C, Quinn J, Mitchel REJ
superoxide production by the cytochrome bc1 complex. J Biol (2006) The adaptive response and protection against heritable
Chem 281:38459–38465 mutations and fetal malformation. Dose Response 4:317–326
85. Bilban M, Haschemi A, Wegiel B, Chin BY, Wagner O, 106. Mothersill C, Seymour CB (2006) Radiation-induced bystander
Otterbein LE (2008) Heme oxygenase and carbon monoxide effects and the DNA paradigm: an ‘‘out of field’’ perspective.
initiate homeostatic signaling. J Mol Med 86:267–279 Mutat Res 597:5–10
86. Calabrese V, Cornelius C, Rizzarelli E, Owen JB, Dinkova- 107. Sakai K, Nomura T, Ina Y (2006) Enhancement of bio-protec-
Kostova AT, Butterfield DA (2009) Nitric oxide in cell survival: tive functions by low dose/dose-rate radiation. Dose Response
a Janus molecule. Antioxid Redox Signal 11:2717–2739 4:327–332
87. Wang W, Fang H, Groom L, Cheng A, Zhang W, Liu J, Wang 108. Scott BR, DiPalma J (2006) Sparsely ionizing diagnostic and
X, Li K, Han P, Zheng M, Yin J, Wang W, Mattson MP, Kao JP, natural background radiations are likely preventing cancer and

123
Neurochem Res (2010) 35:1880–1915 1913

other genomic-instability-associated diseases. Dose Response Seymour C, Sinclair DA, Smith-Sonneborn J, Snow ET, Spear
5:230–255 L, Stevenson DE, Thomas Y, Tubiana M, Williams GM,
109. Redpath JL (2006) Suppression of neoplastic transformation in Mattson MP (2007) Biological stress response terminology:
vitro by low doses of low LET radiation. Dose Response Integrating the concepts of adaptive response and precondi-
4:302–308 tioning stress within a hormetic dose-response framework.
110. Calabrese EJ, Baldwin LA (2000) Chemical hormesis: Its his- Toxicol Appl Pharmacol 222:122–128
torical foundations as a biological hypothesis. Hum Exp Toxicol 132. Flood JF, Smith GE, Cherkin A (1982) Memory retention—
19:2–31 enhancement by cholinergic drug-combinations in mice. Ger-
111. Calabrese EJ, Baldwin LA (2000) The marginalization of hor- ontologist 22:230–231
mesis. Hum Exp Toxicol 19:32–40 133. Flood JF, Smith GE, Cherkin A (1983) Memory retention—
112. Calabrese EJ, Baldwin LA (2000) Radiation hormesis: its his- potentiation of cholinergic drug-combinations in mice. Neuro-
torical foundations as a biological hypothesis. Hum Exp Toxicol biol Aging 4:37–43
19:41–75 134. Flood JF, Smith GE, Cherkin A (1984) Memory retention—
113. Calabrese EJ, Baldwin LA (2000) Radiation hormesis: the enhancement by synergistic oral cholinergic drug-combination
demise of a legitimate hypothesis. Hum Exp Toxicol 19:76–84 in mice. Gerontologist 24:149–158
114. Calabrese EJ, Baldwin LA (2003) Ethanol and hormesis. Crit 135. Flood JF, Smith GE, Cherkin A (1985) Memory enhancement—
Rev Toxicol 33:407–424 supra-additive effect of subcutaneous chlolinergic drug-combi-
115. Calabrese EJ (2008) Hormesis. In: Melnick E, Everitt B (eds) nations in mice. Psychopharmacology 86:61–67
Encyclopedia of quantitative risk assessment and analysis. 136. Calabrese EJ (2008) Neuroscience and hormesis: overview and
Wiley, Chichester, pp 838–844 general findings. Crit Rev Toxicol 38:249–252
116. Calabrese EJ (2009) The road to linearity: shy linearity at low 137. Calabrese EJ, Baldwin LA (2002) Hormesis and high risk
doses became the basis for carcinogen risk assessment. Arch groups. Regul Toxicol Pharmacol 35:14–428
Toxicol 83:203–225 138. Calabrese EJ (2005) Cancer biology and hormesis: human tumor
117. Stebbing ARD (1976) Effects of low metal levels on a clonal cell lines commonly display hormetic (biphasic) dose responses.
hydroid. J Mar Biol Assoc UK 56:977–994 Crit Rev Toxicol 35:463–582
118. Szabadi E (1977) Model of 2 functionally antagonistic receptor 139. Randall WA, Price CW, Welch H (1947) Demonstration of
populations activated by same agonist. J Theor Biol 69:101–112 hormesis (increase in fatality rate) by penicillin. Am J Pub
119. Luckey TD (1992) Radiation hormesis. CRC, Boca Raton Health 37:421–425
120. Calabrese EJ, Baldwin LA (2000) Tales of two similar 140. Welch H, Price CW, Randall WA (1946) Increase in fatality rate
hypotheses: the rise and fall of chemical and radiation hormesis. of E. Typhosa for white mice by streptomycin. J Am Pharm
Hum Exp Toxicol 19:85–97 35:155–158
121. Calabrese EJ, Blain R (2005) The occurrence of hormetic dose 141. Abramowitz J, Dai C, Hirschi KK, Dmitieva RI, Doris PA, Liu
responses in the toxicological literature, the hormesis database: L, Allen JC (2003) Ouabain- and marinobufagenin-induced
an overview. Toxicol Appl Pharmacol 202:289–301 proliferation of human umbilical vein smooth muscle cells and a
122. Mitchel REJ (2006) Low doses of radiation are protective in rat vascular smooth muscle cell lines, A7r5. Circulation 108:
vitro and in vivo: evolutionary origins. Dose Response 4:75–90 3048–3053
123. Thong H-Y, Maibach HI (2008) Hormesis [biological effects of 142. Chueh S-C, Guh J-H, Chen J, Lai M-K, Teng C-M (2001) Dual
low level exposure (BELLE)] and dermatology. Dose Response effects of ouabain on the regulation of proliferation and apoptosis
6:1–15 in human prostatic smooth muscle cells. J Urol 166:347–353
124. Eaton DL, Klaassen CD (2003) Principles of toxicology. In: 143. Gerich FJ, Funke F, Hildebrandt B, Faßhauer M, Müller M (2009)
Casarett & Doull’s essentials of toxicology, Chap 2. The H2O2-mediated modulation of cytosolic signaling and organelle
McGraw-Hill Companies, Inc. pp. 6–20 function in rat hippocampus. Pflugers Arch 458:937–952
125. Calabrese EJ, Ricci PF (2010) Hormesis in environmental 144. Huddleston AT, Tang W, Takeshima H, Hamilton SL, Klann E
health: how hormesis will change the risk assessment process. (2008) Superoxide-induced potentiation in the hippocampus
Encycl Environ Health (in press) requires activation of ryanodine receptor type 3 and ERK.
126. Calabrese EJ (1999) Evidence that hormesis represents an J Neurophysiol 99:1565–1571
‘‘overcompensation’’ response to a disruption in homeostasis. 145. Gopalakrishna R, Gundimeda U, Schiffman JE, McNeill TH
Ecotoxicol Environ Saf 42:135–137 (2008) A direct redox regulation of protein kinase C isoenzymes
127. Calabrese EJ, Blain RB (2009) Hormesis and plant biology. mediates oxidant-induced neuritogenesis in PC12 cells. J Biol
Environ Poll. 157:42–48 Chem 283:14430–14444
128. Calabrese EJ, Baldwin LA (2003) Inorganics and hormesis. Crit 146. Terada LS (2006) Specificity in reactive oxidant signaling: think
Rev Toxicol 33:215–304 globally, act locally. J Cell Biol 174:615–623
129. Calabrese EJ, Baldwin LA (2003) Chemotherapeutics and hor- 147. Mattson MP, Goodman Y, Luo H, Fu W, Furukawa K (1997)
mesis. Crit Rev Toxicol 33:305–354 Activation of NF-kappaB protects hippocampal neurons against
130. Calabrese EJ, Baldwin LA (2003) Peptides and hormesis. Crit oxidative stress-induced apoptosis: evidence for induction of
Rev Toxicol 33:355–406 manganese superoxide dismutase and suppression of peroxyni-
131. Calabrese EJ, Bachmann KA, Bailer AJ, Bolger PM, Borak J, trite production and protein tyrosine nitration. J Neurosci Res
Cai L, Cedergreen N, Cherian MG, Chiueh CC, Clarkson TW, 49:681–697
Cook RR, Diamond DM, Doolittle DJ, Dorato MA, Duke SO, 148. Bruce-Keller AJ, Geddes JW, Knapp PE, McFall RW, Keller
Feinendegen L, Gardner DE, Hart RW, Hastings KL, Hayes JN, Holtsberg FW, Parthasarathy S, Steiner SM, Mattson MP
AW, Hoffmann GR, Ives JA, Jaworowski Z, Johnson TE, Jonas (1999) Anti-death properties of TNF against metabolic poison-
WB, Kaminski NE, Keller JG, Klaunig JE, Knudsen TB, ing: mitochondrial stabilization by MnSOD. J Neuroimmunol
Kozumbo WJ, Lettieri T, Liu SZ, Maisseu A, Maynard KI, 93:53–71
Masoro EJ, McClellan RO, Mehendale HM, Mothersill C, 149. Ekdahl CT, Kokaia Z, Lindvall O (2009) Brain inflammation
Newlin DB, Nigg HN, Oehme FW, Phalen RF, Philbert MA, and adult neurogenesis: the dual role of microglia. Neuroscience
Rattan SI, Riviere JE, Rodricks J, Sapolsky RM, Scott BR, 158:1021–1029

123
1914 Neurochem Res (2010) 35:1880–1915

150. Jiang F, Zhang ZG, Katakowski M, Robin AM, Faber M, Zhang Parkinson’s disease-related insults through the activation of
F, Chopp M (2004) Angiogenesis induced by photodynamic XBP1. Brain Res 1257:16–24
therapy in normal rat brains. Photochem Photobiol 79:494–498 172. Madeo F, Eisenberg T, Kroemer G (2009) Autophagy for the
151. Bandopadhyay R, de Belleroche J (2010) Pathogenesis of Par- avoidance of neurodegeneration. Genes Dev 23:2253–2259
kinson’s disease: emerging role of molecular chaperones. 173. Pennuto M, Tinelli E, Malaguti M, Del Carro U, D’Antonio M,
Trends Mol Med 16:27–36 Ron D, Quattrini A, Feltri ML, Wrabetz L (2008) Ablation of the
152. Morimoto RI (2008) Proteotoxic stress and inducible chaperone UPR-mediator CHOP restores motor function and reduces demy-
networks in neurodegenerative disease and aging. Genes Dev elination in Charcot-Marie-Tooth 1B mice. Neuron 57:393–405
22:1427–1438 174. Sakaki K, Wu J, Kaufman RJ (2008) Protein kinase Cu is
153. Csermely P, Söti C, Blatch GL (2007) Chaperones as parts of required for autophagy in response to stress in the endoplasmic
cellular networks. Adv Exp Med Biol 594:55–63 reticulum. J Biol Chem 283:15370–15380
154. Amijee H, Madine J, Middleton DA, Doig AJ (2009) Inhibitors 175. Schulz TJ, Zarse K, Voigt A, Urban N, Birringer M, Ristow M
of protein aggregation and toxicity. Biochem Soc Trans (2007) Glucose restriction extends Caenorhabditis elegans life
37:692–696 span by inducing mitochondrial respiration and increasing oxi-
155. Mattson MP, Magnus T (2006) Ageing and neuronal vulnera- dative stress. Cell Metab 6:280–293
bility. Nat Rev Neurosci 7:278–294 176. Azad MB, Chen Y, Gibson SB (2009) Regulation of autophagy
156. Saibil HR (2008) Chaperone machines in action. Curr Opin by reactive oxygen species (ROS): implications for cancer
Struct Biol 18:35–42 progression and treatment. Antioxid Redox Signal 11:777–790
157. Glabe CG (2008) Structural classification of toxic amyloid 177. Fujioka Y, Noda NN, Nakatogawa H, Ohsumi Y, Inagaki F
oligomers. J Biol Chem 283:29639–29643 (2010) Dimeric coiled-coil structure of Saccharomyces cerevi-
158. Broadley SA, Hartl FU (2009) The role of molecular chaperones siae Atg16 and its functional significance in autophagy. J Biol
in human misfolding diseases. FEBS Lett 583:2647–2653 Chem 285:1508–1515
159. Gidalevitz T, Kikis EA, Morimoto RI (2010) A cellular per- 178. Jakubowicz-Gil J, Langner E, Wertel I, Piersiak T, Rzeski W
spective on conformational disease: the role of genetic back- (2010) Temozolomide, quercetin and cell death in the MOG-
ground and proteostasis networks. Curr Opin Struct Biol GCCM astrocytoma cell line. Chem Biol Interact [Epub ahead
20:23–32 of print]
160. Nassif M, Matus S, Castillo K, Hetz C (2010) Amyotrophic 179. Yogev O, Goldberg R, Anzi S, Yogev O, Shaulian E (2010) Jun
lateral sclerosis pathogenesis: a journey through the secretory proteins are starvation-regulated inhibitors of autophagy. Cancer
pathway. Antioxid Redox Signal. [Epub ahead of print] Res 70:2318–2327
161. Eizirik DL, Cardozo AK, Cnop M (2008) The role for endo- 180. Luk KC, Mills IP, Trojanowski JQ, Lee VM (2008) Interactions
plasmic reticulum stress in diabetes mellitus. Endocr Rev between Hsp70 and the hydrophobic core of alpha-synuclein
29:42–61 inhibit fibril assembly. Biochemistry 47:12614–12625
162. Hoozemans JJ, van Haastert ES, Nijholt DA, Rozemuller AJ, 181. Rump TJ, Muneer PM, Szlachetka AM, Lamb A, Haorei C,
Eikelenboom P, Scheper W (2009) The unfolded protein Alikunju S, Xiong H, Keblesh J, Liu J, Zimmerman MC, Jones
response is activated in pretangle neurons in Alzheimer’s dis- J, Donohue TM Jr, Persidsky Y, Haorah J (2010) Acetyl-L-
ease hippocampus. Am J Pathol 174:1241–1251 carnitine protects neuronal function from alcohol-induced oxi-
163. Scheper W, Hoozemans JJ (2009) Endoplasmic reticulum pro- dative damage in the brain. Free Radic Biol Med 49:1494–1504
tein quality control in neurodegenerative disease: the good, the 182. Calabrese V, Stella AMG, Calvani M, Butterfield DA (2006)
bad and the therapy. Curr Med Chem 16:615–626 Acetylcarnitine and cellular stress response: roles in nutritional
164. Naidoo N (2009) ER and aging-protein folding and the ER stress redox homeostasis andregulation of longevity genes. J Nutr
response. Ageing Res Rev 8:150–159 Biochem 17:73–88
165. Schröder M (2008) Endoplasmic reticulum stress responses. Cell 183. Jones LL, McDonald D, Borum PR (2009) Acylcarnitines: role
Mol Life Sci 65:862–894 in brain. Progr Lipid Res
166. Verkhratsky A (2004) Endoplasmic reticulum calcium signaling 184. Calabrese V, Rizza V (1999) Formation of propionate after
in nerve cells. Biol Res 37:693–699 short-term ethanol treatment and its interaction with the carni-
167. Jiao J, Huang X, Feit-Leithman RA, Neve RL, Snider W, Dartt tine pool in rat. Alcohol 19:169–176
DA, Chen DF (2005) Bcl-2 enhances Ca(2?) signaling to sup- 185. Calabrese V, Calvani M, Butterfield DA (2004) Increased for-
port the intrinsic regenerative capacity of CNS axons. EMBO J mation of short-chain organic acids after chronic ethanol
24:1068–1078 administration and its interaction with the carnitine pool in rat.
168. Kitao Y, Ozawa K, Miyazaki M, Tamatani M, Kobayashi T, Arch Biochem Biophys 431:271–278
Yanagi H, Okabe M, Ikawa M, Yamashima T, Stern DM, Hori 186. Noland RC, Koves TR, Seiler SE, Lum H, Lust RM, Ilkayeva O,
O, Ogawa S (2001) Expression of the endoplasmic reticulum Stevens RD, Hegardt FG, Muoio DM (2009) Carnitine insuffi-
molecular chaperone (ORP150) rescues hippocampal neurons ciency caused by aging and overnutrition compromises mito-
from glutamate toxicity. J Clin Invest 108:1439–1450 chondrial performance and metabolic control. J Biol Chem
169. Um HS, Kang EB, Leem YH, Cho IH, Yang CH, Chae KR, 284:22840–22852
Hwang DY, Cho JY (2008) Exercise training acts as a thera- 187. Bieber LL (1998) Carnitine. Annu Rev Biochem 57:261–283
peutic strategy for reduction of the pathogenic phenotypes for 188. Evans AM, Fornasini G (2003) Pharmacokinetics of L-carnitine.
Alzheimer’s disease in an NSE=APPsw-transgenic model. Int J Clin Pharmacokinet 42:941–967
Mol Med 22:529–539 189. Rebouche CJ (2004) Kinetics, pharmacokinetics, and regulation
170. Matus S, Lisbona F, Torres M, León C, Thielen P, Hetz C of L-carnitine and acetyl-L-carnitine metabolism. Ann N Y Acad
(2008) The stress rheostat: an interplay between the unfolded Sci 1033:30–41
protein response (UPR) and autophagy in neurodegeneration. 190. Steiber A, Kerner J, Hoppel CL (2004) Carnitine: a nutritional,
Curr Mol Med 8:157–172 biosynthetic, and functional perspective. Mol Aspects Med
171. Sado M, Yamasaki Y, Iwanaga T, Onaka Y, Ibuki T, Nishihara 25:455–473
S, Mizuguchi H, Momota H, Kishibuchi R, Hashimoto T, Wada 191. Koves TR, Ussher JR, Noland RC, Slentz D, Mosedale M,
D, Kitagawa H, Watanabe TK (2009) Protective effect against Ilkayeva O, Bain J, Stevens R, Dyck JR, Newgard CB,

123
Neurochem Res (2010) 35:1880–1915 1915

Lopaschuk GD, Muoio DM (2008) Mitochondrial overload and 206. Traina G, Bernardi R, Rizzo M, Calvani M, Durante M, Brunelli
incomplete fatty acid oxidation contribute to skeletal muscle M (2006) Acetyl-L-carnitine up-regulates expression of voltage-
insulin resistance. Cell Metab 7:45–56 dependent anion channel in the rat brain. Neurochem Int
192. Lombard KA, Olson AL, Nelson SE, Rebouche CJ (1989) 48:673–678
Carnitine status of lactoovovegetarians and strict vegetarian 207. Binienda Z, Virmani A, Przybyla-Zawislak B, Schmued L
adults and children. Am J Clin Nutr 50:301–306 (2004) Neuroprotective effect of L-carnitine in the 3-nitroprop-
193. Brass EP, Hoppel CL, Hiatt WR (1994) Effect of intravenous ionic acid [3-NPA]-evoked neurotoxicity in rats. Neurosci Lett
L-carnitine on carnitine homeostasis and fuel metabolism during 367:264–267
exercise in humans. Clin Pharmacol Ther 55:681–692 208. Lesnefsky EJ, He D, Moghaddas S, Hoppel CL (2006) Reversal
194. Murakami R, Tanaka A, Nakamura H (1997) The effect of of mitochondrial defects before ischemia protects the aged heart.
starvation on brain carnitine concentration in neonatal rats. FASEB J 20:1543–1545
J Pediatr Gastroenterol Nutr 25:385–387 209. Chiechio S, Copani A, Nicoletti F, Gereau RW 4th (2006) L-
195. Bremer J (1983) Carnitine-metabolism and functions. Physiol acetylcarnitine: a proposed therapeutic agent for painful
Rev 63:1420–1480 peripheral neuropathies. Curr Neuropharmacol 4:233–237
196. Zhao S, Xu W, Jiang W, Yu W, Lin Y, Zhang T, Yao J, Zhou L, 210. Chiechio S, Copani A, Zammataro M, Battaglia G, Gereau RW
Zeng Y, Li H, Li Y, Shi J, An W, Hancock SM, He F, Qin L, 4th, Nicoletti F (2010) Transcriptional regulation of type-2
Chin J, Yang P, Chen X, Lei Q, Xiong Y, Guan KL (2010) metabotropic glutamate receptors: an epigenetic path to novel
Regulation of cellular metabolism by protein lysine acetylation. treatments for chronic pain. Trends Pharmacol Sci 31:153–160
Science 327:1000–1004 211. Di Cesare ML, Ghelardini C, Calvani M, Nicolai R, Mosconi L,
197. Alonso-Montes C, Castro MG, Reguero JR, Perrot A, Ozcelik C, Vivoli E, Pacini A, Bartolini A (2007) Protective effect of
Geier C, Posch MG, Morris C, Alvarez V, Ruiz-Ortega M, Coto acetyl-L-carnitine on the apoptotic pathway of peripheral neu-
E (2008) Mitochondrial transcription factor TFA, TFB1 and ropathy. Eur J Neurosci 26:820–827
TFB2: a search for DNA variant/haplotypes and the risk of 212. Watt MJ, Hevener AL (2008) Fluxing the mitochondria to
cardiac hyperthrophy. Dis Markers 25:131–139 insulin resistance. Cell Metab 7:5–6
198. Takahashi H, McCaffery JM, Irizarry RA, Boeke JD (2006) Nu- 213. Adams SH, Hoppel CL, Lok KH, Zhao L, Wong SW, Minkler
cleocytosolic acetylcoenzyme a synthetase is required for histone PE, Hwang DH, Newman JW, Garvey WT (2009) Plasma
acetylation and global transcription. Mol Cell 23:207–217 acylcarnitine profiles suggest incomplete long-chain fatty acid
199. Distler AM, Kerner J, Hoppel CL (2007) Post-translational beta-oxidation and altered tricarboxylic acid cycle activity
modifications of rat liver mitochondrial outer membrane pro- in type 2 diabetic African-American women. J Nutr 139:
teins identified by mass spectrometry. Biochim Biophys Acta 1073–1081
1774:628–636 214. Stephens FB, Constantin-Teodosiu D, Greenhaff PL (2007) New
200. Infante JP, Huszagh VA (2000) Secondary carnitine deficiency insights concerning the role of carnitine in the regulation of fuel
and impaired docosahexaenoic [22:6n–3] acid synthesis: a com- metabolism in skeletal muscle. J Physiol 581:431–444
mon denominator in thepathophysiology of diseases of oxidative 215. Heo YR, Kang CW, Cha S (2001) L-carnitine changes the levels
phosphorylation and beta-oxidation. FEBS Lett 468:1–5 of insulin-like growth factors (IGFs) and IGF binding proteins in
201. Calabrese V, Ravagna A, Colombrita C, Scapagnini G, Gua- streptozotocin-induced diabetic rat. J Nutr Sci Vitaminol
gliano E, Calvani M, Butterfield DA, Giuffrida Stella AM (Tokyo) 47:329–334
(2005) Acetylcarnitine induces heme oxygenase in rat astrocytes 216. Randle PJ, Garland PB, Hales CN, Newsholme EA (1963) The
and protects against oxidative stress: involvement of the tran- glucose fatty-acid cycle. Its role in insulin sensitivity and the
scription factor Nrf2. J Neurosci Res 79:509–521 metabolic disturbances of diabetes mellitus. Lancet 1:785–789
202. McDaniel MA, Maier SF, Einstein GO (2003) ‘‘Brain-specific’’ 217. Abdul HM, Calabrese V, Calvani M, Butterfield DA (2006)
nutrients: a memory cure? Nutrition 19:957–975 Acetyl-L-carnitine-induced up-regulation of heat shock proteins
203. Traina G, Federighi G, Brunelli M, Scuri R (2009) Cytopro- protects cortical neurons against amyloid-beta peptide 1–42-
tective effect of cetyl-L-carnitine evidenced by analysis of gene mediated oxidative stress and neurotoxicity: implications for
expression in the rat brain. Mol Neurobiol 39:101–106 Alzheimer’s disease. J Neurosci Res 84:398–408
204. Calabrese V, Scapagnini G, Ravagna A, Colombrita C, Spadaro 218. Wallace DC (2010) The epigenome and the mitochondrion:
F, Butterfield DA, Giuffrida Stella AM (2004) Increased bioenergetics and the environment. Genes Dev 24:1571–1573
expression of heat shock proteins in rat brain during aging: 219. Kuhn TS (1996) The structure of scientific revolutions. Uni-
relationship with mitochondrial function and glutathione redox versity of Chicago Press, Chicago
state. Mech Ageing Dev 125:325–335 220. Wallace DC (2007) Why do we still have a maternally inherited
205. Traina G, Valleggi S, Bernardi R (2004) Identification of dif- mitochondrial DNA? Insights from evolutionary medicine.
ferentially expressed genes induced in the rat brain by acetyl-L- Annu Rev Biochem 76:781–821
carnitine as evidenced by suppression subtractive hybridisation. 221. Wallace DC (2010) Mitochondrial DNA mutations in disease
Mol Brain Res 132:57–63 and aging. Environ Mol Mutagen 51:440–450

123

You might also like