Tải bản đầy đủ (.pdf) (18 trang)

Báo cáo khoa học: Calcium, mitochondria and oxidative stress in neuronal pathology Novel aspects of an enduring theme pdf

Bạn đang xem bản rút gọn của tài liệu. Xem và tải ngay bản đầy đủ của tài liệu tại đây (188.84 KB, 18 trang )

REVIEW ARTICLE
Calcium, mitochondria and oxidative stress in neuronal
pathology
Novel aspects of an enduring theme
Christos Chinopoulos and Vera Adam-Vizi
Department of Medical Biochemistry, Semmelweis University, Neurobiochemical Group, Hungarian Academy of Sciences, Szentagothai
Knowledge Center, Budapest, Hungary
Background
A long-standing perception is that upon activation of
glutamate receptors followed by a robust Ca
2+
influx,
in situ mitochondria generate reactive oxygen species
(ROS) [1–6]. These studies inferred that mitochondrial
Ca
2+
sequestration is a prerequisite for production of
ROS: abolition of mitochondrial membrane potential
(DYm) by mitochondrial poisons, and thus, electro-
phoretic calcium uptake or direct inhibition of the uni-
porter with ruthenium red prevented ROS generation.
Parallel to these reports, the response of isolated mito-
chondria to calcium loading in terms of ROS produc-
tion has also been scrutinized; it was found that
mitochondrial Ca
2+
uptake led to free radical produc-
tion [7–12]. On the other hand, it was shown that ROS
formation depends steeply on DYm [13–15], and from
a thermodynamic point of view, Ca
2+


uptake occur-
ring at the expense of membrane potential should
result in a decrease in ROS production (in the absence
of respiratory chain inhibitors), as it has also been
demonstrated (reviewed in [16,17]). Nevertheless, brain
mitochondria also generate ROS in a DYm-independ-
ent manner [18–20]. The reason behind the opposing
observations that mitochondrial ROS production
increases or decreases upon Ca
2+
uptake is not
Keywords
alpha-ketoglutarate dehydrogenase;
oxidative stress; permeability transition
pore;store-operated Ca
2+
entry; transient
receptor potential; TRPM2; TRPM7
Correspondence
V. Adam-Vizi, Semmelweis University,
Department of Medical Biochemistry,
Budapest H-1444, PO Box 262, Hungary
Fax: +36 1 2670031
Tel: +36 1 2662773
E-mail:
(Received 18 October 2005, accepted
14 December 2005)
doi:10.1111/j.1742-4658.2005.05103.x
The interplay among reactive oxygen species (ROS) formation, elevated
intracellular calcium concentration and mitochondrial demise is a recurring

theme in research focusing on brain pathology, both for acute and chronic
neurodegenerative states. However, causality, extent of contribution or the
sequence of these events prior to cell death is not yet firmly established.
Here we review the role of the alpha-ketoglutarate dehydrogenase complex
as a newly identified source of mitochondrial ROS production. Further-
more, based on contemporary reports we examine novel concepts as poten-
tial mediators of neuronal injury connecting mitochondria, increased
[Ca
2+
]
c
and ROS ⁄ reactive nitrogen species (RNS) formation; specifically:
(a) the possibility that plasmalemmal nonselective cationic channels con-
tribute to the latent [Ca
2+
]
c
rise in the context of glutamate-induced
delayed calcium deregulation; (b) the likelihood of the involvement of the
channels in the phenomenon of ‘Ca
2+
paradox’ that might be implicated in
ischemia ⁄ reperfusion injury; and (c) how ROS ⁄ RNS and mitochondrial sta-
tus could influence the activity of these channels leading to loss of ionic
homeostasis and cell death.
Abbreviations
2-APB, 2-aminoethoxydiphenyl borate; ADPR, ADP-ribose; DAG, diacylglycerols; DCD, delayed calcium deregulation; KGDHC, a-ketoglutarate
dehydrogenase complex; NMDA, N-methyl-
D-aspartate; PTP, permeability transition pore; RNS, reactive nitrogen species; ROS, reactive
oxygen species; siRNA, short interfering RNA; SOC channel, store-operated Ca

2+
channel.
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 433
entirely clear; a plausible explanation lies in the condi-
tion in which mitochondria are probed for ROS,
specifically whether or not the organelles undergo per-
meability transition pore (PTP) formation. Among the
many features accompanying mitochondrial permeabil-
ity transition (for a full list see [16] and references
therein) loss of glutathione, cytochrome c, substrates
and pyridine nucleotides are characteristic. This leads
to an increase in ROS production from the impaired
mitochondria by multiple means: (a) loss of glutathi-
one from the matrix decreases the antioxidant capacity
resulting in a net ‘steady-state’ increase in the amount
of ROS [21]; (b) loss of cytochrome c impairs the flow
of electrons in the respiratory chain inducing over-
reduction of the complexes, favouring the generation
of ROS [16,17,22]; (c) reduction in the matrix concen-
tration of electron acceptors, i.e. NAD
+
, results in
ROS emission from the a-ketoglutarate dehydrogenase
complex (KGDHC) [23,24].
Mitochondrial formation of ROS-the
role of KGDHC
The first observation of ROS production in mitoch-
ondrial fragments was reported in 1966 by Jensen [25].
Subsequent studies by Britton Chance’s group, estab-
lished that mitochondria generate ROS [26,27]. The

sites of ROS formation within the organelle have been
extensively reviewed elsewhere [17,20,28]. Among
them, complex I [29–31] and III [32–35] of the respirat-
ory chain have attracted most attention. However, in
light of recent results on the substantial contribution
of matrix enzymes (especially KGDHC) on ROS gen-
eration, we believe that in addition to the respiratory
chain, the components of the Krebs cycle should also
be considered as a possible important source of ROS
in mitochondria.
Almost all studies have used respiratory chain inhib-
itors as tools to maximize and to identify potential
sites of ROS production in isolated mitochondria.
They revealed that inhibition of complexes I and III,
respectively, with specific mitochondrial toxins such as
rotenone and antimycin A, results in high rates of
ROS production [29,36,37]. For complex I in partic-
ular, the ‘reverse electron transport’ mode of ROS pro-
duction has gained momentum throughout the past
four decades [38]; reverse electron transport requires
high DYm and is abolished by the complex I inhibitor,
rotenone [18], but the pathophysiological relevance of
this mode of ROS generation is questionable. Similar
approaches have been used successfully to study ROS
production in in situ brain mitochondria present in
isolated nerve terminals (synaptosomes) [39], but no
information is yet available regarding the specific sites
or mechanisms of ROS generation in the absence of
respiratory chain inhibitors.
Numerous reports in isolated or in situ mitochondria

support complex I being regarded as a major site of
ROS production, however, a lingering assumption
remains that all ROS production caused by complex I
inhibitors occurs at the complex I site. There are other
sources of ROS within the mitochondrial matrix that
are in equilibrium with the ratio NAD(P)H ⁄ NAD(P)
+
,
such as the dihydrolipoyl dehydrogenase (Dld) compo-
nent of KGDHC [40]. In intact mitochondria, complex
I inhibition by any means, inevitably results in over-
reduction of most if not all NAD
+
-linked matrix
enzymes.
Among the NAD
+
-linked dehydrogenases that gen-
erate ROS, KGDHC deserves special attention.
KGDHC is a mitochondrial enzyme tightly bound to
the inner mitochondrial membrane on the matrix side
[41]. It (as well as other but not all dehydrogenases)
binds to complex I of the mitochondrial respiratory
chain [42] and may form a part of the TCA cycle
enzyme supercomplex [43]. Mammalian KGDHC is
composed of multiple copies of three enzymes: a-keto-
glutarate dehydrogenase (E1; EC 1.2.4.2), dihydrolipo-
amide succinyltransferase (E2; EC 2.3.1.61), and
dihydrolipoamide dehydrogenase (E3 or Dld; EC
1.8.1.4). Dld is also a part of other multienzyme com-

plexes such as the pyruvate dehydrogenase complex
(PDHC), the branched chain ketoacid dehydrogenase
complex, and the glycine cleavage system [44–47]. The
catalytic mechanism of the a-ketoacid dehydrogenase
complex was reviewed by Bunik [40].
Isolated KGDHC [23] as well as PDHC [24] in isola-
ted and in in situ mitochondria respectively produce
superoxide and H
2
O
2
. Quantitatively, it seems likely
that KGDHC generates the majority of ROS among
dehydrogenases: under conditions of maximum respir-
ation induced with either ADP or an uncoupler,
a-ketoglutarate supports the highest rate of H
2
O
2
pro-
duction [24]. The Dld component of KGDHC, and to
a lesser degree of PDHC, generate ROS in isolated
mouse brain mitochondria [24]. The reasons behind
this quantitative discrepancy among the Dld-contain-
ing dehydrogenases regarding ROS production are at
present, unknown. The isolated Dld subunit is able to
form H
2
O
2

and superoxide radical, accompanying
NADH oxidation [40,48,49]. This observation is
important as to the mechanisms and sites of ROS pro-
duction in mitochondria because the flavin of the Dld
subunit is abundant and possesses a sufficiently negat-
ive redox potential (Em 7.4 ¼ )283 mV) to allow
superoxide formation [50,51]. Moreover, H
2
O
2
produc-
Ca
2+
, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
434 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS
tion by brain mitochondria isolated from heterozygous
knockout mice deficient in Dld is significantly dimin-
ished, as compared to wild-type littermates [24].
Within KGDHC, it is the flavin or the neighbouring
disulfide bridge in the catalytic centre of the Dld com-
ponent that could act as an electron donor for superox-
ide formation [52]. KGDHC is activated by low
concentrations of Ca
2+
and matrix ADP [53–56]. Con-
sidering that KGDHC-mediated ROS production
requires a fully active complex with all the cofactors
and substrates (except NAD
+
), the fact that the enzyme

activity is stimulated by Ca
2+
and ADP may perhaps
account for previous findings that mitochondrial ROS
production was increased by Ca
2+
[7–11,14] and ADP
[30]. Results obtained in our laboratory [23] demon-
strate that Ca
2+
activates ROS production by isolated
KGDHC both in the presence and in the absence of
pyridine nucleotides. Still, the reduced Dld subunit is
the most likely source of ROS under conditions of an
elevated NADPH ⁄ NADP
+
ratio in the mitochondrial
matrix [23,24]. The conditions promoting KGDHC-
mediated ROS production may be any that increase the
intramitochondrial NADH ⁄ NAD
+
ratio (e.g. inhibi-
tion of oxidative phosphorylation or inhibition of any
segment of the mitochondrial electron transport chain).
This hypothesis is favoured by our results showing
that ROS production by isolated KGDHC is strongly
dependent on the NADH ⁄ NAD
+
ratio [23].
The relationship of ROS to KGDHC is extended in

an ‘ouroboros’ fashion to the self-inactivation of the
enzyme by ROS. We demonstrated previously, that
KGDHC is sensitive to inhibition by H
2
O
2
[57]. That
inevitably leads to a decrease in complex I function, as
repeatedly demonstrated [57–61], since KGDHC which
is the rate-limiting step of the TCA cycle provides
NADH as a substrate for the respiratory chain complex.
It is difficult to establish the extent of contribution
of KGDHC and other enzymes to overall ROS pro-
duction in mitochondria, as this is prone to be condi-
tion-dependent (e.g. choice of substrate), in addition to
heavily reliant on non-Krebs cycle enzyme mediated
ROS formation through the respiratory chain; i.e. both
complex I and KGDHC are in equilibrium with the
NAD(P)H ⁄ NAD(P)
+
ratio, and therefore interdepend-
ent on each other concerning ROS formation. Thus,
in organello it might not be possible to accurately esti-
mate the degree of contribution of each ROS-forming
site, because inhibition of ROS production in the one
may aggravate ROS formation in the other, and vice
versa.
The observation that KGDHC generates and is also
self-inactivated by ROS, is of paramount importance in
neuronal pathology. A compelling body of evidence

indicates that mitochondria are the major source of
ROS in several neurodegenerative conditions [37,62].
Also, KGDHC activity is severely reduced in a variety
of neurodegenerative diseases associated with impaired
mitochondrial functions, specifically, Alzheimer’s dis-
ease [63–67], Parkinson’s disease [68–71], progressive su-
pranuclear palsy [72,73] and Wernicke–Korsakoff
syndrome [74]. It is not known if the physical associ-
ation of KGDHC with complex I (see above) plays a
role in the dual deficiency of these protein complexes in
Parkinson’s disease. It appears that neuronal pathology
is preferentially associated with KGDHC deficiency: in
an animal model of diminished KGDHC activity caused
by thiamine deprivation in the diet, neurons are dying,
while endothelial cells, astrocytes and microglia are not
affected. In fact, KGDHC activity is increased in these
non-neuronal cell types [63], which might indicate that
KGDHC deficiency has an etiologic role in the manifes-
tation of some neurodegenerative diseases [75,76]. It
must be emphasized that this multienzyme is the rate-
limiting step of the Krebs cycle, and if altered that
would inpact on the overall energy production in the
affected tissue. Moreover, in vivo studies suggested that
reduced activity of KGDHC predisposes to damage by
toxins, such as 1-methyl-4-phenyl-1,2,3,6-tetrahydro-
pyridine (MPTP) or malonate, reducing the capacity of
neurons to respond to stress [77,78]. In addition, it was
shown recently that reduction in the E2 subunit of
KGDHC is associated with diminished growth of cells
and impaired antioxidant defence systems, without a

reduction in the overall activity of the complex [79].
This finding should come at no surprise: several
enzymes of the TCA cycle (and at least one glycolytic
enzyme [80]) have roles beyond those of just being cycle
participants for the provision of reducing equivalents:
aconitase, isocitrate dehydrogenase and kgd2p (a sub-
unit of KGDHC in yeast equivalent to E2 in mammals),
have two or more different functions, in addition to
having supporting functions for oxidative defences [79],
involving the thioredoxin system [40]. Aconitase acts
also as an iron-responsive element binding protein, iso-
citrate dehydrogenase is an RNA-binding protein, while
kgd2p is a mitochondrial DNA binding protein [81–84].
Mitochondria from different brain regions contain
different amounts of KGDHC [85,86], which may
account for regional vulnerability. For instance, the
cholinergic neurons of the nucleus basalis of Meynert
have high levels of KGDHC, and these neurons are
particularly vulnerable in Alzheimer disease [64].
Nevertheless, the relationship between KGDHC
activity and mitochondrial damage per se is much less
clear. One can speculate that KGDHC-mediated
oxidative stress predisposes the cell to succumb to con-
C. Chinopoulos and V. Adam-Vizi Ca
2+
, mitochondria, ROS in neuronal disease
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 435
comitant adverse conditions; in addition, a diminished
KGDHC activity will lead to insufficient provision of
reducing equivalents, lowering the energetic capacity of

the mitochondria of the affected cell. However, studies
with the KGDHC inhibitor KMV (alpha-keto-beta-
methyl-n-valeric acid) suggest that inhibition of the
enzyme might contribute to cell death by induction of
permeability transition [87].
Permeability transition pore in situ
Permeability transition pore is considered to be a chan-
nel with a large conductance provided by proteins resi-
ding in both the inner and outer mitochondrial
membrane, that is activated by mitochondrial Ca
2+
overloading and other factors including oxidative stress
[88,89]. In neurons the presence of PTP in situ has not
gained wide acceptance among investigators and
results published in the literature support views of both
its presence and absence in several in vitro models of
neurodegeneration [90–98]. One of the possible reasons
for this discrepancy is that sensitivity to cyclosporin A
is considered pathognomonic for mitochondrial PTP
(see also [90]). Cyclosporin A is a potent inhibitor of
PTP in isolated liver mitochondria [99] that has been
demonstrated to be effective also in situ in this and
other organs [100–103]. The sensitivity of isolated
brain mitochondria to cyclosporin A depends highly
on the conditions: in the absence of adenine nucleo-
tides and magnesium, cyclosporin A mitigates Ca
2+
-in-
duced mitochondrial pore formation [104,105]
however, in the presence of 3 mm ATP plus 1 mm free

Mg
2+
, cyclosporin A is only marginally effective, pro-
vided that mitochondria are challenged by boluses of
CaCl
2
[104]. In the case that Ca
2+
loading occurs
slowly, cyclosporin A delays onset of PTP in brain
mitochondria extensively, even in the presence of aden-
ine nucleotides and magnesium [106]. The caveat here
is that despite the decreased ATP levels to less than
the millimolar range during ischemic deenergizing,
ADP levels approximate 400 lm [107], and the K
i
for
inhibition of the PTP by ADP is in the low micromo-
lar range [108]. Moreover, in situ neuronal mitochon-
dria are exposed to bolus-like additions of Ca
2+
[109]
during intense glutamate receptor stimulation for the
duration of seizure activity or reversal of glutamate
transporters throughout ischemia [110]. Ca
2+
cycling
across the mitochondrial inner membrane ensues sub-
sequently [111]. On the other hand, intense stimulation
of N-methyl-d-aspartate (NMDA) receptors on cul-

tured cerebellar granule and hippocampal neurons cau-
ses major ultrastructural alterations of mitochondria,
implying the activation of some form of PTP [112,113].
Mitochondrial alterations suggestive of pore opening is
also demonstrated in vivo, during the postischemic per-
iod in the gerbil brain [114]. Yet, to identify these
in situ mitochondrial alterations as the PTP on the basis
of the functional ⁄ morphological ⁄ pharmacological cri-
teria applied for isolated mitochondria is rather hasty.
Collectively, the sensitivity of glutamate-induced
neuronal damage to cyclosporin A as diagnostic for
PTP occurrence is unreliable. This ambiguity is also nur-
tured by the complex pharmacology of cyclosporin A
and its affinity to non-PTP targets [90,115] that could be
involved in the manifestation of neuronal injury [116], in
addition to the fact that PTP may not have a causal role
in excitotoxic cell death. It is to be noted that the magni-
tude of the literature involving cyclosporin A unrelated
to mitochondria is 12 times larger than that implicating
PTP! The nonimmunosuppressant analogue, N-methyl-
valine-4-cyclosporin also gave contrasting results, con-
ferring neuronal protection against excitotoxicity in
some studies [92,117,118], but not in others [94].
What could be important though, is the role of the
in situ mitochondrial pore formation in dictating the
type of death that the ill-fated neuron will follow. A
most simplistic view is that this pore will promote
apoptosis due to release of cytochrome c followed by
activation of caspases [119,120], provided that pertain-
ing conditions divert the type of cell death from the

necrotic to the apoptotic pathway [121,122]. The role
of mitochondria in apoptosis and necrosis has been
extensively reviewed elsewhere [121,123–131]. Recently
however, a blow was delivered to the conception that
PTP contributes to apoptotic cell death by three
almost simultaneous and independent reports using
cyclophilin D knockout mice [132–134]. Cyclophilin D
is a component of the PTP complex [135,136] and it is
the target for cyclosporin A. As expected, mitochon-
dria isolated from the cyclophilin D knockout mice
were much less susceptible to various PTP-inducing
regimes, that are otherwise sensitive to cyclosporin A
treatment (see also [137]). Unexpectedly though, tissues
obtained from mutant mice were not more resistant to
several apoptotic stimuli than those from their wild-
type littermates; however, the resistance of the mutant
mice to treatments known to result in necrotic cell
death was much higher than in control mice.
Mitochondrial Ca
2+
-flux pathways and
relation to signal transduction
In general, the contribution of mitochondria to intra-
cellular Ca
2+
homeostasis is ascribed to uptake and
release through the uniporter, the mitochondrial
Na
+
⁄ Ca

2+
exchanger, the PTP (both high- and low-
Ca
2+
, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
436 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS
conductance mode) and other less well characterized
pathways, such as the ‘Na
+
-independent pathway for
Ca
2+
efflux’ and a H
+
⁄ Ca
2+
antiporter [89,138]. With
the exception of the high-conductance mode of PTP
and the uniporter, none of these molecular complexit-
ies have been described to be modulated by any signal
transduction mediators. High-conductance PTP is
known to be affected by matrix Ca
2+
and ROS [89].
Also the uniporter is supposed to be activated only if
extramitochondrial Ca
2+
levels exceed a certain thresh-
old concentration, termed the ‘set-point’ [139]; how-
ever, this has been challenged recently, showing that

in situ mitochondria accumulate Ca
2+
well below the
set-point, in permeabilized rat adrenal glomerulosal
cells [140]. Nonetheless, despite that mitochondria are
increasingly viewed as active mediators of [Ca
2+
]
c
regulation, the pathways that these organelles use to
achieve this task are rather passive.
To this repertoire of Ca
2+
influx and efflux mecha-
nisms across the mitochondrial membranes, a novel
Ca
2+
-efflux-only machinery has been recently added: a
channel located in the inner membrane activated by dia-
cylglycerols (DAGs) [141]. This is either a single channel
with numerous substates (mean conductance  200 pS),
or multiple channels with unequal conductance. DAGs
cause a biphasic form of Ca
2+
efflux in Ca
2+
-loaded
mitochondria: the first wave of efflux is attributed to the
activation of the DAG-sensitive nonselective cationic
channels; the second wave is due to opening of the PTP.

It is not yet known how activation of the former leads
to induction of the latter. One is tempted to hypothesize
that the initial Ca
2+
efflux through DAG-sensitive
channels causes intense Ca
2+
cycling due to reuptake
by the uniporter, leading to PTP. However, cyclospo-
rin A fails to defend against the secondary Ca
2+
efflux
in liver mitochondria in the presence of DAGs, in which
the immunosuppressant otherwise confers significant
protection against PTP induction.
The role of DAG-sensitive mitochondrial channels in
physiological [Ca
2+
]
c
regulation can easily be envis-
aged: upon phosphatidylinositol (4,5) bisphosphate
(PIP
2
) hydrolysis, inositol-1,4,5-triphosphate (IP
3
) dif-
fuses in the cytosol to activate IP
3
receptors on the

endoplasmic reticulum releasing Ca
2+
to the cytoplasm,
followed by triggering of Ca
2+
influx from the extracel-
lular space [142]. The role of mitochondria in shaping
Ca
2+
transients during such events is recognized in lim-
iting Ca
2+
diffusion, and secondarily relieving Ca
2+
-
mediated negative feedback on the Ca
2+
flux pathways
themselves [143]. However, the other obligatory meta-
bolite of PIP
2
catabolism ) DAG ) may regulate the
role of mitochondria in shaping those [Ca
2+
]
c
tran-
sients: mitochondrial DAG-sensitive channels would
re-release sequestered matrix Ca
2+

only in the vicinity
where DAGs are formed most likely in microdomains,
since this second messenger is extremely lipophilic and
does not diffuse into the aqueous cytosol.
Mitochondrial permeabilization and the
delayed calcium deregulation
The association of ROS to a possible PTP induction
prior to neuronal cell death has received much atten-
tion in relation to the delayed, irreversible rise in
[Ca
2+
]
c
following a prolonged glutamate stimulus,
coined by Nicholls’ group as ‘delayed calcium deregu-
lation, DCD’ [144] that commits a neuron to die
[145–148]. DCD was originally described by Manev
and colleagues [149], further characterized by the
groups of Thayer [150] and Tymianski [146]. However,
credit should also be given to an earlier work by Con-
nor and colleagues, showing that a short exposure
(1–3 s) of CA1 hippocampal neurons to NMDA causes
an abrupt elevation in [Ca
2+
]
c
that returns to baseline;
a subsequent exposure to NMDA of the same duration
a few minutes later leads to an irreversible and sus-
tained increase in intracellular [Ca

2+
]
c
in apical dend-
rites [151]. DCD is invariably demonstrated in every
neuronal cell type studied, i.e. spinal [146], hippocam-
pal [150], cerebellar granule [152], striatal [117] and
cortical neurons [93,153]. The phenomenon is not
observed if high extracellular K
+
is alternatively
employed to elevate [Ca
2+
]
c
; this led to the proposal
of a ‘source specificity’ of Ca
2+
-induced neurotoxicity
[146]. However, this was subsequently challenged by
studies demonstrating that activation of NMDA recep-
tors produces much larger Ca
2+
entry than activation
of voltage-dependent Ca
2+
channels by high extracel-
lular K
+
[154].

This secondary [Ca
2+
]
c
rise is not inhibitable by
postglutamate addition of antagonists of NMDA or
non-NMDA receptors [94,145,149,150], nor by block-
ing voltage-dependent Ca
2+
or Na
+
channels
[145,149,150,155]. Results supporting views that DCD
is comprised of an active Ca
2+
influx pathway
[93,146,149,150,155–159] as well as those indicating a
failure in Ca
2+
efflux mechanisms [160–162], are avail-
able in the literature. It is anticipated that these seem-
ingly opposing observations represent two-facets of the
same problem: even in the earliest report on DCD by
Manev and colleagues [149] it was shown that during
the postglutamate period neurons still accumulate
45
Ca
2+
within 30 s exposure to the isotope, without
any statistically significant difference seen in the pres-

ence or absence of N-methyl-d-aspartate receptors/non-
N-methyl-d-aspartate receptors/voltage dependent Ca
2+
C. Chinopoulos and V. Adam-Vizi Ca
2+
, mitochondria, ROS in neuronal disease
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 437
channels (NMDAR ⁄ non-NMDAR ⁄ VDCC blockers).
That attests to the presence of a discrete pathway for
Ca
2+
influx. Yet, it was recently demonstrated that in
an almost identical paradigm of excitotoxicity, the plas-
malemmal Na
+
⁄ Ca
2+
exchanger (in particular the
NCX3 isoform) is cleaved by calpain, severing the high
capacity Ca
2+
efflux pathway in neurons [161]. Provi-
ded that the Ca
2+
influx pathway is most likely a chan-
nel, it must saturate [163] imposing a continuous load of
calcium to the neuron. The turning point upon which
the cell looses the ability to buffer the incoming calcium
resulting in an abrupt, sustained and irreversible
increase in [Ca

2+
]
c
, probably coincides with the clea-
vage of the exchanger (but see [164]). Therefore, inhibi-
tion of the, as yet unidentified, Ca
2+
influx pathway or
prevention of NCX proteolysis should thwart DCD.
The question arises: what is the nature of the Ca
2+
influx pathway?
Non-selective cationic channel(s) and
the DCD
As mentioned above, inhibition of NMDAR⁄ non-
NMDAR ⁄ voltage-dependent Ca
2+
or Na
+
channels
after the initial Ca
2+
and Na
2+
influx through the glu-
tamate receptors, failed to prevent DCD. Yet, DCD
demands the existence of a discrete pathway as it pre-
cedes, and eventually leads to, plasma membrane leaki-
ness and cell death [145,146,148]. The notion that
DCD is not attributed to the ‘traditionally’ recognized

Ca
2+
channels, such as glutamate receptor-operated or
voltage-gated Ca
2+
channels has been proposed previ-
ously [157,158]. Along this line, it was shown that a
secondary activation of a nonselective cation conduct-
ance, termed postexposure current (I
pe
), is induced sub-
sequent to excitotoxic application of NMDA to
hippocampal neurons that probably contributes to the
delayed Ca
2+
rise [156].
Relevant to the inability of the glutamate receptor
blockers to prevent DCD, antiexcitotoxic therapy util-
izing these compounds failed to produce a better out-
come in clinical trials concerning stroke treatment
[165–167]. To address this setback, Aarts and collea-
gues [159] examined the possibility that an overlooked
neurotoxic process was occurring in a well-established
in vitro model of excitotoxicity, by subjecting cultured
neurons to oxygen–glucose deprivation. This treatment
results in neuronal demise through NMDAR activa-
tion [168,169]. It was found that a member of the
melastatin branch of the transient receptor potential
channel (TRP) family, TRPM7 [170], mediates a lethal
cation current loading the neurons with Ca

2+
and
Na
+
. This nonselective current was activated by ROS
and reactive nitrogen species (RNS), and its abolition
permitted the survival of neurons previously destined
to die from prolonged anoxia, regardless of the pres-
ence or absence of NMDAR blockers.
In a subsequent study, we explored the hypothesis
that a TRP channel contributes to the manifestation of
DCD [93]. A pharmacological approach was used,
applying 2-aminoethoxydiphenyl borate (2-APB) or
La
3+
to cultured cortical neurons challenged by pro-
longed glutamatergic stimulation. We observed that
2-APB and La
3+
diminished the delayed Ca
2+
rise
with a 50% inhibitory concentration of 62 ± 9 lm
and 7.2 ± 3 lm, respectively. Both substances are
known to inhibit TRP channels in addition to acting
on many other targets; 2-APB blocks store-operated
Ca
2+
(SOC) channels [171], the IP
3

receptor [172], the
sarco-endoplasmic reticulum Ca
2+
ATPase (SERCA)
pump [173], voltage-dependent K
+
channels [174], gap
junctions [175] and the cyclosporin A-insensitive PTP
[104], while La
3+
blocks SOC [176] and voltage-
dependent Ca
2+
channels [177]. Almost all non-TRP
targets are irrelevant or have been previously excluded
concerning the origin of DCD, except for the cyclospo-
rin A-insensitive PTP that is abolished by 2-APB in
isolated brain mitochondria [104]. However, in our
hands, bongkrekic acid ameliorated the cyclosporin A-
insensitive PTP but not the DCD [93,104]. From this
study we concluded that a TRP channel could be
responsible for the Ca
2+
influx part of DCD. In gen-
eral, the two inhibitors that we used do not distinguish
among individual members of the TRP family, but for
reasons explained below, it is tempting to speculate
that it is the TRPM7. Unfortunately, we could not
achieve silencing of TRPM7 expression in our cultures
with short interfering RNA (siRNA); primary neurons

are notoriously vulnerable to transfection techniques,
as opposed to the ease and the high efficiency of the
procedure in cell lines. Hopefully, the development of
novel approaches such as the conjugation of siRNA to
penetratins [178,179] will assist transfection protocols
and allow research on primary neuronal cultures to
benefit from the tremendous potential of siRNA.
The connection of TRPM7 to DCD may lie in the
observation that this channel is activated by ROS and
RNS [159]. For a long time, ROS were considered to
be responsible for DCD [180]; however, in a recent
study it was deduced that the increased ROS produc-
tion is a consequence, rather than a cause of DCD
[181]. In the latter study the authors also demonstrated
that the increase in superoxide radical formation is
predominantly associated with extramitochondrial
phospholipase A(2) (PLA
2
) activation, and it does not
emanate from mitochondria. That may be in contrast
Ca
2+
, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
438 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS
with previous reports claiming that ROS are the induc-
ers of DCD. However over the years concerns have
arisen as for the reliability of ROS-detecting dyes,
given that some are affected by confounding parame-
ters such as mitochondrial membrane potential (see
discussion in [181]). The development of new dyes des-

cribed recently will no doubt contribute to the clarifi-
cation of these matters [182].
In light of the recent observations though, one could
argue that TRPM7 is not the Ca
2+
influx pathway of
DCD, as the increase in superoxide radical appears
after the secondary [Ca
2+
]
c
rise. However, the exact
species activating TRPM7 is not known, and the extent
of ROS production necessary to activate the channel
maybe less than the detection level of the probes used.
In addition, ROS ⁄ RNS could be just one of the many
activators of the channel [183], while others that might
play a significant role could be also mobilized upon
prolonged glutamate exposure. We have found that by
elevating intracellular [Mg
2+
]
i
DCD is abolished in cul-
tured cortical neurons [93], and it is known that
TRPM7 receives strong negative feedback by intracel-
lular Mg
2+
[170]. In addition, TRPM7 currents
induced by oxygen–glucose deprivation promote fur-

ther ROS production [159], and this could partially
explain the results of Vesce and colleagues, detecting
an increase in superoxide formation after the delayed
secondary [Ca
2+
]
c
rise [181]. In our opinion, TRPM7
is one of the best possible candidates for the Ca
2+
influx part of DCD; other good candidates are TRPM2
(see below) and the calcium-permeable acid-sensing ion
channel [184] (not reviewed here).
Nonselective cationic channels and the
’Ca
2+
paradox’
In spite of the widely accepted role of [Ca
2+
]
c
deregula-
tion in the manifestation of neurodegeneration, exactly
how Ca
2+
ions mediate neural cell death is less clear
[185]. One of the most important unresolved issues is the
mechanism by which [Ca
2+
]

c
increases to excessively
high levels in neurons following periods of intense neur-
onal activation. Reaching further from the possibility of
the involvement of TRP channels in the delayed calcium
deregulation, these proteins could participate in an addi-
tional overlooked pathway of Ca
2+
influx that may per-
tain during ischemia ⁄ reperfusion or other type of
pathology. Large [Ca
2+
]
c
increases are known to be trig-
gered by reintroduction of ‘normal’ Ca
2+
concentra-
tions to the extracellular milieu after the tissue has
experienced a [Ca
2+
]
e
-free challenge, or at least a severe
reduction in extracellular calcium concentration, termed
‘Ca
2+
paradox’. The free extracellular calcium concen-
tration falls dramatically in several brain disease states:
(a) during or after ischemia (0.1–0.28 mm [186–189]); (b)

traumatic brain injury (0.1 mm [190]); (c) severe hypo-
glycemia (0.12 mm [191]); and (d) spreading depression
(0.06–0.08 mm [192]). Reduction of extracellular Ca
2+
is mostly due to robust influx of the cation to the intra-
cellular milieu, although the appearance of lactate in the
interstitium during ischemia, with the ability to chelate
divalent ions significantly, also plays a role [193,194].
The Ca
2+
paradox
Paradoxical Ca
2+
increases were originally described in
isolated heart preparations [195] and subsequently
shown to be associated with tissue damage in this and
other organs, including the kidney and skeletal muscle
[196,197], but not in others, i.e. liver [198]. Interestingly,
the possibility that paradoxical Ca
2+
influx contributes
to neuronal degeneration was put forward almost
20 years ago [199], but the vast majority of subsequent
work on [Ca
2+
]
c
elevation during excitotoxicity has
since concentrated on other Ca
2+

entry routes, inclu-
ding glutamate receptors and voltage-gated Ca
2+
chan-
nels. Unfortunately, this emphasis has not resulted in
any clinically useful intervention to limit the neuronal
damage following ischemia ⁄ reperfusion or other brain
injury. Inescapably, within a context of ischemia ⁄ reper-
fusion in which a Ca
2+
paradox is encompassed [200],
concomitant adverse conditions, e.g. oxygen–glucose
deprivation, associated ROS production and many
more ) reviewed in [201] ) contribute to irreversible
tissue damage. Nevertheless, the paradoxical Ca
2+
rise
per se remains a poorly understood phenomenon. What
is known though, is that abolition of in situ mitochond-
rial respiration and oxidative phosphorylation protects
against the Ca
2+
paradox [202]. The reasons behind
this unexpected finding are not yet understood. A num-
ber of theories were put forward, including the deleteri-
ous effect of overloading mitochondria with Ca
2+
that
can only happen in respiring mitochondria.
Possible mechanisms underlying neuronal

paradoxical Ca
2+
-increases
While multiple mechanisms could contribute to para-
doxical Ca
2+
increases, the most current interest is the
activation of novel nonselective cation channels. It is
known that reduction of [Ca
2+
]
e
activates nonselective
cation currents in hippocampal neurons [203] and neo-
cortical nerve terminals [204] termed csNSC and NSC,
respectively, as well as in thalamic neurons [205], vagal
afferent nerves [206] and ventricular myocytes [207].
Such currents may underlie paradoxical Ca
2+
increases
C. Chinopoulos and V. Adam-Vizi Ca
2+
, mitochondria, ROS in neuronal disease
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 439
activated by transient [Ca
2+
]
e
removal. We have also
observed the appearance of a nonselective, noninacti-

vating cation conductance upon reducing extracellular
Ca
2+
and Mg
2+
in cultures of cortical neurons, as well
as in cortical and hippocampal neurons in brain slices
from adult mice, raising the possibility that such cur-
rents are readily available in these cells (C. Chinopou-
los, unpublished data). Furthermore, we have recently
reported that cultured cortical neurons exhibit para-
doxical Ca
2+
entry [93] and it is conceivable that the
[Ca
2+
]
c
rise is a result of the ‘tails’ of these currents.
Alternative mechanisms for paradoxical Ca
2+
rise lie
in a diversity of molecular complexities: lowering
[Ca
2+
]
e
reduces the shielding of negatively charged
groups located at the membrane surface affecting the
voltage-dependent activation of various ion channels

[163,208]. In addition, it is the biophysical property of
many types of channels to conduct monovalents in a
less controlled manner in the absence of divalent cati-
ons, such as the I
crac
-conducting channel [209,210],
voltage-gated Ca
2+
channels [211–215], Na
+
channels
[216,217], K
+
channels [218], other unidentified chan-
nels [203–207] and many members of the TRP family
of channels (see below). In extreme cases, channel
selectivity is lost when [Ca
2+
]
e
is reduced to ultra-low
(<1 lm) concentrations [219].
Apart from this biophysical property of channels, a
number of receptor-based mechanisms are modulated
by [Ca
2+
]
e
: (a) the Ca
2+

-sensing receptor is activated by
millimolar changes in [Ca
2+
]
e
, and is widely distributed
in mammalian tissues including brain [220]; (b) hemi-
gap channels in horizontal cells of the catfish retina are
activated by [Ca
2+
]
e
decreases [221] and it is likely that
gap junctional regulation could be strongly modified by
[Ca
2+
]
e
in the central nervous system [222]; (c) metabo-
tropic glutamate receptors 1, 3 and 5 [223] are activated
by physiological [Ca
2+
]
e
fluctuations in the synaptic
cleft [224]; and (d) the Gamma-aminobutyric acid (B)
GABA
B
receptor also possesses Ca
2+

sensing proper-
ties, potentiating GABA responses upon increase of
[Ca
2+
]
e
[225]. It is not yet known whether these addi-
tional Ca
2+
-sensing mechanisms may act alone or in
concert with nonselective Ca
2+
channels in producing
significant excitotoxic Ca
2+
increases following ischemic
insults.
TRP channels as candidates for paradoxical
Ca
2+
-increases
TRP channels are widely expressed in mammalian tis-
sues, especially in neurons of the central nervous
system [226]. With a few notable exceptions, the phy-
siological roles of TRP channels in neurons remain
largely unknown [226–231]. Diverse neuropathological
conditions were also found to implicate TRP family
members: (a) mucolipidosis type IV [232] involving a
channel from the distant polycystin branch (TRPP);
(b) TRPV4 in neuropathic pain [233], and – as dis-

cussed above ) (c) TRPM7 in neuronal death caused
by oxygen–glucose deprivation [159]; the latter study
also proposed the possibility of TRPM2 involvement,
a view supported by more recent observations on oxi-
dative stress-induced cell death [234]. Furthermore,
ROS were specifically shown to trigger the opening of
TRPC3 [235], TRPM2 [236–238] and TRPM7 [159]. In
preliminary experiments, we have observed that the
presence of ROS abolishes [Ca
2+
]
c
decay during the
paradoxical Ca
2+
rise and converts it to a progressive
[Ca
2+
]
c
rise (C. Chinopoulos, unpublished data).
Of particular interest however, are the observations
that a number of TRP channels are activated by a
decrease in [Ca
2+
]
e
, raising the possibility that they
could contribute to paradoxical Ca
2+

increases. Recent
descriptions have included the Drosophila TRP channel
[239], TRPC1 and TRPC3 [240], TRPC6 [241], TRPC7
[242,243], and TRPM7 [159].
Mitochondrial permeabilization and a
possible link to TRP channel activation
Among the known activators of some members of the
TRP family, NAD
+
and its catabolite ADP-ribose
(ADPR) were described to activate TRPM2 [244–247],
in addition to the fact that the channel is stimulated
by ROS ⁄ RNS [236,238,246]. Furthermore, it was dem-
onstrated that the major source of free ADPR medi-
ating the activation of TRPM2 in cultured cells were
the mitochondria [248]. One could link these observa-
tions to the fact that opening of the PTP causes the
release of mitochondrial NAD
+
followed by its hydro-
lysis by an extramitochondrial NAD
+
glycohydrolase
to ADPR [103,249]. It is tempting to speculate that
this ADPR in conjunction with ROS produced upon
loss of mitochondrial integrity, activates the nonselec-
tive TRPM2 allowing a large Ca
2+
and Na
+

load to
enter the cytosol. Since both high [Ca
2+
]
c
and ROS
promote mitochondrial pore formation, it seems that
the order of appearance of a pore or TRPM2 activa-
tion is trivial; what is probably more important is that
activation of the one can lead to activation of the
other, completing a vicious cycle. Intriguingly, silen-
cing the expression of TRPM7 with siRNA, led to an
accompanying decrease in TRPM2 expression. This
suggests that the two transcripts might be coordinately
regulated, raising the possibility that a fraction of the
oxygen–glucose deprivation-induced current recorded
Ca
2+
, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
440 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS
earlier [159] is mediated by TRPM2 or TRPM7 hetero-
multimers, a structural arrangement commonly occur-
ring among TRP channels [250,251]. Further
implications of TRP channels in relation to the overall
metabolic state of the cell in hypoxia have been
reviewed elsewhere [252].
Trp channels and ionic homeostasis
In view of the fact that most TRP channels are nonse-
lective, in addition to allowing Ca
2+

ions to enter the
cytosol they also permit Na
+
influx and K
+
efflux
[226,253,254]. The ominous effects of an elevated
[Na
+
]
i
are mostly associated with cell swelling and acti-
vation of the Na
+
⁄ Ca
2+
exchanger causing Ca
2+
influx. However, it is possible that the effect of an
increased [Na
+
]
i
may be directly on mitochondria as
recently demonstrated, diminishing the half-life of mit-
ochondrially encoded mRNA, without involving Ca
2+
[255,256]. In addition it was recently shown that in
mature hippocampal slices, NAD(P)H transients during
postsynaptic neuronal activation are not mediated by

Ca
2+
, but rather reflect alterations in [Na
+
]
i
. That may
explain our previous results in isolated nerve terminals
showing that in the presence of an oxidative stress a
concomitant elevation in [Na
+
]
i
acts deleteriously on in
situ mitochondria [257]. The effect of K
+
loss from the
cytoplasm is commonly ignored; however, it was shown
that it can promote neuronal apoptosis [258–260]. To
what extent ) if any ) the activation of TRP channels
is associated with alterations of Na
+
and K
+
homeos-
tasis in neurodegeneration, is currently unknown. Nev-
ertheless, the fact that these proteins are intensely
expressed in the central nervous system [251,254,261]
and their ever-increasing roles in physiology and
pathology being discovered [253,262], identify them as

excellent novel targets amenable to pharmacological
manipulation [254,263,264].
References
1 Reynolds IJ & Hastings TG (1995) Glutamate induces
the production of reactive oxygen species in cultured
forebrain neurons following NMDA receptor activa-
tion. J Neurosci 15, 3318–3327.
2 Dugan LL, Sensi SL, Canzoniero LM, Handran SD,
Rothman SM, Lin TS, Goldberg MP & Choi DW
(1995) Mitochondrial production of reactive oxygen
species in cortical neurons following exposure to
N-methyl-D-aspartate. J Neurosci 15, 6377–6388.
3 Luetjens CM, Bui NT, Sengpiel B, Munstermann G,
Poppe M, Krohn AJ, Bauerbach E, Krieglstein J &
Prehn JH (2000) Delayed mitochondrial dysfunction in
excitotoxic neuron death: cytochrome c release and a
secondary increase in superoxide production. J Neu-
rosci 20, 5715–5723.
4 Carriedo SG, Sensi SL, Yin HZ & Weiss JH (2000)
AMPA exposures induce mitochondrial Ca
2+
overload
and ROS generation in spinal motor neurons in vitro.
J Neurosci 20, 240–250.
5 Sengpiel B, Preis E, Krieglstein J & Prehn JH (1998)
NMDA-induced superoxide production and neurotoxi-
city in cultured rat hippocampal neurons: role of mito-
chondria. Eur J Neurosci 10, 1903–1910.
6 Gunasekar PG, Kanthasamy AG, Borowitz JL & Isom
GE (1995) NMDA receptor activation produces con-

current generation of nitric oxide and reactive oxygen
species: implication for cell death. J Neurochem 65,
2016–2021.
7 Dykens JA (1994) Isolated cerebral and cerebellar
mitochondria produce free radicals when exposed to
elevated CA
2+
and Na
+
: implications for neurodegen-
eration. J Neurochem 63, 584–591.
8 Kowaltowski AJ, Netto LE & Vercesi AE (1998) The
thiol-specific antioxidant enzyme prevents mitochon-
drial permeability transition. Evidence for the partici-
pation of reactive oxygen species in this mechanism.
J Biol Chem 273, 12766–12769.
9 Kowaltowski AJ, Castilho RF & Vercesi AE (1995)
Ca
2+
-induced mitochondrial membrane permeabiliza-
tion: role of coenzyme Q redox state. Am J Physiol
269, C141–C147.
10 Kowaltowski AJ, Castilho RF & Vercesi AE (1996)
Opening of the mitochondrial permeability transition
pore by uncoupling or inorganic phosphate in the pre-
sence of Ca
2+
is dependent on mitochondrial-generated
reactive oxygen species. FEBS Lett 378, 150–152.
11 Kowaltowski AJ, Naia-d, a-Silva ES, Castilho RF &

Vercesi AE (1998) Ca
2+
-stimulated mitochondrial
reactive oxygen species generation and permeability
transition are inhibited by dibucaine or Mg
2+
. Arch
Biochem Biophys 359, 77–81.
12 Maciel EN, Vercesi AE & Castilho RF (2001) Oxidative
stress in Ca
2+
-induced membrane permeability trans-
ition in brain mitochondria. J Neurochem 79, 1237–1245.
13 Korshunov SS, Skulachev VP & Starkov AA (1997)
High protonic potential actuates a mechanism of pro-
duction of reactive oxygen species in mitochondria.
FEBS Lett 416, 15–18.
14 Starkov AA, Polster BM & Fiskum G (2002) Regulation
of hydrogen peroxide production by brain mitochondria
by calcium and Bax. J Neurochem 83, 220–228.
15 Starkov AA & Fiskum G (2003) Regulation of brain
mitochondrial H
2
O
2
production by membrane potential
and NAD(P) H redox state. J Neurochem 86, 1101–1107.
16 Starkov AA, Chinopoulos C & Fiskum G (2004) Mito-
chondrial calcium and oxidative stress as mediators of
ischemic brain injury. Cell Calcium 36, 257–264.

C. Chinopoulos and V. Adam-Vizi Ca
2+
, mitochondria, ROS in neuronal disease
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 441
17 Andreyev AY, Kushnareva YE & Starkov AA (2005)
Mitochondrial metabolism of reactive oxygen species.
Biochemistry (Moscow) 70, 200–214.
18 Votyakova TV & Reynolds IJ (2001) DeltaPsi (m)-
dependent and -independent production of reactive
oxygen species by rat brain mitochondria. J Neurochem
79, 266–277.
19 Sipos I, Tretter L & Adam-Vizi V (2003) The produc-
tion of reactive oxygen species in intact isolated nerve
terminals is independent of the mitochondrial mem-
brane potential. Neurochem Res 28, 1575–1581.
20 Adam-Vizi V (2005) Production of reactive oxygen spe-
cies in brain mitochondria: contribution by electron
transport chain and non-electron transport chain
sources. Antioxid Redox Signal 7, 1140–1149.
21 Anderson MF & Sims NR (2002) The effects of focal
ischemia and reperfusion on the glutathione content of
mitochondria from rat brain subregions. J Neurochem
81, 541–549.
22 Cai J & Jones DP (1998) Superoxide in apoptosis.
Mitochondrial generation triggered by cytochrome c
loss. J Biol Chem 273, 11401–11404.
23 Tretter L & Adam-Vizi V (2004) Generation of
reactive oxygen species in the reaction catalyzed by
alpha-ketoglutarate dehydrogenase. J Neurosci 24,
7771–7778.

24 Starkov AA, Fiskum G, Chinopoulos C, Lorenzo BJ,
Browne SE, Patel MS & Beal MF (2004) Mitochon-
drial alpha-ketoglutarate dehydrogenase complex
generates reactive oxygen species. J Neurosci 24,
7779–7788.
25 Jensen PK (1966) Antimycin-insensitive oxidation of
succinate and reduced nicotinamide-adenine dinucleo-
tide in electron-transport particles. I. pH dependency
and hydrogen peroxide formation. Biochim Biophys
Acta 122, 157–166.
26 Loschen G, Flohe L & Chance B (1971) Respiratory
chain linked H
2
O
2
production in pigeon heart mito-
chondria. FEBS Lett 18, 261–264.
27 Boveris A & Chance B (1973) The mitochondrial gen-
eration of hydrogen peroxide. General properties and
effect of hyperbaric oxygen. Biochem J 134, 707–716.
28 Turrens JF (2003) Mitochondrial formation of reactive
oxygen species. J Physiol 552, 335–344.
29 Lenaz G (2001) The mitochondrial production of react-
ive oxygen species: mechanisms and implications in
human pathology. IUBMB Life 52, 159–164.
30 Barja G (1999) Mitochondrial oxygen radical genera-
tion and leak: sites of production in states 4 and 3,
organ specificity, and relation to aging and longevity.
J Bioenerg Biomembr 31, 347–366.
31 Herrero A & Barja G (2000) Localization of the site of

oxygen radical generation inside the Complex I of
heart and nonsynaptic brain mammalian mitochondria.
J Bioenerg Biomembr 32, 609–615.
32 Turrens JF & Boveris A (1980) Generation of super-
oxide anion by the NADH dehydrogenase of bovine
heart mitochondria. Biochem J 191, 421–427.
33 Loschen G, Azzi A, Richter C & Flohe L (1974) Super-
oxide radicals as precursors of mitochondrial hydrogen
peroxide. FEBS Lett 42, 68–72.
34 Dionisi O, Galeotti T, Terranova T & Azzi A (1975)
Superoxide radicals and hydrogen peroxide formation
in mitochondria from normal and neoplastic tissues.
Biochim Biophys Acta 403, 292–300.
35 Boveris A, Cadenas E & Stoppani AO (1976) Role of
ubiquinone in the mitochondrial generation of hydro-
gen peroxide. Biochem J 156, 435–444.
36 Turrens JF (1997) Superoxide production by the mito-
chondrial respiratory chain. Biosci Report 17, 3–8.
37 Murphy AN, Fiskum G & Beal MF (1999) Mitochon-
dria in neurodegeneration: bioenergetic function in cell
life and death. J Cereb Blood Flow Metab 19, 231–245.
38 Hinkle PC, Butow RA, Racker E & Chance B (1967)
Partial resolution of the enzymes catalyzing oxidative
phosphorylation. XV. Reverse electron transfer in the
flavin-cytochrome beta region of the respiratory chain
of beef heart submitochondrial particles. J Biol Chem
242, 5169–5173.
39 Sipos I, Tretter L & Adam-Vizi V (2003) Quantitative
relationship between inhibition of respiratory com-
plexes and formation of reactive oxygen species in iso-

lated nerve terminals. J Neurochem 84, 112–118.
40 Bunik VI (2003) 2-Oxo acid dehydrogenase complexes
in redox regulation. Eur J Biochem 270, 1036–1042.
41 Maas E & Bisswanger H (1990) Localization of the
alpha-oxoacid dehydrogenase multienzyme complexes
within the mitochondrion. FEBS Lett 277, 189–190.
42 Sumegi B & Srere PA (1984) Complex I binds several
mitochondrial NAD-coupled dehydrogenases. J Biol
Chem 259, 15040–15045.
43 Lyubarev AE & Kurganov BI (1989) Supramolecular
organization of tricarboxylic acid cycle enzymes. Bio-
systems 22, 91–102.
44 Koike K, Hamada M, Tanaka N, Otsuka KI, Ogasa-
hara K & Koike M (1974) Properties and subunit com-
position of the pig heart 2-oxoglutarate dehydrogenase.
J Biol Chem 249, 3836–3842.
45 Patel MS & Roche TE (1990) Molecular biology and
biochemistry of pyruvate dehydrogenase complexes.
FASEB J 4, 3224–3233.
46 Reed LJ & Hackert ML (1990) Structure-function rela-
tionships in dihydrolipoamide acyltransferases. J Biol
Chem 265, 8971–8974.
47 Kochi H, Seino H & Ono K (1986) Inhibition of gly-
cine oxidation by pyruvate, alpha-ketoglutarate, and
branched-chain alpha-keto acids in rat liver mitochon-
dria: presence of interaction between the glycine clea-
vage system and alpha-keto acid dehydrogenase
complexes. Arch Biochem Biophys 249, 263–272.
Ca
2+

, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
442 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS
48 Huennekens FM, Basford RE & Gabrio BW (1955)
An oxidase for reduced diphosphopyridine nucleotide.
J Biol Chem 213, 951–967.
49 Gazaryan IG, Krasnikov BF, Ashby GA, Thorneley
RN, Kristal BS & Brown AM (2002) Zinc is a potent
inhibitor of thiol oxidoreductase activity and stimulates
reactive oxygen species production by lipoamide dehy-
drogenase. J Biol Chem 277, 10064–10072.
50 Kunz WS & Gellerich FN (1993) Quantification of the
content of fluorescent flavoproteins in mitochondria
from liver, kidney cortex, skeletal muscle, and brain.
Biochem Med Metab Biol 50, 103–110.
51 Kunz WS & Kunz W (1985) Contribution of different
enzymes to flavoprotein fluorescence of isolated rat liver
mitochondria. Biochim Biophys Acta 841, 237–246.
52 Bunik VI & Sievers C (2002) Inactivation of the 2-oxo
acid dehydrogenase complexes upon generation of
intrinsic radical species. Eur J Biochem 269, 5004–5015.
53 Hamada M, Koike K, Nakaula Y, Hiraoka T & Koike
M (1975) A kinetic study of the alpha-keto acid dehy-
drogenase complexes from pig heart mitochondria.
J Biochem (Tokyo) 77, 1047–1056.
54 Mcminn CL & Ottaway JH (1977) Studies on the
mechanism and kinetics of the 2-oxoglutarate dehydro-
genase system from pig heart. Biochem J 161, 569–581.
55 Wan B, Lanoue KF, Cheung JY & Scaduto RC Jr
(1989) Regulation of citric acid cycle by calcium. J Biol
Chem 264, 13430–13439.

56 Kiselevsky YV, Ostrovtsova SA & Strumilo SA (1990)
Kinetic characterization of the pyruvate and oxogluta-
rate dehydrogenase complexes from human heart. Acta
Biochim Pol 37, 135–139.
57 Tretter L & Adam-Vizi V (2000) Inhibition of Krebs
cycle enzymes by hydrogen peroxide: a key role of
[alpha]-ketoglutarate dehydrogenase in limiting NADH
production under oxidative stress. J Neurosci 20, 8972–
8979.
58 Kumar MJ, Nicholls DG & Andersen JK (2003) Oxi-
dative a-ketoglutarate dehydrogenase inhibition via
subtle elevations in monoamine oxidase B levels
results in loss of spare respiratory capacity: Implica-
tions for Parkinson’s disease. J Biol Chem 278,
46432–46439.
59 Chinopoulos C, Tretter L & Adam-Vizi V (1999)
Depolarization of in situ mitochondria due to hydro-
gen peroxide-induced oxidative stress in nerve term-
inals: inhibition of alpha-ketoglutarate dehydrogenase.
J Neurochem 73, 220–228.
60 Chinopoulos C, Tretter L & Adam-Vizi V (2000)
Reversible depolarization of in situ mitochondria by
oxidative stress parallels a decrease in NAD(P)H level
in nerve terminals. Neurochem Int 36, 483–488.
61 Chinopoulos C & Adam-Vizi V (1999) Depolarization
of in situ mitochondria by hydrogen peroxide in nerve
terminals. Ann NY Acad Sci 893, 269–272.
62 Beal MF (1996) Mitochondria, free radicals, and
neurodegeneration. Curr Opin Neurobiol 6, 661–666.
63 Gibson GE, Blass JP, Beal MF & Bunik V (2005) The

alpha-ketoglutarate-dehydrogenase complex: a media-
tor between mitochondria and oxidative stress in
neurodegeneration. Mol Neurobiol 31, 43–64.
64 Gibson GE, Sheu KF, Blass JP, Baker A, Carlson KC,
Harding B & Perrino P (1988) Reduced activities of
thiamine-dependent enzymes in the brains and periph-
eral tissues of patients with Alzheimer’s disease. Arch
Neurol 45, 836–840.
65 Butterworth RF & Besnard AM (1990) Thiamine-
dependent enzyme changes in temporal cortex of
patients with Alzheimer’s disease. Metab Brain Dis 5,
179–184.
66 Mastrogiacomo F, Bergeron C & Kish SJ (1993) Brain
alpha-ketoglutarate dehydrogenase complex activity in
Alzheimer’s disease. J Neurochem 61, 2007–2014.
67 Terwel D, Bothmer J, Wolf E, Meng F & Jolles J (1998)
Affected enzyme activities in Alzheimer’s disease are sen-
sitive to antemortem hypoxia. J Neurol Sci 161, 47–56.
68 Mizuno Y, Matuda S, Yoshino H, Mori H, Hattori N
& Ikebe S (1994) An immunohistochemical study on
alpha-ketoglutarate dehydrogenase complex in Parkin-
son’s disease. Ann Neurol 35, 204–210.
69 Mizuno Y, Ikebe S, Hattori N, Nakagawa-Hattori Y,
Mochizuki H, Tanaka M & Ozawa T (1995) Role of
mitochondria in the etiology and pathogenesis of Par-
kinson’s disease. Biochim Biophys Acta 1271, 265–274.
70 Jimenez-Jimenez FJ, Molina JA, Hernanz A, Fernan-
dez-Vivancos EBF, Barcenilla B, Gomez-Escalonilla C,
Zurdo M, Berbel A & Villanueva C (1999) Cerebro-
spinal fluid levels of thiamine in patients with

Parkinson’s disease. Neurosci Lett 271, 33–36.
71 Gibson GE, Kingsbury AE, Xu H, Lindsay JG,
Daniel S, Foster OJ, Lees AJ & Blass JP (2003)
Deficits in a tricarboxylic acid cycle enzyme in brains
from patients with Parkinson’s disease. Neurochem
Int 43, 129–135.
72 Albers DS, Augood SJ, Park LC, Browne SE, Martin
DM, Adamson J, Hutton M, Standaert DG, Vonsattel
JP, Gibson GE & Beal MF (2000) Frontal lobe dys-
function in progressive supranuclear palsy: evidence for
oxidative stress and mitochondrial impairment.
J Neurochem 74, 878–881.
73 Park LC, Albers DS, Xu H, Lindsay JG, Beal MF &
Gibson GE (2001) Mitochondrial impairment in the
cerebellum of the patients with progressive supranuc-
lear palsy. J Neurosci Res 66, 1028–1034.
74 Butterworth RF, Kril JJ & Harper CG (1993) Thia-
mine-dependent enzyme changes in the brains of alco-
holics: relationship to the Wernicke–Korsakoff
syndrome. Alcohol Clin Exp Res 17, 1084–1088.
75 Mastrogiacomo F, Lamarche J, Dozic S, Lindsay G,
Bettendorff L, Robitaille Y, Schut L & Kish SJ (1996)
C. Chinopoulos and V. Adam-Vizi Ca
2+
, mitochondria, ROS in neuronal disease
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 443
Immunoreactive levels of alpha-ketoglutarate dehydro-
genase subunits in Friedreich’s ataxia and spinocerebel-
lar ataxia type 1. Neurodegeneration 5, 27–33.
76 Gibson GE, Zhang H, Sheu KF, Bogdanovich N,

Lindsay JG, Lannfelt L, Vestling M & Cowburn RF
(1998) Alpha-ketoglutarate dehydrogenase in Alzhei-
mer brains bearing the APP670 ⁄ 671 mutation. Ann
Neurol 44, 676–681.
77 Klivenyi P, Starkov AA, Calingasan NY, Gardian G,
Browne SE, Yang L, Bubber P, Gibson GE, Patel MS
& Beal MF (2004) Mice deficient in dihydrolipoamide
dehydrogenase show increased vulnerability to MPTP,
malonate and 3-nitropropionic acid neurotoxicity.
J Neurochem 88, 1352–1360.
78 Sheu KF, Calingasan NY, Lindsay JG & Gibson GE
(1998) Immunochemical characterization of the defi-
ciency of the alpha-ketoglutarate dehydrogenase com-
plex in thiamine-deficient rat brain. J Neurochem 70,
1143–1150.
79 Shi Q, Chen HL, Xu H & Gibson GE (2005)
Reduction in the E2k subunit of the alpha-
ketoglutarate dehydrogenase complex has effects inde-
pendent of complex activity. J Biol Chem 280,
10888–10896.
80 Sun YJ, Chou CC, Chen WS, Wu RT, Meng M &
Hsiao CD (1999) The crystal structure of a multifunc-
tional protein: phosphoglucose isomerase ⁄ autocrine
motility factor ⁄ neuroleukin. Proc Natl Acad Sci USA
96, 5412–5417.
81 Kennedy MC, Mende-Mueller L, Blondin GA & Bei-
nert H (1992) Purification and characterization of cyto-
solic aconitase from beef liver and its relationship to
the iron-responsive element binding protein. Proc Natl
Acad Sci USA 89, 11730–11734.

82 Elzinga SD, van Bednarz ALOK, Dekker PJ & Grivell
LA (1993) Yeast mitochondrial NAD(+)-dependent
isocitrate dehydrogenase is an RNA-binding protein.
Nucl Acids Res 21, 5328–5331.
83 Jeffery CJ (1999) Moonlighting proteins. Trends Bio-
chem Sci 24, 8–11.
84 Kaufman BA, Newman SM, Hallberg RL, Slaughter
CA, Perlman PS & Butow RA (2000) In organello for-
maldehyde crosslinking of proteins to mtDNA: identifi-
cation of bifunctional proteins. Proc Natl Acad Sci
USA 97, 7772–7777.
85 Calingasan NY, Baker H, Sheu KF & Gibson GE (1994)
Distribution of the alpha-ketoglutarate dehydrogenase
complex in rat brain. J Comp Neurol 346, 461–479.
86 Park LC, Calingasan NY, Sheu KF & Gibson GE
(2000) Quantitative alpha-ketoglutarate dehydrogenase
activity staining in brain sections and in cultured cells.
Anal Biochem 277, 86–93.
87 Huang HM, Ou HC, Xu H, Chen HL, Fowler C &
Gibson GE (2003) Inhibition of alpha-ketoglutarate
dehydrogenase complex promotes cytochrome c release
from mitochondria, caspase-3 activation, and necrotic
cell death. J Neurosci Res 74 , 309–317.
88 Zoratti M & Szabo I (1995) The mitochondrial perme-
ability transition. Biochim Biophys Acta 1241, 139–176.
89 Bernardi P (1999) Mitochondrial transport of cations:
channels, exchangers, and permeability transition.
Physiol Rev 79, 1127–1155.
90 Duchen MR (2000) Mitochondria and Ca
2+

in cell phys-
iology and pathophysiology. Cell Calcium 28, 339–348.
91 Nicholls DG & Budd SL (2000) Mitochondria and
neuronal survival. Physiol Rev 80, 315–360.
92 Vergun O, Keelan J, Khodorov BI & Duchen MR
(1999) Glutamate-induced mitochondrial depolarisation
and perturbation of calcium homeostasis in cultured rat
hippocampal neurones. J Physiol 519 Part 2, 451–466.
93 Chinopoulos C, Gerencser AA, Doczi J, Fiskum G &
Adam-Vizi V (2004) Inhibition of glutamate-induced
delayed calcium deregulation by 2-APB and La
3+
in
cultured cortical neurones. J Neurochem 91, 471–483.
94 Castilho RF, Hansson O, Ward MW, Budd SL &
Nicholls DG (1998) Mitochondrial control of acute
glutamate excitotoxicity in cultured cerebellar granule
cells. J Neurosci 18 , 10277–10286.
95 Dubinsky JM & Levi Y (1998) Calcium-induced activa-
tion of the mitochondrial permeability transition in
hippocampal neurons. J Neurosci Res 53, 728–741.
96 Hoyt KR, Mclaughlin BA, Higgins DS Jr & Reynolds
IJ (2000) Inhibition of glutamate-induced mitochon-
drial depolarization by tamoxifen in cultured neurons.
J Pharmacol Exp Ther 293, 480–486.
97 Scanlon JM & Reynolds IJ (1998) Effects of oxidants
and glutamate receptor activation on mitochondrial
membrane potential in rat forebrain neurons. J Neuro-
chem 71, 2392–2400.
98 Hoyt KR, Sharma TA & Reynolds IJ (1997) Trifluoper-

azine and dibucaine-induced inhibition of glutamate-
induced mitochondrial depolarization in rat cultured
forebrain neurones. Br J Pharmacol 122, 803–808.
99 Fournier N, Ducet G & Crevat A (1987) Action of
cyclosporine on mitochondrial calcium fluxes. J Bio-
energ Biomembr 19, 297–303.
100 Johans M, Milanesi E, Franck M, Johans C, Liobikas
J, Panagiotaki M, Greci L, Principato G, Kinnunen
PK, Bernardi P, Costantini P & Eriksson O (2005)
Modification of permeability transition pore arginine(s)
by phenylglyoxal derivatives in isolated mitochondria
and mammalian cells. Structure-function relationship of
arginine ligands. J Biol Chem 280, 12130–12136.
101 Soriano ME, Nicolosi L & Bernardi P (2004) Desensiti-
zation of the permeability transition pore by cyclos-
porin a prevents activation of the mitochondrial
apoptotic pathway and liver damage by tumor necrosis
factor-alpha. J Biol Chem 279, 36803–36808.
102 Petronilli V, Penzo D, Scorrano L, Bernardi P & Di
Lisa F (2001) The mitochondrial permeability transi-
Ca
2+
, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
444 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS
tion, release of cytochrome c and cell death. Correla-
tion with the duration of pore openings in situ. J Biol
Chem 276, 12030–12034.
103 Di Lisa F, Menabo R, Canton M, Barile M &
Bernardi P (2001) Opening of the mitochondrial
permeability transition pore causes depletion of mito-

chondrial and cytosolic NAD+ and is a causative
event in the death of myocytes in postischemic
reperfusion of the heart. J Biol Chem 276, 2571–
2575.
104 Chinopoulos C, Starkov AA & Fiskum G (2003)
Cyclosporin A-insensitive permeability transition in
brain mitochondria: inhibition by 2-aminoethoxydiphe-
nyl borate. J Biol Chem 278, 27382–27389.
105 Kristian T, Weatherby TM, Bates TE & Fiskum G
(2002) Heterogeneity of the calcium-induced permeabil-
ity transition in isolated non-synaptic brain mitochon-
dria. J Neurochem 83, 1297–1308.
106 Chalmers S & Nicholls DG (2003) The relationship
between free and total calcium concentrations in the
matrix of liver and brain mitochondria. J Biol Chem
278, 19062–19070.
107 Ekholm A, Katsura K, Kristian T, Liu M, Folbergrova
J & Siesjo BK (1993) Coupling of cellular energy state
and ion homeostasis during recovery following brain
ischemia. Brain Res 604, 185–191.
108 Haworth RA & Hunter DR (1980) Allosteric inhibition
of the Ca
2+
-activated hydrophilic channel of the mit-
ochondrial inner membrane by nucleotides. J Membr
Biol 54, 231–236.
109 Peng TI, Jou MJ, Sheu SS & Greenamyre JT (1998)
Visualization of NMDA receptor-induced mitochon-
drial calcium accumulation in striatal neurons. Exp
Neurol 149, 1–12.

110 Rossi DJ, Oshima T & Attwell D (2000) Glutamate
release in severe brain ischaemia is mainly by reversed
uptake. Nature 403, 316–321.
111 Wang GJ & Thayer SA (2002) NMDA-induced
calcium loads recycle across the mitochondrial inner
membrane of hippocampal neurons in culture.
J Neurophysiol 87, 740–749.
112 Isaev NK, Zorov DB, Stelmashook EV, Uzbekov RE,
Kozhemyakin MB & Victorov IV (1996) Neurotoxic
glutamate treatment of cultured cerebellar granule cells
induces Ca
2+
-dependent collapse of mitochondrial
membrane potential and ultrastructural alterations of
mitochondria. FEBS Lett 392, 143–147.
113 Pivovarova NB, Nguyen HV, Winters CA, Brantner
CA, Smith CL & Andrews SB (2004) Excitotoxic cal-
cium overload in a subpopulation of mitochondria trig-
gers delayed death in hippocampal neurons. J Neurosci
24, 5611–5622.
114 Dux E, Mies G, Hossmann KA & Siklos L (1987) Cal-
cium in the mitochondria following brief ischemia of
gerbil brain. Neurosci Lett 78, 295–300.
115 Hallak H, Ramadan B & Rubin R (2001) Tyrosine
phosphorylation of insulin receptor substrate-1 (IRS-1)
by oxidant stress in cerebellar granule neurons: modu-
lation by N-methyl-D-aspartate through calcineurin
activity. J Neurochem 77, 63–70.
116 Dawson TM, Steiner JP, Dawson VL, Dinerman JL,
Uhl GR & Snyder SH (1993) Immunosuppressant

FK506 enhances phosphorylation of nitric oxide
synthase and protects against glutamate neurotoxicity.
Proc Natl Acad Sci USA 90, 9808–9812.
117 Alano CC, Beutner G, Dirksen RT, Gross RA &
Sheu SS (2002) Mitochondrial permeability transition
and calcium dynamics in striatal neurons upon
intense NMDA receptor activation. J Neurochem 80,
531–538.
118 Khaspekov L, Friberg H, Halestrap A, Viktorov I &
Wieloch T (1999) Cyclosporin A and its nonimmuno-
suppressive analogue N-Me-Val-4-cyclosporin A miti-
gate glucose ⁄ oxygen deprivation-induced damage to rat
cultured hippocampal neurons. Eur J Neurosci 11,
3194–3198.
119 Liu X, Kim CN, Yang J, Jemmerson R & Wang X
(1996) Induction of apoptotic program in cell-free
extracts: requirement for dATP and cytochrome c. Cell
86, 147–157.
120 Hengartner MO (2000) The biochemistry of apoptosis.
Nature 407, 770–776.
121 Kim JS, He L & Lemasters JJ (2003) Mitochondrial
permeability transition: a common pathway to necrosis
and apoptosis. Biochem Biophys Res Commun 304,
463–470.
122 Lemasters JJ, Nieminen AL, Qian T, Trost LC, Elmore
SP, Nishimura Y, Crowe RA, Cascio WE, Bradham
CA, Brenner DA & Herman B (1998) The mitochon-
drial permeability transition in cell death: a common
mechanism in necrosis, apoptosis and autophagy. Bio-
chim Biophys Acta 1366, 177–196.

123 Polster BM & Fiskum G (2004) Mitochondrial
mechanisms of neural cell apoptosis. J Neurochem 90,
1281–1289.
124 Kroemer G (2003) Mitochondrial control of apoptosis:
an introduction. Biochem Biophys Res Commun 304,
433–435.
125 Van Loo G, Saelens X, Van Gurp M, Macfarlane M,
Martin SJ & Vandenabeele P (2002) The role of mito-
chondrial factors in apoptosis: a Russian roulette with
more than one bullet. Cell Death Differ 9, 1031–1042.
126 Zamzami N & Kroemer G (2001) The mitochondrion
in apoptosis: how Pandora’s box opens. Nat Rev Mol
Cell Biol 2, 67–71.
127 Desagher S & Martinou JC (2000) Mitochondria as the
central control point of apoptosis. Trends Cell Biol 10,
369–377.
128 Green DR & Reed JC (1998) Mitochondria and apop-
tosis. Science 281, 1309–1312.
C. Chinopoulos and V. Adam-Vizi Ca
2+
, mitochondria, ROS in neuronal disease
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 445
129 Susin SA, Zamzami N & Kroemer G (1998) Mitochon-
dria as regulators of apoptosis: doubt no more. Bio-
chim Biophys Acta 1366, 151–165.
130 Green D & Kroemer G (1998) The central executioners
of apoptosis: caspases or mitochondria? Trends Cell
Biol 8, 267–271.
131 Kroemer G, Dallaporta B & Resche-Rigon M (1998)
The mitochondrial death ⁄ life regulator in apoptosis

and necrosis. Annu Rev Physiol 60, 619–642.
132 Schinzel AC, Takeuchi O, Huang Z, Fisher JK, Zhou
Z, Rubens J, Hetz C, Danial NN, Moskowitz MA &
Korsmeyer SJ (2005) Cyclophilin D is a component of
mitochondrial permeability transition and mediates
neuronal cell death after focal cerebral ischemia. Proc
Natl Acad Sci USA 102, 12005–12010.
133 Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska
H, Hambleton MA, Brunskill EW, Sayen MR, Gott-
lieb RA, Dorn GW, Robbins J & Molkentin JD (2005)
Loss of cyclophilin D reveals a critical role for mito-
chondrial permeability transition in cell death. Nature
434, 658–662.
134 Nakagawa T, Shimizu S, Watanabe T, Yamaguchi O,
Otsu K, Yamagata H, Inohara H, Kubo T & Tsuji-
moto Y (2005) Cyclophilin D-dependent mitochondrial
permeability transition regulates some necrotic but not
apoptotic cell death. Nature 434, 652–658.
135 Woodfield K, Ruck A, Brdiczka D & Halestrap AP
(1998) Direct demonstration of a specific
interaction between cyclophilin-D and the adenine
nucleotide translocase confirms their role in the mito-
chondrial permeability transition. Biochem J 336,
287–290.
136 Crompton M, Virji S & Ward JM (1998) Cyclophilin-
D binds strongly to complexes of the voltage-depen-
dent anion channel and the adenine nucleotide
translocase to form the permeability transition pore.
Eur J Biochem 258, 729–735.
137 Basso E, Fante L, Fowlkes J, Petronilli V, Forte MA

& Bernardi P (2005) Properties of the permeability
transition pore in mitochondria devoid of Cyclophilin
D. J Biol Chem 280, 18558–18561.
138 Nicholls D & Akerman K (1982) Mitochondrial cal-
cium transport. Biochim Biophys Acta 683, 57–88.
139 Nicholls DG (1978) The regulation of extramitochond-
rial free calcium ion concentration by rat liver
mitochondria. Biochem J 176, 463–474.
140 Pitter JG, Maechler P, Wollheim CB & Spat A (2002)
Mitochondria respond to Ca
2+
already in the sub-
micromolar range: correlation with redox state. Cell
Calcium 31, 97–104.
141 Chinopoulos C, Starkov AA, Grigoriev S, Dejean LM,
Kinnally KW, Liu X, Ambudkar IS & Fiskum G
(2005) Diacylglycerols activate mitochondrial cationic
channel(s) and release sequestered Ca
2+
. J Bioenerg
Biomembr 37, 237–247.
142 Mikoshiba K & Hattori M (2000) IP3 receptor-oper-
ated calcium entry. Sci STKE 2000, E1.
143 Bianchi K, Rimessi A, Prandini A, Szabadkai G &
Rizzuto R (2004) Calcium and mitochondria: mechan-
isms and functions of a troubled relationship. Biochim
Biophys Acta 1742, 119–131.
144 Nicholls DG & Budd SL (1998) Mitochondria and
neuronal glutamate excitotoxicity. Biochim Biophys
Acta 1366, 97–112.

145 Tymianski M, Charlton MP, Carlen PL & Tator CH
(1993) Secondary Ca
2+
overload indicates early neur-
onal injury which precedes staining with viability indi-
cators. Brain Res 607, 319–323.
146 Tymianski M, Charlton MP, Carlen PL & Tator CH
(1993) Source specificity of early calcium neurotoxicity
in cultured embryonic spinal neurons. J Neurosci 13,
2085–2104.
147 Witt MR, Dekermendjian K, Frandsen A, Schousboe
A & Nielsen M (1994) Complex correlation between
excitatory amino acid-induced increase in the intracel-
lular Ca
2+
concentration and subsequent loss of neur-
onal function in individual neocortical neurons in
culture. Proc Natl Acad Sci USA 91, 12303–12307.
148 Limbrick DD Jr, Churn SB, Sombati S & Delorenzo
RJ (1995) Inability to restore resting intracellular cal-
cium levels as an early indicator of delayed neuronal
cell death. Brain Res 690, 145–156.
149 Manev H, Favaron M, Guidotti A & Costa E (1989)
Delayed increase of Ca
2+
influx elicited by glutamate:
role in neuronal death. Mol Pharmacol 36, 106–112.
150 Randall RD & Thayer SA (1992) Glutamate-induced
calcium transient triggers delayed calcium overload and
neurotoxicity in rat hippocampal neurons. J Neurosci

12, 1882–1895.
151 Connor JA, Wadman WJ, Hockberger PE & Wong
RK (1988) Sustained dendritic gradients of Ca
2+
induced by excitatory amino acids in CA1 hippocampal
neurons. Science 240, 649–653.
152 Budd SL & Nicholls DG (1996) Mitochondria, calcium
regulation, and acute glutamate excitotoxicity in cul-
tured cerebellar granule cells. J Neurochem 67, 2282–
2291.
153 Rajdev S & Reynolds IJ (1994) Glutamate-induced
intracellular calcium changes and neurotoxicity in corti-
cal neurons in vitro: effect of chemical ischemia. Neuro-
science 62, 667–679.
154 Hyrc K, Handran SD, Rothman SM & Goldberg MP
(1997) Ionized intracellular calcium concentration
predicts excitotoxic neuronal death: observations with
low-affinity fluorescent calcium indicators. J Neurosci
17, 6669–6677.
155 Hartley DM & Choi DW (1989) Delayed rescue of
N-methyl-D-aspartate receptor-mediated neuronal
injury in cortical culture. J Pharmacol Exp Ther 250,
752–758.
Ca
2+
, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
446 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS
156 Chen QX, Perkins KL, Choi DW & Wong RK (1997)
Secondary activation of a cation conductance is
responsible for NMDA toxicity in acutely isolated

hippocampal neurons. J Neurosci 17, 4032–4036.
157 Limbrick DD Jr, Pal S & Delorenzo RJ (2001) Hippo-
campal neurons exhibit both persistent Ca
2+
influx
and impairment of Ca
2+
sequestration ⁄ extrusion mech-
anisms following excitotoxic glutamate exposure. Brain
Res 894, 56–67.
158 Limbrick DD Jr, Sombati S & Delorenzo RJ (2003)
Calcium influx constitutes the ionic basis for the main-
tenance of glutamate-induced extended neuronal depo-
larization associated with hippocampal neuronal death.
Cell Calcium 33, 69–81.
159 Aarts M, Iihara K, Wei WL, Xiong ZG, Arundine M,
Cerwinski W, Macdonald JF & Tymianski M (2003) A
key role for TRPM7 channels in anoxic neuronal
death. Cell 115, 863–877.
160 Khodorov B, Pinelis V, Golovina V, Fajuk D, Andre-
eva N, Uvarova T, Khaspekov L & Victorov I (1993)
On the origin of a sustained increase in cytosolic Ca
2+
concentration after a toxic glutamate treatment of the
nerve cell culture. FEBS Lett 324, 271–273.
161 Bano D, Young KW, Guerin CJ, Lefeuvre R, Rothwell
NJ, Naldini L, Rizzuto R, Carafoli E & Nicotera P
(2005) Cleavage of the plasma membrane Na
+
⁄ Ca

2+
exchanger in excitotoxicity. Cell 120, 275–285.
162 Ward MW, Rego AC, Frenguelli BG & Nicholls DG
(2000) Mitochondrial membrane potential and gluta-
mate excitotoxicity in cultured cerebellar granule cells.
J Neurosci 20, 7208–7219.
163 Hille B (2001) Ionic Channels of Excitable Membranes.
Sinauer Associates, Inc, Sunderland, MA.
164 Kiedrowski L, Czyz A, Baranauskas G, Li XF & Lyt-
ton J (2004) Differential contribution of plasmalemmal
Na ⁄ Ca exchange isoforms to sodium-dependent cal-
cium influx and NMDA excitotoxicity in depolarized
neurons. J Neurochem 90, 117–128.
165 Ikonomidou C & Turski L (2002) Why did
NMDA receptor antagonists fail clinical trials for
stroke and traumatic brain injury? Lancet Neurol 1,
383–386.
166 Muir KW & Lees KR (2003) Excitatory amino acid
antagonists for acute stroke. Cochrane Database Syst
Rev CD001244.
167 Birmingham K (2002) Future of neuroprotective drugs
in doubt. Nat Med 8,5.
168 Goldberg MP, Weiss JH, Pham PC & Choi DW (1987)
N-methyl-D-aspartate receptors mediate hypoxic
neuronal injury in cortical culture. J Pharmacol Exp
Ther 243, 784–791.
169 Goldberg MP & Choi DW (1993) Combined oxygen
and glucose deprivation in cortical cell culture: cal-
cium-dependent and calcium-independent mechanisms
of neuronal injury. J Neurosci 13, 3510–3524.

170 Nadler MJ, Hermosura MC, Inabe K, Perraud AL,
Zhu Q, Stokes AJ, Kurosaki T, Kinet JP, Penner R,
Scharenberg AM & Fleig A (2001) LTRPC7 is a Mg.
ATP-regulated divalent cation channel required for cell
viability. Nature 411, 590–595.
171 Ma HT, Venkatachalam K, Li HS, Montell C, Kurosa-
ki T, Patterson RL & Gill DL (2001) Assessment of
the role of the inositol 1,4,5-trisphosphate receptor in
the activation of transient receptor potential channels
and store-operated Ca
2+
entry channels. J Biol Chem
276, 18888–18896.
172 Maruyama T, Kanaji T, Nakade S, Kanno T &
Mikoshiba K (1997) 2APB, 2-aminoethoxydiphenyl
borate, a membrane-penetrable modulator of
Ins(1,4,5)P3-induced Ca
2+
release. J Biochem (Tokyo)
122, 498–505.
173 Bilmen JG, Wootton LL, Godfrey RE, Smart OS &
Michelangeli F (2002) Inhibition of SERCA Ca
2+
pumps by 2-aminoethoxydiphenyl borate (2-APB).
2-APB reduces both Ca
2+
binding and phosphoryl
transfer from ATP, by interfering with the pathway
leading to the Ca
2+

-binding sites. Eur J Biochem 269,
3678–3687.
174 Wang Y, Deshpande M & Payne R (2002) 2-Ami-
noethoxydiphenyl borate inhibits phototransduction
and blocks voltage-gated potassium channels in Limu-
lus ventral photoreceptors. Cell Calcium 32, 209–216.
175 Harks EG, Camina JP, Peters PH, Ypey DL, Scheenen
WJ, Van Zoelen EJ & Theuvenet AP (2003) Besides
affecting intracellular calcium signaling, 2-APB reversi-
bly blocks gap junctional coupling in confluent mono-
layers, thereby allowing measurement of single-cell
membrane currents in undissociated cells. FASEB J 17,
941–943.
176 Kwan CY & Putney JW Jr (1990) Uptake and intracel-
lular sequestration of divalent cations in resting and
methacholine-stimulated mouse lacrimal acinar cells.
Dissociation by Sr
2+
and Ba
2+
of agonist-stimulated
divalent cation entry from the refilling of the agonist-
sensitive intracellular pool. J Biol Chem 265, 678–684.
177 Nelson MT, French RJ & Krueger BK (1984) Voltage-
dependent calcium channels from brain incorporated
into planar lipid bilayers. Nature 308, 77–80.
178 Davidson TJ, Harel S, Arboleda VA, Prunell GF,
Shelanski ML, Greene LA & Troy CM (2004) Highly
efficient small interfering RNA delivery to primary
mammalian neurons induces MicroRNA-like effects

before mRNA degradation. J Neurosci 24, 10040–
10046.
179 Derossi D, Chassaing G & Prochiantz A (1998) Trojan
peptides: the penetratin system for intracellular deliv-
ery. Trends Cell Biol 8, 84–87.
180 Nicholls DG & Ward MW (2000) Mitochondrial mem-
brane potential and neuronal glutamate excitotoxicity:
mortality and millivolts. Trends Neurosci 23, 166–174.
C. Chinopoulos and V. Adam-Vizi Ca
2+
, mitochondria, ROS in neuronal disease
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 447
181 Vesce S, Kirk L & Nicholls DG (2004) Relationships
between superoxide levels and delayed calcium deregu-
lation in cultured cerebellar granule cells exposed con-
tinuously to glutamate. J Neurochem 90, 683–693.
182 Setsukinai K, Urano Y, Kakinuma K, Majima HJ &
Nagano T (2003) Development of novel fluorescence
probes that can reliably detect reactive oxygen species
and distinguish specific species. J Biol Chem 278, 3170–
3175.
183 Takezawa R, Schmitz C, Demeuse P, Scharenberg
AM, Penner R & Fleig A (2004) Receptor-mediated
regulation of the TRPM7 channel through its endogen-
ous protein kinase domain. Proc Natl Acad Sci USA
101, 6009–6014.
184 Xiong ZG, Zhu XM, Chu XP, Minami M, Hey J, Wei
WL, Macdonald JF, Wemmie JA, Price MP, Welsh
MJ & Simon RP (2004) Neuroprotection in ischemia;
blocking calcium-permeable Acid-sensing ion channels.

Cell 118, 687–698.
185 Arundine M & Tymianski M (2003) Molecular
mechanisms of calcium-dependent neurodegeneration
in excitotoxicity. Cell Calcium 34, 325–337.
186 Kristian T, Katsura K, Gido G & Siesjo BK (1994)
The influence of pH on cellular calcium influx during
ischemia. Brain Res 641, 295–302.
187 Ekholm A, Kristian T & Siesjo BK (1995) Influence of
hyperglycemia and of hypercapnia on cellular calcium
transients during reversible brain ischemia. Exp Brain
Res 104, 462–466.
188 Silver IA & Erecinska M (1992) Ion homeostasis in rat
brain in vivo: intra- and extracellular [Ca
2+
] and [H
+
]in
the hippocampus during recovery from short-term, tran-
sient ischemia. J Cereb Blood Flow Metab 12, 759–772.
189 Harris RJ, Symon L, Branston NM & Bayhan M
(1981) Changes in extracellular calcium activity in cere-
bral ischaemia. J Cereb Blood Flow Metab 1, 203–209.
190 Nilsson P, Laursen H, Hillered L & Hansen AJ (1996)
Calcium movements in traumatic brain injury: the role
of glutamate receptor-operated ion channels. J Cereb
Blood Flow Metab 16, 262–270.
191 Zhang ET, Hansen AJ, Wieloch T & Lauritzen M
(1990) Influence of MK-801 on brain extracellular cal-
cium and potassium activities in severe hypoglycemia.
J Cereb Blood Flow Metab 10, 136–139.

192 Hansen AJ & Zeuthen T (1981) Extracellular ion con-
centrations during spreading depression and ischemia
in the rat brain cortex. Acta Physiol Scand 113, 437–
445.
193 Martell A & Smith R (1977) Critical Stability Con-
stants. Plenum, New York.
194 Immke DC & Mccleskey EW (2001) Lactate enhances
the acid-sensing Na
+
channel on ischemia-sensing neu-
rons. Nat Neurosci 4, 869–870.
195 Zimmerman AN & Hulsmann WC (1966) Paradoxical
influence of calcium ions on the permeability of the cell
membranes of the isolated rat heart. Nature 211, 646–
647.
196 Duncan CJ & Morton JW (1996) Membrane damage
and the Ca
2+
-paradox in the perfused rat kidney. Kid-
ney Int 49, 639–646.
197 Soza M, Karpati G & Carpenter S (1986) Calcium
paradox in skeletal muscles: physiologic and micro-
scopic observations. Muscle Nerve 9, 222–232.
198 Okuda M, Lee HC, Chance B & Kumar C (1992)
Depletion and repletion of Ca
2+
in the perfused rat
liver. J Laboratory Clin Med 120, 57–66.
199 Young W (1986) Ca paradox in neural injury: a hypo-
thesis. Cent Nerv Syst Trauma 3, 235–251.

200 Siemkowicz E & Hansen AJ (1981) Brain extracellular
ion composition and EEG activity following 10 min-
utes ischemia in normo- and hyperglycemic rats. Stroke
12, 236–240.
201 Schaller B & Graf R (2004) Cerebral ischemia and
reperfusion: the pathophysiologic concept as a basis
for clinical therapy. J Cereb Blood Flow Metab 24,
351–371.
202 Ruigrok TJ, Boink AB & Zimmerman AN (1976)
Influence of ATP or oxygen plus substrate on occur-
rence of the calcium paradox. Recent Adv Stud Cardiac
Struct Metab 11, 565–569.
203 Xiong Z, Lu W & Macdonald JF (1997) Extracellular
calcium sensed by a novel cation channel in hippocampal
neurons. Proc Natl Acad Sci USA 94, 7012–7017.
204 Smith SM, Bergsman JB, Harata NC, Scheller RH &
Tsien RW (2004) Recordings from single neocortical
nerve terminals reveal a nonselective cation channel
activated by decreases in extracellular calcium. Neuron
41, 243–256.
205 Formenti ASA, Arrigoni E & Martina M (2001)
Changes in extracellular Ca
2+
can affect the pattern of
discharge in rat thalamic neurons. J Physiol 535, 33–
45.
206 Undem BJ, Oh EJ, Lancaster E & Weinreich D (2003)
Effect of extracellular calcium on excitability of guinea
pig airway vagal afferent nerves. J Neurophysiol 89,
1196–1204.

207 Mubagwa K, Stengl M & Flameng W (1997) Extracel-
lular divalent cations block a cation non-selective con-
ductance unrelated to calcium channels in rat cardiac
muscle. J Physiol 502 (2), 235–247.
208 Frankenhaeuser B & Hodgkin AL (1957) The action
of calcium on the electrical properties of squid axons.
J Physiol 137, 218–244.
209 Hoth M & Penner R (1993) Calcium release-activated
calcium current in rat mast cells. J Physiol 465, 359–
386.
210 Su Z, Shoemaker RL, Marchase RB & Blalock JE
(2004) Ca
2+
modulation of Ca
2+
release-activated
Ca
2+
channels is responsible for the inactivation of its
monovalent cation current. Biophys J 86, 805–814.
Ca
2+
, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
448 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS
211 Kostiuk PG, Mironov SL & Shuba I (1983) [2 selective
filters in the calcium channel of the somatic membrane
of mollusk neurons]. Neirofiziologiia 15, 420–427.
212 Almers W, Mccleskey EW & Palade PT (1984) A non-
selective cation conductance in frog muscle membrane
blocked by micromolar external calcium ions. J Physiol

353, 565–583.
213 Fukushima Y & Hagiwara S (1985) Currents carried by
monovalent cations through calcium channels in mouse
neoplastic B lymphocytes. J Physiol 358, 255–284.
214 Hess P, Lansman JB & Tsien RW (1986) Calcium
channel selectivity for divalent and monovalent cations.
Voltage and concentration dependence of single chan-
nel current in ventricular heart cells. J Gen Physiol 88,
293–319.
215 Polo-Parada L & Korn SJ (1997) Block of N-type cal-
cium channels in chick sensory neurons by external
sodium. J Gen Physiol 109, 693–702.
216 Armstrong CM & Cota G (1999) Calcium block of
Na
+
channels and its effect on closing rate. Proc Natl
Acad Sci USA 96, 4154–4157.
217 Armstrong CM (1999) Distinguishing surface effects of
calcium ion from pore-occupancy effects in Na
+
chan-
nels. Proc Natl Acad Sci USA 96, 4158–4163.
218 Johnson JP Jr, Balser JR & Bennett PB (2001) A novel
extracellular calcium sensing mechanism in voltage-
gated potassium ion channels. J Neurosci 21, 4143–
4153.
219 Xiong ZG & Macdonald JF (1999) Sensing of extracel-
lular calcium by neurones. Can J Physiol Pharmacol
77, 715–721.
220 Brown EM & Macleod RJ (2001) Extracellular calcium

sensing and extracellular calcium signaling. Physiol Rev
81, 239–297.
221 Devries SH & Schwartz EA (1992) Hemi-gap-junction
channels in solitary horizontal cells of the catfish
retina. J Physiol 445, 201–230.
222 Ebihara L, Liu X & Pal JD (2003) Effect of external
magnesium and calcium on human connexin 46 hemi-
channels. Biophys J 84, 277–286.
223 Kubo Y, Miyashita T & Murata Y (1998) Structural
basis for a Ca
2+
-sensing function of the metabotropic
glutamate receptors. Science 279, 1722–1725.
224 Vassilev PM, Mitchel J, Vassilev M, Kanazirska M &
Brown EM (1997) Assessment of frequency-dependent
alterations in the level of extracellular Ca
2+
in the syn-
aptic cleft. Biophys J 72, 2103–2116.
225 Wise A, Green A, Main MJ, Wilson R, Fraser N &
Marshall FH (1999) Calcium sensing properties of the
GABA(B) receptor. Neuropharmacology 38, 1647–1656.
226 Moran MM, Xu H & Clapham DE (2004) TRP ion
channels in the nervous system. Curr Opin Neurobiol
14, 362–369.
227 Gunthorpe MJ, Benham CD, Randall A & Davis JB
(2002) The diversity in the vanilloid (TRPV) receptor
family of ion channels. Trends Pharmacol Sci 23, 183–
191.
228 Liman ER, Corey DP & Dulac C (1999) TRP2: a can-

didate transduction channel for mammalian pheromone
sensory signaling. Proc Natl Acad Sci USA 96, 5791–
5796.
229 Stowers L, Holy TE, Meister M, Dulac C & Koentges
G (2002) Loss of sex discrimination and male-male
aggression in mice deficient for TRP2. Science 295,
1493–1500.
230 Greka A, Navarro B, Oancea E, Duggan A & Clap-
ham DE (2003) TRPC5 is a regulator of hippocampal
neurite length and growth cone morphology. Nat Neu-
rosci 6, 837–845.
231 Li HS, Xu XZ & Montell C (1999) Activation of a
TRPC3-dependent cation current through the neuro-
trophin BDNF. Neuron 24, 261–273.
232 Sun M, Goldin E, Stahl S, Falardeau JL, Kennedy JC,
Acierno JS Jr, Bove C, Kaneski CR, Nagle J, Bromley
MC, Colman M, Schiffmann R & Slaugenhaupt SA
(2000) Mucolipidosis type IV is caused by mutations in
a gene encoding a novel transient receptor potential
channel. Hum Mol Genet 9, 2471–2478.
233 Alessandri-Haber N, Dina OA, Yeh JJ, Parada CA,
Reichling DB & Levine JD (2004) Transient receptor
potential vanilloid 4 is essential in chemotherapy-
induced neuropathic pain in the rat. J Neurosci 24,
4444–4452.
234 Fonfria E, Marshall IC, Benham CD, Boyfield I,
Brown JD, Hill K, Hughes JP, Skaper SD &
Mcnulty S (2004) TRPM2 channel opening in
response to oxidative stress is dependent on activa-
tion of poly (ADP-ribose) polymerase. Br J Pharma-

col 143, 186–192.
235 Balzer M, Lintschinger B & Groschner K (1999)
Evidence for a role of Trp proteins in the oxidative
stress-induced membrane conductances of porcine
aortic endothelial cells. Cardiovasc Res 42, 543–
549.
236 Wehage E, Eisfeld J, Heiner I, Jungling E, Zitt C &
Luckhoff A (2002) Activation of the Cation Channel
Long Transient Receptor Potential Channel 2
(LTRPC2) by Hydrogen Peroxide. A splice variant
reveals a mode of activation independent of ADP-
ribose. J Biol Chem 277, 23150–23156.
237 Kraft R, Grimm C, Grosse K, Hoffmann A, Sauerb-
ruch S, Kettenmann H, Schultz G & Harteneck C
(2004) Hydrogen peroxide and ADP-ribose induce
TRPM2-mediated calcium influx and cation currents
in microglia. Am J Physiol Cell Physiol 286, C129–
C137.
238 Hara Y, Wakamori M, Ishii M, Maeno E, Nishida M,
Yoshida T, Yamada H, Shimizu S, Mori E, Kudoh J,
Shimizu N, Kurose H, Okada Y, Imoto K & Mori Y
(2002) LTRPC2 Ca
2+
-permeable channel activated by
C. Chinopoulos and V. Adam-Vizi Ca
2+
, mitochondria, ROS in neuronal disease
FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS 449
changes in redox status confers susceptibility to cell
death. Mol Cell 9, 163–173.

239 Hardie RC, Raghu P, Moore S, Juusola M, Baines RA
& Sweeney ST (2001) Calcium influx via TRP channels
is required to maintain PIP2 levels in Drosophila
photoreceptors. Neuron 30, 149–159.
240 Lintschinger B, Balzer-Geldsetzer M, Baskaran T, Gra-
ier WF, Romanin C, Zhu MX & Groschner K (2000)
Coassembly of Trp1 and Trp3 proteins generates dia-
cylglycerol- and Ca
2+
-sensitive cation channels. J Biol
Chem 275, 27799–27805.
241 Jung S, Strotmann R, Schultz G & Plant TD (2002)
TRPC6 is a candidate channel involved in receptor-
stimulated cation currents in A7r5 smooth muscle cells.
Am J Physiol Cell Physiol 282, C347–C359.
242 Okada T, Inoue R, Yamazaki K, Maeda A, Kurosaki
T, Yamakuni T, Tanaka I, Shimizu S, Ikenaka K,
Imoto K & Mori Y (1999) Molecular and functional
characterization of a novel mouse transient receptor
potential protein homologue TRP7. Ca
2+
-permeable
cation channel that is constitutively activated and
enhanced by stimulation of G protein-coupled receptor.
J Biol Chem 274, 27359–27370.
243 Shi J, Mori E, Mori Y, Mori M, Li J, Ito Y & Inoue
R (2004) Multiple regulation by calcium of murine
homologues of transient receptor potential proteins
TRPC6 and TRPC7 expressed in HEK293 cell. J Phy-
siol 561, 415–432.

244 Perraud AL, Fleig A, Dunn CA, Bagley LA, Launay P,
Schmitz C, Stokes AJ, Zhu Q, Bessman MJ, Penner R,
Kinet JP & Scharenberg AM (2001) ADP-ribose gating
of the calcium-permeable LTRPC2 channel revealed by
Nudix motif homology. Nature 411, 595–599.
245 Heiner I, Eisfeld J, Halaszovich CR, Wehage E, Jun-
gling E, Zitt C & Luckhoff A (2003) Expression profile
of the transient receptor potential (TRP) family in neu-
trophil granulocytes: evidence for currents through
LTRPC2 induced by ADP-ribose and NAD. Biochem J
371, 1045–1053.
246 Kolisek M, Beck A, Fleig A & Penner R (2005) Cyclic
ADP-ribose and hydrogen peroxide synergize with
ADP-ribose in the activation of TRPM2 channels. Mol
Cell 18, 61–69.
247 Harteneck C (2005) Function and pharmacology of
TRPM cation channels. Naunyn Schmiedebergs Arch
Pharmacol 371, 307–314.
248 Perraud AL, Takanishi CL, Shen B, Kang S, Smith
MK, Schmitz C, Knowles HM, Ferraris D, Li W,
Zhang J, Stoddard BL & Scharenberg AM (2004)
Accumulation of free ADP-ribose from mitochondria
mediates oxidative stress-induced gating of TRPM2
cation channels. J Biol Chem 280, 6138–6148.
249 Ziegler M (2000) New functions of a long-known mole-
cule. Emerging roles of NAD in cellular signaling. Eur
J Biochem 267, 1550–1564.
250 Strubing C, Krapivinsky G, Krapivinsky L & Clapham
DE (2001) TRPC1 and TRPC5 form a novel cation
channel in mammalian brain. Neuron 29, 645–655.

251 Strubing C, Krapivinsky G, Krapivinsky L & Clapham
DE (2003) Formation of novel TRPC channels by
complex subunit interactions in embryonic brain.
J Biol Chem 278, 39014–39019.
252 Toescu EC (2004) Hypoxia sensing and pathways of
cytosolic Ca
2+
increases. Cell Calcium 36 , 187–199.
253 Montell C (2005) The TRP superfamily of cation chan-
nels. Sci STKE 2005 re3.
254 Montell C (2001) Physiology, phylogeny, and functions
of the TRP superfamily of cation channels. Sci STKE
2001, RE1.
255 Chandrasekaran K, Mehrabian Z, Li XL & Hassel B
(2004) RNase-L regulates the stability of mitochondrial
DNA-encoded mRNAs in mouse embryo fibroblasts.
Biochem Biophys Res Commun 325, 18–23.
256 Mehrabian Z, Liu LI, Fiskum G, Rapoport SI &
Chandrasekaran K (2005) Regulation of mitochondrial
gene expression by energy demand in neural cells.
J Neurochem 93, 850–860.
257 Chinopoulos C, Tretter L, Rozsa A & Adam-Vizi V
(2000) Exacerbated responses to oxidative stress by
an Na
+
load in isolated nerve terminals: the role of
ATP depletion and rise of [Ca
2+
] (i). J Neurosci 20,
2094–2103.

258 Yu SP, Yeh CH, Sensi SL, Gwag BJ, Canzoniero LM,
Farhangrazi ZS, Ying HS, Tian M, Dugan LL & Choi
DW (1997) Mediation of neuronal apoptosis by
enhancement of outward potassium current. Science
278, 114–117.
259 Yu SP, Yeh C, Strasser U, Tian M & Choi DW (1999)
NMDA receptor-mediated K
+
efflux and neuronal
apoptosis. Science 284, 336–339.
260 Yu SP (2003) Regulation and critical role of potas-
sium homeostasis in apoptosis. Prog Neurobiol 70,
363–386.
261 Montell C, Birnbaumer L & Flockerzi V (2002) The
TRP channels, a remarkably functional family. Cell
108, 595–598.
262 Nilius B, Voets T & Peters J (2005) TRP channels in
disease. Sci STKE 2005, re8.
263 Inoue R, Hanano T, Shi J, Mori Y & Ito Y (2003)
Transient receptor potential protein as a novel non-
voltage-gated Ca
2+
entry channel involved in diverse
pathophysiological functions. J Pharmacol Sci 91, 271–
276.
264 Wissenbach U, Niemeyer BA & Flockerzi V (2004)
TRP channels as potential drug targets. Biol Cell 96,
47–54.
265 Shuttleworth CW, Brennan AM & Connor JA (2003)
NAD(P)H fluorescence imaging of postsynaptic neuro-

nal activation in murine hippocampal slices. J Neurosci
23, 3196–3208.
Ca
2+
, mitochondria, ROS in neuronal disease C. Chinopoulos and V. Adam-Vizi
450 FEBS Journal 273 (2006) 433–450 ª 2006 The Authors Journal compilation ª 2006 FEBS

×