ArticlePDF Available

Kirichok Y, Krapivinsky G, Clapham DEThe mitochondrial calcium uniporter is a highly selective ion channel. Nature 427:360-364

Authors:

Abstract and Figures

During intracellular Ca2+ signalling mitochondria accumulate significant amounts of Ca2+ from the cytosol. Mitochondrial Ca2+ uptake controls the rate of energy production, shapes the amplitude and spatio-temporal patterns of intracellular Ca2+ signals, and is instrumental to cell death. This Ca2+ uptake is undertaken by the mitochondrial Ca2+ uniporter (MCU) located in the organelle's inner membrane. The uniporter passes Ca2+ down the electrochemical gradient maintained across this membrane without direct coupling to ATP hydrolysis or transport of other ions. Carriers are characterized by turnover numbers that are typically 1,000-fold lower than ion channels, and until now it has been unclear whether the MCU is a carrier or a channel. By patch-clamping the inner mitochondrial membrane, we identified a previously unknown Ca2+-selective ion channel sensitive to inhibitors of mitochondrial Ca2+ uptake. Our data indicate that this unique channel binds Ca2+ with extremely high affinity (dissociation constant < or =2 nM), enabling high Ca2+ selectivity despite relatively low cytoplasmic Ca2+ concentrations. The channel is inwardly rectifying, making it especially effective for Ca2+ uptake into energized mitochondria. Thus, we conclude that the properties of the current mediated by this novel channel are those of the MCU.
Content may be subject to copyright.
J. Biol. Chem. 273, 23722–23728 (1998).
7. Graeler, M. & Goetzl, E. J. Activation-regulated expression and chemotactic function of sphingosine
1-phosphate receptors in mouse splenic T cells. FASEB J. 16, 1874–1878 (2002).
8. Liu, Y. et al. Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for
vascular maturation. J. Clin. Invest. 106, 951–961 (2000).
9. Chaffin, K. E. & Perlmutter, R. M. A pertussis toxin sensitive process controls thymocyte emigration.
Eur. J. Immunol. 21, 2565–2573 (1991).
10. Rosen, H., Alfonso, C., Surh, C. D. & McHeyzer-Williams, M. G. Rapid induction of medullary
thymocyte phenotypic maturation and egress inhibition by nanomolar sphingosine 1-phosphate
receptor agonist. Proc. Natl Acad. Sci. USA 100, 10907–10912 (2003).
11. Lucas, B., Vasseur, F. & Penit, C. Production, selection, and maturation of thymocytes with high
surface density of TCR. J. Immunol. 153, 53–62 (1994).
12. Gabor, M. J., Godfrey, D. I. & Scollay, R. Recent thymic emigrants are distinct from most medullary
thymocytes. Eur. J. Immunol. 27, 2010–2015 (1997).
13. Chu, P. et al. Systematic identification of regulatory proteins critical for T-cell activation. J. Biol. 2,
211–2116 (2003).
14. Parrot, D. M. V. & de Sousa, M. Thymus-dependent and thymus-independent populations: origin,
migratory patterns and lifespan. Clin. Exp. Immunol. 8, 663–673 (1971).
15. Hall, J. G. & Morris, B. The immediate effect of antigens on the cell output of a lymph node. Br. J. Exp.
Pathol. 46, 450–454 (1965).
16. Sprent, J., Miller, J. F. A. P. & Mitchell, G. F. Antigen-induced selective recruitment of circulating
lymphocytes. Cell. Immunol. 2, 171–181 (1971).
17. Hla, T. Sphingosine 1-phosphate receptors. Prostaglandins Lipid Mediat. 64, 135–142 (2001).
18. Edsall, L. C. & Spiegel, S. Enzymatic measurement of sphingosine 1-phosphate. Anal. Biochem. 272,
80–86 (1999).
19. Murata, N. et al. Interaction of sphingosine 1-phosphate with plasma components, including
lipoproteins, regulates the lipid receptor-mediated actions. Biochem. J. 352, 809–815 (2000).
20. Caligan, T. B. et al. A high-performance liquid chromatographic method to measure sphingosine
1-phosphate and related compounds from sphingosine kinase assays and other biological samples.
Anal. Biochem. 281, 36–44 (2000).
21. Graeler, M., Shankar, G. & Goetzl, E. J. Cutting edge: suppression of T cell chemotaxis by sphingosine
1-phosphate. J. Immunol. 169, 4084–4087 (2002).
22. Idzko, M. et al. Sphingosine 1-phosphate induces chemotaxis of immature and modulates cytokine-
release in mature human dendritic cells for emergence of Th2 immune responses. FASEB J. 16,
625–627 (2002).
23. Hargreaves, D. C. et al. A coordinated change in chemokine responsiveness guides plasma cell
movements. J. Exp. Med. 194, 45–56 (2001).
24. Reif, K. et al. Balanced responsiveness to chemoattractants from adjacent zones determines B-cell
position. Nature 416, 94–99 (2002).
Supplementary Information accompanies the paper on www.nature.com/nature.
Acknowledgements We are grateful to C. Low for technical assistance; C. McArthur and S. Jiang
for cell sorting; R. Albert at Novartis Institutes for BioMedical Research for synthesis of
FTY720-P; and T. Okada, C. Allen and S. Watson for helpful discussions. M.M. is supported by
the Pfizer Postdoctoral Fellowship in Immunology and Rheumatology and the Rosalind Russell
Medical Research Center for Arthritis at University of California, San Francisco; J.G.C. is a
Packard fellow and an HHMI assistant investigator. This work was supported in part by grants
from the National Institutes of Health.
Competing interests statement The authors declare that they have no competing financial
interests.
Correspondence and requests for materials should be addressed to J.G.C. (cyster@itsa.ucsf.edu).
..............................................................
The mitochondrial calcium uniporter
is a highly selective ion channel
Yuriy Kirichok, Grigory Krapivinsky & David E. Clapham
Howard Hughes Medical Institute, Department of Cardiovascular Research,
Childrens Hospital, and Department of Neurobiology, Harvard Medical School,
320 Longwood Avenue, Boston, Massachusetts 02115, USA
.............................................................................................................................................................................
During intracellular Ca
21
signalling mitochondria accumulate
significant amounts of Ca
21
from the cytosol
1,2
. Mitochondrial
Ca
21
uptake controls the rate of energy production
1,3,4
, shapes
the amplitude and spatio-temporal patterns of intracellular Ca
21
signals
1,5–8
, and is instrumental to cell death
9,10
. This Ca
21
uptake
is undertaken by the mitochondrial Ca
21
uniporter (MCU)
located in the organelle’s inner membrane
11,12
. The uniporter
passes Ca
21
down the electrochemical gradient maintained
across this membrane without direct coupling to ATP hydrolysis
or transport of other ions
11
. Carriers are characterized by turn-
over numbers that are typically 1,000-fold lower than ion chan-
nels, and until now it has been unclear whether the MCU is a
carrier or a channel
13
. By patch-clamping the inner mitochon-
drial membrane, we identified a previously unknown Ca
21
-
selective ion channel sensitive to inhibitors of mitochondrial
Ca
21
uptake. Our data indicate that this unique channel binds
Ca
21
with extremely high affinity (dissociation constant #2 nM),
enabling high Ca
21
selectivity despite relatively low cytoplasmic
Ca
21
concentrations. The channel is inwardly rectifying, making
it especially effective for Ca
21
uptake into energized mitochon-
dria. Thus, we conclude that the properties of the current
mediated by this novel channel are those of the MCU.
Since its discovery in 1961 (ref. 14), mitochondrial Ca
2þ
uptake
has been measured in suspensions of isolated mitochondria. This
approach does not reliably control ion concentrations or voltage
gradients across the inner mitochondrial membrane, resulting in
inaccuracies in experimental results
11,12
. The patch-clamp method
solves these problems but its implementation in mitochondria is a
severe technical challenge. Here we patch-clamp single mitoplasts
(2–5-
m
m vesicles of inner mitochondrial membrane) isolated from
COS-7 cells (Fig. 1a, b), to measure directly whole-mitoplast and
single-channel Ca
2þ
currents.
In the whole-mitoplast configuration, voltage ramps from
2160 mV (potential across the inner membrane of energized
mitochondria) to þ80 mV elicited an inwardly rectifying current
that gradually increased as free Ca
2þ
concentration at the cytosolic
surface of the inner membrane ([Ca
2þ
]
c
) was varied from 20
m
Mto
100
m
M. These concentrations are comparable to [Ca
2þ
] in micro-
domains near endoplasmic/sarcoplasmic reticulum Ca
2þ
release or
plasma membrane Ca
2þ
channels, sensed by neighbouring mito-
chondria
1,2,15,16
. At 100
m
M [Ca
2þ
]
c
, the inwardly rectifying current
reached amplitudes of 20–30 pA (Fig. 1c). As mitoplasts are small,
this amplitude corresponds to a significant current density
(55 ^ 19 pA pF
21
at 2160 mV, n ¼ 4). To examine the level at
which the current saturates, [Ca
2þ
]
c
was elevated from 20
m
Mto
105 mM (Fig. 1c–e). Notably, the Ca
2þ
carrying capacity of the
current was enormous, reaching half-saturation at approximately
20 mM [Ca
2þ
]
c
. Elimination of Na
þ
in the bath did not reduce the
current, suggesting that Ca
2þ
was the primary charge carrier (not
shown). We refer to the mitoplast inwardly rectifying Ca
2þ
current
as I
MiCa
(for mitochondrial Ca
2þ
current).
I
MiCa
elicited by voltage steps rapidly inactivated to a plateau level
(Fig. 1f). The time-independent plateau was the major component
of the current with the transient component averaging 21 ^ 14% of
the net peak current (n ¼ 16). I
MiCa
amplitude was not noticeably
altered when pipette (intramitoplast) [Ca
2þ
]wasvariedfrom
,10 nM to about 10
m
M (no EGTA or EDTA). Thus, unlike many
Ca
2þ
-selective currents, I
MiCa
lacked Ca
2þ
-dependent inactivation
at energized mitochondrial potentials (approximately 2160 mV).
Similar to MCU-mediated mitochondrial Ca
2þ
uptake
11,17–19
,
Ruthenium 360 (Ru360) inhibited I
MiCa
in a dose-dependent
manner (half-maximal inhibitory concentration (IC
50
), 2 nM)
and was a more potent inhibitor than ruthenium red (RuR, IC
50
9 nM; Fig. 2).
RuR-sensitive (200 nM) I
MiCa
conducted Ca
2þ
and Sr
2þ
equally,
whereas Mg
2þ
was not measurably permeant (Fig. 3a). The relative
divalent ion conductance was Ca
2þ
< Sr
2þ
.. Mn
2þ
< Ba
2þ
(Fig. 3b), identical to the relative divalent ion permeability of the
MCU
13,20–22
. I
MiCa
currents carried by any of these four permeant
divalent ions were substantially reduced by low concentrations of
other permeant divalent ions, indicating competition for binding
at the selectivity site. The monovalent ions K
þ
and Na
þ
do not
contribute to I
MiCa
in the presence of [Ca
2þ
]
c
(Fig. 1d; see also
Supplementary Fig. 1). We refer to MiCa (mitochondrial calcium)
as the highly Ca
2þ
-selective conductance producing large current
letters to nature
NATURE | VOL 427 | 22 JANUARY 2004 | www.nature.com/nature360
© 2004
Nature
Publishing
Group
densities in mitochondria, consistent with an ion channel.
Ion channel Ca
2þ
selectivity is the result of high-affinity Ca
2þ
binding to the permeation pathway, and removal of external Ca
2þ
enables these ion channels to conduct monovalent ions
23–26
. Indeed,
in low-divalent (5 mM EGTA/5 mM EDTA) solution, we recorded a
RuR- and Ru360-sensitive linear Na
þ
current (I
MiNa
). Interestingly,
I
MiNa
was relatively impermeant to K
þ
(Fig. 3c, top row). Increasing
[Ca
2þ
]
c
to 10 nM partially restored Ca
2þ
selectivity, whereas
100 nM led to complete inhibition of the Na
þ
conductance
(Fig. 3c, bottom left panel; estimated Ca
2þ
-binding equilibrium
constant #2 nM; see Methods). Although MiCa does not conduct
Mg
2þ
, in the absence of other divalent ions MiCa (I
MiNa
) binds
Mg
2þ
(Fig. 3c, bottom right panel) with an estimated binding
constant #250 nM (Methods). In 0.2 mM [Ca
2þ
]
c
,0.5mM
[Mg
2þ
]
c
reduced I
MiCa
to 41 ^ 8% of its conductance in nominally
Mg
2þ
-free solution (not shown). Thus, I
MiCa
will be reduced by
local cytosolic [Mg
2þ
]
c
, in agreement with data on the MCU
27,28
.
On the basis of its high sensitivity to RuR, Ru360 and Ca
2þ
,we
conclude that I
MiNa
is the monovalent current in divalent-free
conditions.
Single MiCa channel activity was recorded from inside-out
mitoplast patches. Under the ionic conditions used for the whole-
mitoplast configuration, 3–7 channels were active in each patch and
they closely mimicked the inward-rectifying I
MiCa
current–voltage
relationship (Fig. 4a). This density of single channels agrees with the
high I
MiCa
current density in whole mitoplasts. Single-channel
current amplitudes in 105 mM [Sr
2þ
]
c
(pipette) conditions were
similar to those in 105 mM [Ca
2þ
]
c
conditions; however, in agree-
ment with whole-mitoplast I
MiCa
selectivity, 105 mM [Mg
2þ
]
c
currents were not observed. RuR or Ru360 (200 nM; pipette)
rapidly blocked inward single-channel activity on stepping to
2180 mV (Fig. 4e). Block by 200 nM RuR and Ru360 was relieved
on returning to potentials above 0 mV (not shown). Thus, RuR and
Ru360 were ineffective in blocking i
MiCa
outward current (where i
indicates unitary current).
In symmetrical 105 mM [Ca
2þ
] conditions, outward single-
channel openings at positive voltages (always correlated with inward
openings at negative voltages) rapidly flickered between open and
closed states (Fig. 4a). Substitution of bath (matrix) Ca
2þ
by Mg
2þ
abolished these outward channels (Fig. 4b), whereas Sr
2þ
-mediated
outward currents were comparable to those in Ca
2þ
. In symmetrical
150 mM Na-gluconate low-divalent solution, single i
MiNa
Na
þ
channel currents had about tenfold higher conductance than
i
MiCa
, were selective for Na
þ
over K
þ
, exhibited multiple conduc-
tance states, and were blocked by 200 nM RuR. The rapid flickering
of outward single-channel currents observed in 105 mM Ca
2þ
were
not observed for the outward monovalent current. This suggests
that open channels are transiently blocked by outward-going Ca
2þ
Figure 1 Ca
2þ
current through the inner mitochondrial membrane. a, COS-7 cell
transfected with mitochondrial-targeted YFP. b, Transmitted and fluorescent images of a
mito-YFP-labelled mitoplast. Scale bar: 3
m
m. c, I
MiCa
elicited by voltage ramps. Negative
current flows from the cytoplasmic face of the inner mitochondrial membrane (bath) to the
interior of the mitoplast. Bath ¼ 0 mV. Inside mitoplast (pipette), Cs-gluconate solution;
bath, HEPES-Tris solution. d, I
MiCa
increases with bath [Ca
2þ
]
c
. Bath, Na-gluconate
solution. e, Peak amplitude of I
MiCa
at 2160 mV at varying [Ca
2þ
]
c
. Currents were
normalized to peak I
MiCa
in 105 mM [Ca
2þ
]
c
and fitted by I ¼ I
max
=ð1 þ
ðK
0:5
=½Ca
2þ
Þ
h
Þ; n ¼ 5. f, I
MiCa
in response to voltage steps (5 mM [Ca
2þ
]
c
Na-
gluconate solution); DV ¼ 20 mV.
letters to nature
NATURE | VOL 427 | 22 JANUARY 2004 | www.nature.com/nature 361
© 2004
Nature
Publishing
Group
ions. Indeed, the amplitude of ensemble-averaged Ca
2þ
currents
from multi-channel patches was about nine times less in the out-
ward than in the inward direction (Fig. 4c).
To determine single-channel kinetics from patches containing
only one active channel (Fig. 4d), smaller areas of membrane
were patched by using greatly reduced pipette tip diameters. In
symmetrical 105 mM CaCl
2
solution, the activity of single Ca
2þ
currents was time-independent at all voltages between 2200 and
þ140 mV. Single MiCa channels had multiple subconductance
states between 2.6 pS and 5.2 pS (2160 mV). Most importantly,
the open probability (P
open
) of the channel was about 99% at
2200 mV and declined to approximately 11% at 280 mV. This
dependence of P
open
on membrane potential accounts for much of
the inward rectification of the macroscopic I
MiCa
current over this
range of potentials (Supplementary Fig. 2).
In summary, a unique Ca
2þ
-selective channel of the inner
mitochondrial membrane accounts for the properties of the
MCU. MiCa is the only known functional intracellular Ca
2þ
-
selective channel (inositol-1,4,5-trisphosphate and ryanodine
Figure 2 RuR and Ru360 sensitivity. a, RuR (2–200 nM) inhibited I
MiCa
which did not fully
recover by 7 min after washout (red trace). The pipette (intramitoplast) contained Na-
gluconate solution whereas the HEPES-Tris bath solution contained 5 mM Ca
2þ
. b, Dose
response of I
MiCa
to RuR (n ¼ 5). c, I
MiCa
inhibition by the MCU blocker Ru360. I
MiCa
recovered 4 min after washout (red trace). The pipette was filled with Na-gluconate
solution (no EGTA/EDTA). d, Estimated dose–response curve of I
MiCa
in response to
Ru360. The time courses of inhibition were fitted by single exponential functions with
t ¼ 5 ^ 2s(n ¼ 4) at 2 nM and 270 ^ 160 ms (n ¼ 4) at 50 nM. Steady state
inhibition is estimated from fits for 2 and 5 nM.
Figure 3 I
MiCa
selectivity. a, I
MiCa
in 5 mM Ca
2þ
,Sr
2þ
,Mn
2þ
,Ba
2þ
(blue traces) or
Mg
2þ
(black traces). Recordings are from the same mitoplast in HEPES-Tris-based
solution, washed between applications with 1 mM EGTA/EDTA solution. Pipettes
contained Na-gluconate solution. b, Relative conductance of I
MiCa
to divalent cations.
Averaged peak current densities at 2160 mV (n ¼ 5; ^s.d.). c, I
MiCa
conducts Na
þ
in
the absence of divalent ions (I
MiNa
). Top left panel: I
MiNa
in Na-gluconate (black trace) and
K-gluconate (red trace) low-divalent (LD) solution. Top right panel: I
MiNa
Na
þ
(LD) current
was inhibited by RuR and Ru360. Bottom left panel: [Ca
2þ
]
c
at 10 nM and 100 nM
inhibited I
MiNa
. Bottom right panel: [Mg
2þ
]
c
at 1
m
M inhibited I
MiNa
.
letters to nature
NATURE | VOL 427 | 22 JANUARY 2004 | www.nature.com/nature362
© 2004
Nature
Publishing
Group
receptors are nonselective, but are Ca
2þ
-permeant). MiCa is Ca
2þ
-
selective even in low cytoplasmic [Ca
2þ
], it conducts Ca
2þ
prefer-
entially into mitochondria, has relative divalent conductances
Ca
2þ
< Sr
2þ
.. Mn
2þ
< Ba
2þ
; is blocked by nanomolar concen-
trations of RuR and Ru360, and can provide sufficient current
densities to explain mitochondrial uptake by the MCU.
There is one marked difference between our data and Ca
2þ
flux
studies using suspensions of isolated mitochondria. Here, in
105 mM [Ca
2þ
]
c
, I
MiCa
is close to saturation with a maximum
ion flux through the single MiCa molecule of approximately 5 £ 10
6
Ca
2þ
s
21
(half-activation constant, K
0.5
¼ 19 mM), significantly
higher than the approximately 2 £ 10
4
Ca
2þ
s
21
and K
0.5
of about
10
m
M estimated for MCU from suspensions of isolated mitochon-
dria
11,13
. This difference in flux estimates stems from the fact that in
mitochondrial suspensions, the rapid dissipation of the mitochon-
drial potential during Ca
2þ
entry
11,12
would appear to cause satura-
tion of Ca
2þ
flux at much lower [Ca
2þ
]
c
. At [Ca
2þ
]
c
above 100
m
M,
I
MiCa
current densities are so large that the potential across the inner
membrane can only be maintained by voltage clamp. On the basis of
an I
MiCa
density of 1,000–2,000 pA pF
21
, .90% open probability,
and 0.5–1 pA single-channel amplitude at ½Ca
2þ
c
¼ 105 mM and
V
m
¼ 2160 mV, MiCa channel density in the inner mitochondrial
membrane is approximately 10–40 channels per
m
m
2
, slightly lower
than voltage-gated Ca
2þ
channel density (about 100 per
m
m
2
) (ref.
29) in excitable cell membranes. Nonetheless, at micromolar con-
centrations of [Ca
2þ
]
c
, MiCa channels provide current densities
comparable to those of voltage-gated Ca
2þ
channels at millimolar
Ca
2þ
concentrations. This feat is accomplished by the large electro-
chemical gradient for Ca
2þ
across the inner membrane of energized
mitochondria and the unusually high probability of MiCa being in
the open state. Ca
2þ
-dependent inactivation, a negative feedback
mechanism for Ca
2þ
-permeant channels
29
, is surprisingly absent for
MiCa at normal potentials for energized mitochondria.
Na
þ
current through MiCa appears only at extremely low
concentrations of Ca
2þ
, suggesting that binding sites inside the
pore have an exceptionally high affinity (dissociation constant
#2 nM) for Ca
2þ
. This property guarantees its high selectivity for
cytosolic Ca
2þ
where monovalent cations outnumber Ca
2þ
ions by
10
3
–10
6
-fold. MiCa is impermeant to Mg
2þ
and K
þ
, thus prevent-
ing mitochondrial depolarization by these abundant ions. In short
the high Ca
2þ
selectivity of MiCa results in mitochondrial Ca
2þ
accumulation with the least dissipation of mitochondrial potential.
On the basis of the properties demonstrated in our experiments, we
conclude that the mitochondrial Ca
2þ
uniporter is the Ca
2þ
-
selective ion channel MiCa. A
Methods
Preparation of the mitoplasts
COS-7 cells were homogenized by a Teflon pestle in a glass grinder with 250 mM sucrose,
5 mM HEPES and 1 mM EGTA (pH 7.2 with KOH). Mitochondria were isolated from the
lysed COS-7 cell by differential centrifugation in the same solution
30
. To obtain mitoplasts,
mitochondria were subjected to osmotic shock for 5 min in hypotonic solution containing
5 mM sucrose, 5 mM HEPES and 1 mM EGTA (pH 7.2 with KOH). Mitoplasts were
sedimented from this solution by centrifugation at 3,920g
max
for 5 min and subsequently
re-suspended in 750 mM KCl, 100 mM HEPES and 1 mM EGTA (pH 7.2 with KOH).
Mitoplast isolation was carried out at 2 8C and mitoplasts stored on ice. Just before
experiments, 1–3
m
l of the mitoplast suspension was added to the experimental solution
containing 150 mM Na-gluconate, 10 mM HEPES, 1 mM EGTA and 1 mM EDTA (pH 7.2
with NaOH). Isolated mitoplasts were transparent vesicles with one to several black spots,
presumably formed by the remnants of the outer membrane in contact sites between two
mitochondrial membranes. In some experiments, mitoplasts were isolated from COS-7
cells transfected with yellow fluorescent protein (YFP) targeted to the inner membrane of
mitochondria (BD Clontech) to confirm the mitochondrial origin of the vesicles.
Mitoplasts of 2–5
m
M diameter (membrane capacitance, C
m
¼ 0.35–1 pF) were used for
patch-clamp experiments.
Whole-mitoplast recordings
After formation of a GQ seal between the patch-clamp pipette and inner mitochondrial
membrane, capacitance transients were completely compensated. Voltage steps of 200–
500 mV and 3–300 ms were then applied to rupture the membrane and obtain the whole-
mitoplast configuration as monitored by reappearance of capacitance transients and
increase in baseline noise. In some experiments BSA (0.05%) was added to the bath
solution to increase the survival of mitoplasts. Elevation of pipette solution tonicity (£1.5
bath solution) significantly reduced the access resistance associated with the mitoplast
shrinkage after break-in. The ionic compositions of pipette and bath solutions were
chosen to reduce potential contamination by K
þ
and Cl
2
currents, especially in the inward
Figure 4 Single i
MiCa
Ca
2þ
channels from inner mitochondrial membrane inside-out
patches (105 mM CaCl
2
solution at the cytoplasmic surface (pipette)). a, Multi-channel
patch currents. The bath contained 150 mM Na-gluconate, 1 mM EGTA/EDTA (free Ca
2þ
concentration at the matrix surface of the inner membrane ([Ca
2þ
]
m
) ,10 nM) (red trace)
or 105 mM [Ca
2þ
]
m
(black trace). b, i
MiCa
in 105 mM [Ca
2þ
]
m
(black trace) or 105 mM
[Mg
2þ
]
m
(red trace). c, Ensemble averages of 45 multi-channel responses. Voltage ramp:
2160 mV to þ160 mV. d, Rare single i
MiCa
. [Ca
2þ
]
m
and [Ca
2þ
]
c
were both 105 mM.
Note the fast open/close kinetics of the attenuated outward current. e, [RuR]
c
or [Ru360]
c
in the pipette inhibited i
MiCa
at 200 nM. Red traces indicate ensemble averages of 20
multi-channel responses.
letters to nature
NATURE | VOL 427 | 22 JANUARY 2004 | www.nature.com/nature 363
© 2004
Nature
Publishing
Group
direction. Three different pipette solutions were used: Cs-gluconate solution (150 mM
CsOH, 5 mM CsCl, 135 mM sucrose, 10 mM HEPES, 1.5 mM EGTA and 1.5 mM EDTA
(pH 7.2 with
D-gluconic acid)); Na-gluconate solution (150 mM Na-gluconate, 5 mM
NaCl, 135 mM sucrose, 10 mM HEPES, 1.5 mM EGTA and 1.5 mM EDTA (pH 7.2 with
NaOH)); Na-gluconate solution without Ca
2þ
buffer (150 mM Na-gluconate, 5 mM NaCl,
135 mM sucrose, 10 mM HEPES (pH 7.2 with NaOH)).
Signals were recorded using an Axopatch 200B patch-clamp amplifier, filtered at 5 kHz,
and sampled at 10 kHz. Capacitance of the whole-mitoplast membrane ranged from 0.35
to 1 pF. Access resistances, usually 30–60 MQ, were compensated by about 70%. Bath Ca
2þ
concentration was varied from 20
m
M to 5 mM by addition of the corresponding amount
of 1 M CaCl
2
stock solution into one of two solutions: Na-gluconate solution (150 mM
Na-gluconate, 10 mM HEPES (pH 7.2 with NaOH)) or HEPES-Tris solution (205 mM
HEPES (pH 7.2 with Trisma base)). Bath solution with 105 mM [Ca
2þ
]
c
(105 mM CaCl
2
solution) contained 105 mM CaCl
2
, 10 mM HEPES (pH 7.2 with Trisma base or NaOH).
This solution was dissolved with Na-gluconate solution or HEPES-Tris solution to obtain
26 mM [Ca
2þ
]
c
. Bath solutions with 5 mM of Sr
2þ
,Ba
2þ
,Mn
2þ
or Mg
2þ
were obtained by
addition of 1 M stock solution of the chloride salt into HEPES-Tris solution. HEPES-K
bath solution (KOH 75 mM (pH 7.2 with about 210 mM HEPES)); HEPES-Na bath
solution (NaOH 75 mM (pH 7.2 with about 210 mM HEPES)); Na-gluconate and K-
gluconate low-divalent solutions (150 mM Na(K)-gluconate, 10 mM HEPES, 5 mM
EGTA, 5 mM EDTA (pH 7.2 with NaOH)). CaCl
2
or MgCl
2
was added to Na-gluconate
low-divalent (divalent-free) solution in the amounts calculated by the WinMAXC v2.05
program (C. Patton, Stanford University) to obtain free Ca
2þ
and Mg
2þ
concentrations.
Osmolarity was approximately 280 mmol kg
21
. Statistical data was calculated as the
mean ^ s.d.
Single-channel recordings
All single-channel recordings were made in the inside-out configuration of the patch-
clamp technique. Patches were excised from the inner mitochondrial membrane in a bath
solution containing 150 mM Na-gluconate, 10 mM HEPES, 1 mM EGTA and 1 mM EDTA
(pH 7.2 with NaOH). Pipettes were filled with 105 mM CaCl
2
solution. A total of 105 mM
CaCl
2
solution was also used as the bath solution for single channels and whole-mitoplast
recordings. Signals were filtered at 1 kHz and sampled at 5 kHz. In Supplementary Fig. 2,
traces were filtered at 200 Hz before amplitude analysis.
Estimation of Ca
21
- and Mg
21
-binding constants
Ca
2þ
buffering is unreliable below 10 nM. To estimate the MiNa Ca
2þ
-binding constant,
we assumed that I
MiNa
in low-divalent solution was #I
MiNa
at theoretical 0 [Ca
2þ
]
c
. Given
that I
MiNa
at 10 nM [Ca
2þ
]
c
was about eightfold less than I
MiNa
in low-divalent solution,
the Ca
2þ
-binding equilibrium constant calculated from the Hill equation was #2nM
assuming a Hill coefficient of 1. Similarly, the MiNa-binding constant for Mg
2þ
was
estimated to be #250 nM.
Received 20 August; accepted 18 November 2003; doi:10.1038/nature02246.
1. Rizzuto, R., Bernardi, P. & Pozzan, T. Mitochondria as all-roundplayers of the calcium game. J. Physiol.
(Lond.) 529, 37–47 (2000).
2. Montero, M. et al. Chromaffin-cell stimulation triggers fast millimolar mitochondrial Ca
2þ
transients
that modulate secretion. Nature Cell Biol. 2, 57–61 (2000).
3. McCormack, J. G., Halestrap, A. P. & Denton, R. M. Role of calcium ions in regulation of mammalian
intramitochondrial metabolism. Physiol. Rev. 70, 391–425 (1990).
4. Hajnoczky, G., Robb-Gaspers, L. D., Seitz, M. B. & Thomas, A. P. Decoding of cytosolic calcium
oscillations in the mitochondria. Cell 82, 415–424 (1995).
5. Jouaville, L. S., Ichas, F., Holmuhamedov, E. L., Camacho, P. & Lechleiter, J. D. Synchronization of
calcium waves by mitochondrial substrates in Xenopus laevis oocytes. Nature 377, 438–441 (1995).
6. Budd, S. L. & Nicholls, D. G. A re-evaluation of the role of mitochondria in neuronal Ca
2þ
homeostasis. J. Neurochem. 66, 403–411 (1996).
7. Babcock, D. F., Herrington, J., Goodwin, P. C., Park, Y. B. & Hille, B. Mitochondrial participation in
the intracellular Ca
2þ
network. J. Cell Biol. 136, 833–844 (1997).
8. Boitier, E., Rea, R. & Duchen, M. R. Mitochondria exert a negative feedback on the propagation of
intracellular Ca
2þ
waves in rat cortical astrocytes. J. Cell Biol. 145, 795–808 (1999).
9. Scorrano, L. et al. BAX and BAK regulation of endoplasmic reticulum Ca
2þ
: a control point for
apoptosis. Science 300, 135–139 (2003).
10. Demaurex, N. & Distelhorst, C. Cell biology. Apoptosis
the calcium connection. Science 300, 65–67
(2003).
11. Gunter, K. K. & Gunter, T. E. Transport of calcium by mitochondria. J. Bioenerg. Biomembr. 26,
471–485 (1994).
12. Bernardi, P. Mitochondrial transport of cations: channels, exchangers, and permeability transition.
Physiol. Rev. 79, 1127–1155 (1999).
13. Gunter, T. E. & Pfeiffer, D. R. Mechanisms by which mitochondria transport calcium. Am. J. Physiol.
258, C755–C786 (1990).
14. DeLuca, H. F. & Engstrom, G. W. Calcium uptake by rat kidney mitochondria. Proc. Natl Acad. Sci.
USA 47, 1744–1750 (1961).
15. Rizzuto, R., Brini, M., Murgia, M. & Pozzan, T. Microdomains with high Ca
2þ
close to IP
3
-sensitive
channels that are sensed by neighboring mitochondria. Science 262, 744–747 (1993).
16. Neher, E. Vesicle pools and Ca
2þ
microdomains: new tools for understanding their roles in
neurotransmitter release. Neuron 20, 389–399 (1998).
17. Tashmukhamedov, B. A., Gagelgans, A. I., Mamatkulov, K. & Makhmudova, E. M. Inhibition of Ca
2þ
transport in mitochondria by selective blockade of membrane mucopolysaccharides by hexamine
cobalt chloride. FEBS Lett. 28, 239–242 (1972).
18. Ying, W. L., Emerson, J., Clarke, M. J. & Sanadi, D. R. Inhibition of mitochondrial calcium ion
transport by an oxo-bridged dinuclear ruthenium amine complex. Biochemistry 30, 4949–4952
(1991).
19. Matlib, M. A. et al. Oxygen-bridged dinuclear ruthenium amine complex specifically inhibits Ca
2þ
uptake into mitochondria in vitro and in situ in single cardiac myocytes. J. Biol. Chem. 273,
10223–10231 (1998).
20. Drahota, Z., Gazzotti, P., Carafoli, E. & Rossi, C. S. A comparison of the effects of different divalent
cations on a number of mitochondrial reactions linked to ion translocation. Arch. Biochem. Biophys.
130, 267–273 (1969).
21. Vainio, H., Mela, L. & Chance, B. Energy dependent bivalent cation translocation in rat liver
mitochondria. Eur. J. Biochem. 12, 387–391 (1970).
22. Selwyn, M. J., Dawson, A. P. & Dunnett, S. J. Calcium transport in mitochondria. FEBS Lett. 10, 1–5
(1970).
23. Kostyuk, P. G. & Krishtal, O. A. Effects of calcium and calcium-chelating agents on the inward and
outward current in the membrane of mollusc neurones. J. Physiol. (Lond.) 270, 569–580 (1977).
24. Almers, W., McCleskey, E. W. & Palade, P. T. A non-selective cation conductance in frog muscle
membrane blocked by micromolar external calcium ions. J. Physiol. (Lond.) 353, 565–583 (1984).
25. McCleskey, E. W. & Almers, W. The Ca channel in skeletal muscle is a large pore. Proc. Natl Acad. Sci.
USA 82, 7149–7153 (1985).
26. Prakriya, M. & Lewis, R. S. CRAC channels: activation, permeation, and the search for a molecular
identity. Cell Calcium 33, 311–321 (2003).
27. Vinogradov, A. & Scarpa, A. The initial velocities of calcium uptake by rat liver mitochondria. J. Biol.
Chem. 248, 5527–5531 (1973).
28. Bragadin, M., Pozzan, T. & Azzone, G. F. Kinetics of Ca
2þ
carrier in rat liver mitochondria.
Biochemistry 18, 5972–5978 (1979).
29. Hille, B. Ion Channels of Excitable Membranes (Sinauer Associates, Sunderland, Massachusetts, 2001).
30. Cannon, B. & Lindberg, O. Mitochondria from brown adipose tissue: isolation and properties.
Methods Enzymol. 55, 65–78 (1979).
Supplementary Information accompanies the paper on www.nature.com/nature.
Acknowledgements We would like to thank J. Borecky for advice on the whole-mitoplast
configuration; L. DeFelice, H. Xu, B. Desai, V. Sandler and P. Smith for discussions; and S. Gapon
and Y. Manasian for technical assistance.
Competing interests statement The authors declare that they have no competing financial
interests.
Correspondence and requests for materials should be addressed to D.E.C.
(dclapham@enders.tch.harvard.edu).
..............................................................
Two mitotic kinesins cooperate
to drive sister chromatid
separation during anaphase
Gregory C. Rogers
1
, Stephen L. Rogers
2
, Tamara A. Schwimmer
1
,
Stephanie C. Ems-McClung
3
, Claire E. Walczak
3
, Ronald D. Vale
2
,
Jonathan M. Scholey
4
& David J. Sharp
1
1
Department of Physiology and Biophysics, Albert Einstein College of Medicine,
Bronx, New York 10461, USA
2
The Howard Hughes Medical Institute and Department of Cellular and
Molecular Pharmacology, University of California, San Francisco, California
94143, USA
3
Medical Sciences Program, Indiana University, Bloomington, Indiana 47405,
USA
4
Center for Genetics and Development and Section of Molecular and Cellular
Biology, University of California, Davis, California 95616, USA
.............................................................................................................................................................................
During anaphase identical sister chromatids separate and move
towards opposite poles of the mitotic spindle
1,2
. In the spindle,
kinetochore microtubules
3
have their plus ends embedded in the
kinetochore and their minus ends at the spindle pole. Two models
have been proposed to account for the movement of chromatids
during anaphase. In the ‘Pac-Man model, kinetochores induce
the depolymerization of kinetochore microtubules at their plus
ends, which allows chromatids to move towards the pole by
chewing up’ microtubule tracks
4,5
. In the ‘poleward flux’ model,
kinetochores anchor kinetochore microtubules and chromatids
are pulled towards the poles through the depolymerization of
kinetochore microtubules at the minus ends
6
. Here, we show that
two functionally distinct microtubule-destabilizing KinI kinesin
letters to nature
NATURE | VOL 427 | 22 JANUARY 2004 | www.nature.com/nature364
© 2004
Nature
Publishing
Group
... Our simulations have revealed that in the presence of membrane potential, pentamidine exhibits a notable preference for entering cells over exiting, as indicated by the differences in energy barriers. This distinctive behavior shares some similarities with inwardly rectifying ion channels, such as the mitochondrial calcium uniporter 29 . ...
Article
Full-text available
Pentamidine and melarsoprol are primary drugs used to treat the lethal human sleeping sickness caused by the parasite Trypanosoma brucei. Cross-resistance to these two drugs has recently been linked to aquaglyceroporin 2 of the trypanosome (TbAQP2). TbAQP2 is the first member of the aquaporin family described as capable of drug transport; however, the underlying mechanism remains unclear. Here, we present cryo-electron microscopy structures of TbAQP2 bound to pentamidine or melarsoprol. Our structural studies, together with the molecular dynamic simulations, reveal the mechanisms shaping substrate specificity and drug permeation. Multiple amino acids in TbAQP2, near the extracellular entrance and inside the pore, create an expanded conducting tunnel, sterically and energetically allowing the permeation of pentamidine and melarsoprol. Our study elucidates the mechanism of drug transport by TbAQP2, providing valuable insights to inform the design of drugs against trypanosomiasis.
... Numerous reviews [6,7], describe how mammalian cells control [Ca 2+ ]CYT levels and the significance of the nature of variations in [Ca 2+ ]CYT (such as [Ca 2+ ]CYT oscillations and localized changes in Ca 2+ ). As Figure 1 shows a few of the primary calcium exchangers, pumps, and channels in the pathways that signal calcium. ...
Article
Numerous cellular processes, including the release of neurotransmitters and the contraction of muscles, are largely triggered and regulated by Ca2+ inflow via Ca2+ permeable ion channels. In addition, Ca2+ influx regulates cellular migration and proliferation, two mechanisms linked to cancer. This study focuses on calcium influx in breast cancer cells and discusses how future drugs for breast cancer therapy may be pharmacological modulators of particular Ca2+ influx channels. Certain breast tumors have altered expression of particular calcium permeable ion channels. Such alterations may occasionally be connected to the prognosis and subtype of breast cancer. These days, models both in vivo and in vitro have assisted in identifying particular Ca2+ channels that are crucial for the growth and invasiveness of cancerous breast cells. Nonetheless, additional research is still needed to fully understand several features of Ca2+ influx in breast cancer. These include figuring out the processes behind the changed expression and the best treatment plan to target breast cancer cells via particular Ca2+ channels. In the upcoming ten years, research should concentrate on the function of Ca2+ influx in mechanisms other than the migration and proliferation of breast cancer cells
... In 2004, Clapham et al. discovered the existence of a Ca 2+ -selective mitochondrial ion channel. The analysis revealed that the current was mediated by the mitochondrial calcium uniporter (MCU), which controls the stressdependent transport of Ca 2+ through the inner mitochondrial membrane [9,10]. ...
Article
Full-text available
Epilepsy is one of the most common neurological disorders in the world. Common epileptic drugs generally affect ion channels or neurotransmitters and prevent the emergence of seizures. However, up to a third of the patients suffer from drug-resistant epilepsy, and there is an urgent need to develop new therapeutic strategies that go beyond acute antiepileptic (antiseizure) therapies towards therapeutics that also might have effects on chronic epilepsy comorbidities such as cognitive decline and depression. The mitochondrial calcium uniporter (MCU) mediates rapid mitochondrial Ca²⁺ transport through the inner mitochondrial membrane. Ca²⁺ influx is essential for mitochondrial functions, but longer elevations of intracellular Ca²⁺ levels are closely associated with seizure-induced neuronal damage, which are underlying mechanisms of cognitive decline and depression. Using neuronal-specific MCU knockout mice (MCU−/−ΔN), we demonstrate that neuronal MCU deficiency reduced hippocampal excitability in vivo. Furthermore, in vitro analyses of hippocampal glioneuronal cells reveal no change in total Ca²⁺ levels but differences in intracellular Ca²⁺ handling. MCU−/−ΔN reduces ROS production, declines metabolic fluxes, and consequently prevents glioneuronal cell death. This effect was also observed under pathological conditions, such as the low magnesium culture model of seizure-like activity or excitotoxic glutamate stimulation, whereby MCU−/−ΔN reduces ROS levels and suppresses Ca²⁺ overload seen in WT cells. This study highlights the importance of MCU at the interface of Ca²⁺ handling and metabolism as a mediator of stress-related mitochondrial dysfunction, which indicates the modulation of MCU as a potential target for future antiepileptogenic therapy.
... In this context, electrophysiology of organellar membranes is technically feasible by enlarging the compartment (Bertholet, 2021;Chen et al., 2017;Shapovalov et al., 2017). By resorting to enlargement, several successful attempts have been made to record directly from mitochondrial membranes (Kirichok et al., 2004;Sorgato et al., 1987), lysosomal membranes (Chen et al., 2017; and the endoplasmic reticulum (ER) (Mak et al., 2013;Vais et al., 2020). In contrast, fluorescence-based methods for recording org and its dynamics are emerging as an appealing complement to traditional electrophysiology, providing less-invasive conditions, better targeting specificity and technical simplicity. ...
Article
Full-text available
The eukaryotic cell is highly compartmentalized with organelles. Owing to their function in transporting metabolites, metabolic intermediates and byproducts of metabolic activity, organelles are important players in the orchestration of cellular function. Recent advances in optical methods for interrogating the different aspects of organellar activity promise to revolutionize our ability to dissect cellular processes with unprecedented detail. The transport activity of organelles is usually coupled to the transport of charged species; therefore, it is not only associated with the metabolic landscape but also entangled with membrane potentials. In this context, the targeted expression of fluorescent probes for interrogating organellar membrane potential (Ψorg) emerges as a powerful approach, offering less‐invasive conditions and technical simplicity to interrogate cellular signalling and metabolism. Different research groups have made remarkable progress in adapting a variety of optical methods for measuring and monitoring Ψorg. These approaches include using potentiometric dyes, genetically encoded voltage indicators, hybrid fluorescence resonance energy transfer sensors and photoinduced electron transfer systems. These studies have provided consistent values for the resting potential of single‐membrane organelles, such as lysosomes, the Golgi and the endoplasmic reticulum. We can foresee the use of dynamic measurements of Ψorg to study fundamental problems in organellar physiology that are linked to serious cellular disorders. Here, we present an overview of the available techniques, a survey of the resting membrane potential of internal membranes and, finally, an open‐source mathematical model useful to interpret and interrogate membrane‐bound structures of small volume by using the lysosome as an example. image
... To achieve the cytosolic basal concentrations again and allow a new signaling wave, Ca 2+ is continuously removed from the cytosol. This action also involves several transporters, such as PMCA, SERCA, MCU, and the NCX (Carafoli 2002;Berridge et al. 2003;Kirichok et al. 2004). PMCA and NCX are indicated as the two main systems remove Ca 2+ from cytosol (Carafoli 1987;Khananshvili 1998;Brini and Carafoli 2011), with PMCA acting constitutively to maintain a primary Ca 2+ gradient across the plasma membrane at the expense of energy expenditure, although NCX responds to transient changes of [Ca 2+ ] c (Carafoli 1987;Blaustein and Lederer 1999). ...
Article
The heart beats approximately 100,000 times per day in humans, imposing substantial energetic demands on cardiac muscle. Adenosine triphosphate (ATP) is an essential energy source for normal function of cardiac muscle during each beat, as it powers ion transport, intracellular Ca ²⁺ handling, and actin–myosin cross-bridge cycling. Despite this, the impact of excitation–contraction coupling on the intracellular ATP concentration ([ATP] i ) in myocytes is poorly understood. Here, we conducted real-time measurements of [ATP] i in ventricular myocytes using a genetically encoded ATP fluorescent reporter. Our data reveal rapid beat-to-beat variations in [ATP] i . Notably, diastolic [ATP] i was <1 mM, which is eightfold to 10-fold lower than previously estimated. Accordingly, ATP-sensitive K ⁺ (K ATP ) channels were active at physiological [ATP] i . Cells exhibited two distinct types of ATP fluctuations during an action potential: net increases (Mode 1) or decreases (Mode 2) in [ATP] i . Mode 1 [ATP] i increases necessitated Ca ²⁺ entry and release from the sarcoplasmic reticulum (SR) and were associated with increases in mitochondrial Ca ²⁺ . By contrast, decreases in mitochondrial Ca ²⁺ accompanied Mode 2 [ATP] i decreases. Down-regulation of the protein mitofusin 2 reduced the magnitude of [ATP] i fluctuations, indicating that SR-mitochondrial coupling plays a crucial role in the dynamic control of ATP levels. Activation of β-adrenergic receptors decreased [ATP] i , underscoring the energetic impact of this signaling pathway. Finally, our work suggests that cross-bridge cycling is the largest consumer of ATP in a ventricular myocyte during an action potential. These findings provide insights into the energetic demands of EC coupling and highlight the dynamic nature of ATP concentrations in cardiac muscle.
Article
Mitochondrial membrane potential (MMP) plays a crucial role in the function of cells and organelles, involving various cellular physiological processes, including energy production, formation of reactive oxygen species (ROS), unfolded protein stress, and cell survival. Currently, there is a lack of genetically encoded fluorescence indicators (GEVIs) for MMP. In our screening of various GEVIs for their potential monitoring MMP, the Accelerated Sensor of Action Potentials (ASAP) demonstrated optimal performance in targeting mitochondria and sensitivity to depolarization in multiple cell types. However, mitochondrial ASAPs also displayed sensitivity to ROS in cardiomyocytes. Therefore, two ASAP mutants resistant to ROS were generated. A double mutant ASAP3-ST exhibited the highest voltage sensitivity but weaker fluorescence. Overall, four GEVIs capable of targeting mitochondria were obtained and named mitochondrial potential indicators 1–4 (MPI-1–4). In vivo, fiber photometry experiments utilizing MPI-2 revealed a mitochondrial depolarization during isoflurane-induced narcosis in the M2 cortex.
Article
Full-text available
Intramuscular fat (IMF) in pork holds significant importance for economic performance within the pig industry and dietary calcium supplementation enhances the accumulation of intramuscular fat. Additionally, calcium ions inhibit translation and reduce protein synthesis. However, the mechanism by which calcium regulates IMF deposition in muscle through translation remains largely unknown. In this study, we compared the ribosome profiles of the longissimus dorsi muscles of Duroc ×\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\times$$\end{document} Landrace ×\documentclass[12pt]{minimal} \usepackage{amsmath} \usepackage{wasysym} \usepackage{amsfonts} \usepackage{amssymb} \usepackage{amsbsy} \usepackage{mathrsfs} \usepackage{upgreek} \setlength{\oddsidemargin}{-69pt} \begin{document}$$\times$$\end{document} Large white pigs from the normal calcium (NC) group or calcium supplement (HC) group by Ribo-seq, and RNA-seq. By integrating multiple-omics analysis, we further discovered 437 genes that were transcriptionally unchanged but translationally altered and these genes were significantly enriched in the oxidative phosphorylation signaling pathway. Furthermore, experimental data showed that inhibiting the expression of COX10 and mtND4L increased triglyceride accumulation in C2C12 cells, providing new targets for intramuscular fat deposition. Finally, this work links dietary calcium, translation regulation and IMF deposition, providing a new strategy for both meat quality and economic performance within the pig industry.
Article
Full-text available
Mitochondria, with their intricate networks of functions and information processing, are pivotal in both health regulation and disease progression. Particularly, mitochondrial dysfunctions are identified in many common pathologies, including cardiovascular diseases, neurodegeneration, metabolic syndrome, and cancer. However, the multifaceted nature and elusive phenotypic threshold of mitochondrial dysfunction complicate our understanding of their contributions to diseases. Nonetheless, these complexities do not prevent mitochondria from being among the most important therapeutic targets. In recent years, strategies targeting mitochondrial dysfunction have continuously emerged and transitioned to clinical trials. Advanced intervention such as using healthy mitochondria to replenish or replace damaged mitochondria, has shown promise in preclinical trials of various diseases. Mitochondrial components, including mtDNA, mitochondria-located microRNA, and associated proteins can be potential therapeutic agents to augment mitochondrial function in immunometabolic diseases and tissue injuries. Here, we review current knowledge of mitochondrial pathophysiology in concrete examples of common diseases. We also summarize current strategies to treat mitochondrial dysfunction from the perspective of dietary supplements and targeted therapies, as well as the clinical translational situation of related pharmacology agents. Finally, this review discusses the innovations and potential applications of mitochondrial transplantation as an advanced and promising treatment.
Article
A disturbed mitochondrial function contributes to the pathology of many common diseases. These organelles are therefore important therapeutic targets. On the contrary, many adverse effects of drugs can be explained by a mitochondrial off‐target effect, in particular, due to an interaction with carrier proteins in the inner membrane. Yet this class of transport proteins remains underappreciated and understudied. The aim of this review is to provide a deeper understanding of the role of mitochondrial carriers in health and disease and their significance as drug targets. We present literature‐based evidence that mitochondrial carrier proteins are associated with prevalent diseases and emphasize their potential as drug (off‐)target sites by summarizing known mitochondrial drug–transporter interactions. Studying these carriers will enhance our knowledge of mitochondrial drug on‐ and off‐targets and provide opportunities to further improve the efficacy and safety of drugs.
Article
Full-text available
Calcium can activate mitochondrial metabolism, and the possibility that mitochondrial Ca2+ uptake and extrusion modulate free cytosolic [Ca2+] (Cac) now has renewed interest. We use whole-cell and perforated patch clamp methods together with rapid local perfusion to introduce probes and inhibitors to rat chromaffin cells, to evoke Ca2+ entry, and to monitor Ca2+-activated currents that report near-surface [Ca2+]. We show that rapid recovery from elevations of Cac requires both the mitochondrial Ca2+ uniporter and the mitochondrial energization that drives Ca2+ uptake through it. Applying imaging and single-cell photometric methods, we find that the probe rhod-2 selectively localizes to mitochondria and uses its responses to quantify mitochondrial free [Ca2+] (Cam). The indicated resting Cam of 100–200 nM is similar to the resting Cac reported by the probes indo-1 and Calcium Green, or its dextran conjugate in the cytoplasm. Simultaneous monitoring of Cam and Cac at high temporal resolution shows that, although Cam increases less than Cac, mitochondrial sequestration of Ca2+ is fast and has high capacity. We find that mitochondrial Ca2+ uptake limits the rise and underlies the rapid decay of Cac excursions produced by Ca2+ entry or by mobilization of reticular stores. We also find that subsequent export of Ca2+ from mitochondria, seen as declining Cam, prolongs complete Cac recovery and that suppressing export of Ca2+, by inhibition of the mitochondrial Na+/ Ca2+ exchanger, reversibly hastens final recovery of Cac. We conclude that mitochondria are active participants in cellular Ca2+ signaling, whose unique role is determined by their ability to rapidly accumulate and then release large quantities of Ca2+.
Article
Full-text available
Ruthenium red is a well known inhibitor of Ca2+ uptake into mitochondria in vitro. However, its utility as an inhibitor of Ca2+ uptake into mitochondria in vivo or in situ in intact cells is limited because of its inhibitory effects on sarcoplasmic reticulum Ca2+ release channel and other cellular processes. We have synthesized a ruthenium derivative and found it to be an oxygen-bridged dinuclear ruthenium amine complex. It has the same chemical structure as Ru360 reported previously (Emerson, J., Clarke, M. J., Ying, W-L., and Sanadi, D. R. (1993) J. Am. Chem. Soc.115, 11799–11805). Ru360 has been shown to be a potent inhibitor of Ca2+-stimulated respiration of liver mitochondria in vitro. However, the specificity of Ru360 on Ca2+uptake into mitochondria in vitro or in intact cells has not been determined. The present study reports in detail the potency, the effectiveness, and the mechanism of inhibition of mitochondrial Ca2+ uptake by Ru360 and its specificity in vitro in isolated mitochondria and in situ in isolated cardiac myocytes. Ru360 was more potent (IC50 = 0.184 nm) than ruthenium red (IC50 = 6.85 nm) in inhibiting Ca2+ uptake into mitochondria. 103Ru360 was found to bind to isolated mitochondria with high affinity (K d = 0.34 nm, B max = 80 fmol/mg of mitochondrial protein). The IC50 of 103Ru360 for the inhibition of Ca2+ uptake into mitochondria was also 0.2 nm, indicating that saturation of a specific binding site is responsible for the inhibition of Ca2+uptake. Ru360, as high as 10 μm, produced no effect on sarcoplasmic reticulum Ca2+ uptake or release, sarcolemmal Na+/Ca2+ exchange, actomyosin ATPase activity, L-type Ca2+ channel current, cytosolic Ca2+transients, or cell shortening. 103Ru360 was taken up by isolated myocytes in a time-dependent biphasic manner. Ru360 (10 μm) applied outside intact voltage-clamped ventricular myocytes prevented Ca2+ uptake into mitochondria in situ where the cells were progressively loaded with Ca2+ via sarcolemmal Na+/Ca2+ exchange by depolarization to +110 mV. We conclude that Ru360 specifically blocks Ca2+ uptake into mitochondria and can be used in intact cells.
Article
Full-text available
Background The activation of T cells, mediated by the T-cell receptor (TCR), activates a battery of specific membrane-associated, cytosolic and nuclear proteins. Identifying the signaling proteins downstream of TCR activation will help us to understand the regulation of immune responses and will contribute to developing therapeutic agents that target immune regulation. Results In an effort to identify novel signaling molecules specific for T-cell activation we undertook a large-scale dominant effector genetic screen using retroviral technology. We cloned and characterized 33 distinct genes from over 2,800 clones obtained in a screen of 7 × 108 Jurkat T cells on the basis of a reduction in TCR-activation-induced CD69 expression after expressing retrovirally derived cDNA libraries. We identified known signaling molecules such as Lck, ZAP70, Syk, PLCγ1 and SHP-1 (PTP1C) as truncation mutants with dominant-negative or constitutively active functions. We also discovered molecules not previously known to have functions in this pathway, including a novel protein with a RING domain (found in a class of ubiquitin ligases; we call this protein TRAC-1), transmembrane molecules (EDG1, IL-10Rα and integrin α2), cytoplasmic enzymes and adaptors (PAK2, A-Raf-1, TCPTP, Grb7, SH2-B and GG2-1), and cytoskeletal molecules (moesin and vimentin). Furthermore, using truncated Lck, PLCγ1, EDG1 and PAK2 mutants as examples, we showed that these dominant immune-regulatory molecules interfere with IL-2 production in human primary lymphocytes. Conclusions This study identified important signal regulators in T-cell activation. It also demonstrated a highly efficient strategy for discovering many components of signal transduction pathways and validating them in physiological settings.
Article
Full-text available
Sphingolipid signaling pathways have been implicated in many critical cellular events. Sphingosine-1-phosphate (SPP), a sphingolipid metabolite found in high concentrations in platelets and blood, stimulates members of the endothelial differentiation gene (Edg) family of G protein–coupled receptors and triggers diverse effects, including cell growth, survival, migration, and morphogenesis. To determine the in vivo functions of the SPP/Edg signaling pathway, we disrupted the Edg1 gene in mice. Edg1–/– mice exhibited embryonic hemorrhage leading to intrauterine death between E12.5 and E14.5. Vasculogenesis and angiogenesis appeared normal in the mutant embryos. However, vascular maturation was incomplete due to a deficiency of vascular smooth muscle cells/pericytes. We also show that Edg-1 mediates an SPP-induced migration response that is defective in mutant cells due to an inability to activate the small GTPase, Rac. Our data reveal Edg-1 to be the first G protein–coupled receptor required for blood vessel formation and show that sphingolipid signaling is essential during mammalian development.
Article
Although it has been known for over three decades that mitochondria are endowed with a complex array of Ca2+ transporters and that key enzymes of mitochondrial metabolism are regulated by Ca2+, the possibility that physiological stimuli that raise the [Ca2+] of the cytoplasm could trigger major mitochondrial Ca2+ uptake has long been considered unlikely, based on the low affinity of the mitochondrial transporters and the limited amplitude of the cytoplasmic [Ca2+] rises. The direct measurement of mitochondrial [Ca2+] with highly selective probes has led to a complete reversion of this view, by demonstrating that, after cell stimulation, the cytoplasmic Ca2+ signal is always paralleled by a much larger rise in [Ca2+] in the mitochondrial matrix. This observation has rejuvenated the study of mitochondrial Ca2+ transport and novel, unexpected results have altered long-standing dogmas in the field of calcium signalling. Here we focus on four main topics: (i) the current knowledge of the functional properties of the Ca2+ transporters and of the thermodynamic constraints under which they operate; (ii) the occurrence of mitochondrial Ca2+ uptake in living cells and the key role of local signalling routes between the mitochondria and the Ca2+ sources; (iii) the physiological consequences of Ca2+ transport for both mitochondrial function and the modulation of the cytoplasmic Ca2+ signal; and (iv) evidence that alterations of mitochondrial Ca2+ signalling may occur in pathophysiological conditions.
Article
Work in my laboratory related to the subject of this review is supported by the Behrens-Weise-Stiftung and by the Deutsche Forschungsgemeinschaft (SFB 406 and SFB 523), by a grant from the European Community (No. CHRX-CT94–0500), and by the Human Science Frontier Program (RG-4/95B). I would like to thank my colleagues Eric Kandel, Tom Jessell, Bert Sakmann, Alain Marty, Tobias Moser, and Christian Rosenmund for helpful suggestions on the manuscript.
Article
The rate of aerobic Ca2+ transport is limited by the rate of the H+ pump rather than by the Ca2+ carrier. The kinetics of the Ca2+ carrier has therefore been studied by using the K+ diffusion potential as the driving force. The apparent Vmax of the Ca2+ carrier is, at 20 degrees C, about 900 nmol (mg of protein)-1 min-1, more than twice the rate of the H+ pump. The apparent Vmax is depressed by Mg2+ and Li+. This supports the view that the electrolytes act as noncompetitive inhibitors of the Ca2+ carrier. The degree of sigmoidicity of the kinetics of Ca2+ transport increases with the lowering of the temperature and proportionally with the concentration of impermeant electrolytes such as Mg2+ and Li+ but not choline. The effects of temperature and of electrolyte do not support the view that the sigmoidicity is due to modifications of the surface potential. Rather, they suggest that Ca2+ transport occurs through a multisubunit carrier, where cooperative phenomena are the result of ligand-induced conformational changes due to the interaction of several allosteric effectors with the carrier subunits. In contrast with La3+ which acts as a competitive inhibitor, Ruthenium Red affects the kinetics by inducing phenomena both of positive and of negative cooperativity. The Ruthenium Red induced kinetics has been reproduced through curve-fitting procedures by applying the Koshland sequential interaction hypothesis to a four-subunit Ca2+ carrier model.