Download Endogenous Drp1 Mediates Mitochondrial Autophagy and Protects

Document related concepts

Cell membrane wikipedia , lookup

Extracellular matrix wikipedia , lookup

Cell encapsulation wikipedia , lookup

Signal transduction wikipedia , lookup

Cell cycle wikipedia , lookup

Cellular differentiation wikipedia , lookup

Cell culture wikipedia , lookup

Endomembrane system wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

Mitosis wikipedia , lookup

Cell growth wikipedia , lookup

Amitosis wikipedia , lookup

Cytokinesis wikipedia , lookup

Apoptosis wikipedia , lookup

List of types of proteins wikipedia , lookup

Autophagy wikipedia , lookup

JADE1 wikipedia , lookup

Apoptosome wikipedia , lookup

SULF1 wikipedia , lookup

Programmed cell death wikipedia , lookup

Transcript
 Endogenous Drp1 Mediates Mitochondrial Autophagy and Protects the Heart Against
Energy Stress
Yoshiyuki Ikeda1, Akihiro Shirakabe1, Yasuhiro Maejima1, Peiyong Zhai1, Sebastiano Sciarretta1,2,
Jessica Toli1, Masatoshi Nomura3, Katsuyoshi Mihara4, Kensuke Egashira5,6, Mitsuru Ohishi7, Maha
Abdellatif1, Junichi Sadoshima1
1
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New
Jersey Medical School, Newark, New Jersey 07103, USA; 2IRCCS Neuromed, Pozzilli, Italy;
3
Department of Medicine and Bioregulatory Science, Kyushu University, Japan; 4Department of
Molecular Biology, Graduate School of Medical Science, Kyushu University, Japan; 5Department of
Cardiovascular Medicine, Kyushu University Hospital, Japan; 6Department of Cardiovascular Research,
Development, and Translational Medicine, Graduate School of Medical Science, Kyushu University
Hospital; 7Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and
Dental Science, Kagoshima University, Japan
Running title: Drp1 Mediates Autophagy
Subject codes:
[131] Apoptosis
[138] Cell signaling/signal transduction
[140] Energy metabolism
Address correspondence to:
Dr. Junichi Sadoshima
Department of Cell Biology and Molecular Medicine
Cardiovascular Research Institute
Rutgers New Jersey Medical School
185 South Orange Ave., MSB G609
Newark, NJ 07103
Tel: 973-972-8619
Fax: 973-972-8919
[email protected]
In September, 2014, the average time from submission to first decision for all original research papers
submitted to Circulation Research was 14.29 days.
DOI: 10.1161/CIRCRESAHA.116.303356 1
ABSTRACT
Rationale: Both fusion and fission contribute to mitochondrial quality control. How unopposed fusion
affects survival of cardiomyocytes (CMs) and left ventricular (LV) function in the heart is poorly
understood.
Objective: We investigated the role of Dynamin-related protein 1 (Drp1), a GTPase that mediates
mitochondrial fission, in mediating mitochondrial autophagy, ventricular function, and stress resistance in
the heart.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Methods and Results: Drp1 downregulation induced mitochondrial elongation, accumulation of damaged
mitochondria, and increased apoptosis in CMs at baseline. Drp1 downregulation also suppressed
autophagosome formation and autophagic flux at baseline and in response to glucose deprivation in CMs.
The lack of lysosomal translocation of mitochondrially-targeted Keima indicates that Drp1 downregulation
suppressed mitochondrial autophagy. Mitochondrial elongation and accumulation of damaged
mitochondria were also observed in tamoxifen-inducible cardiac-specific Drp1 knockout (Drp1-CKO) mice.
Following Drp1 downregulation, Drp1-CKO mice developed LV dysfunction, preceded by mitochondrial
dysfunction, and died within 13 weeks. Autophagic flux is significantly suppressed in Drp1-CKO mice.
Although LV function in cardiac-specific Drp1 heterozygous KO (Drp1-hetCKO) mice was normal at 12
weeks of age, LV function decreased more severely after 48 hours of fasting and the infarct size/area at risk
after ischemia/reperfusion (I/R) was significantly greater in Drp1-hetCKO than in control mice.
Conclusions: Disruption of Drp1 induces mitochondrial elongation, inhibits mitochondrial autophagy, and
causes mitochondrial dysfunction, thereby promoting cardiac dysfunction and increased susceptibility to
I/R.
Keywords:
Mitochondria, Drp1, heart, mitochondrial autophagy, ischemia/reperfusion.
Nonstandard Abbreviations and Acronyms:
Ad
adenovirus
Ad-shAtg7
adenovirus harboring Atg7 shRNA
Ad-shDrp1
adenovirus harboring Drp1 shRNA
Ad-shScr
adenovirus harboring scramble shRNA
Ad-tf-LC3
adenovirus harboring tandem fluorescent mRFP-GFP-LC3
MHC
alpha myosin heavy chain
CM
cardiomyocyte
COX IV
cytochrome c oxidase subunit IV
CsA
cyclosporin A
Drp1
dynamin-related protein 1
Drp1-CKO
cardiac-specific conditional Drp1 knockout
Drp1-hetCKO
cardiac-specific heterozygous Drp1 knockout
EM
electron microscopic/microscopy
GD
glucose deprivation
Keima-MLS
Keima with mitochondrial localization signal
I/R
ischemia/reperfusion
LV
left ventricular
Mfn
Mitofusin
MI/AAR
infarct size/area at risk
mPTP
mitochondrial permeability transition pore
DOI: 10.1161/CIRCRESAHA.116.303356 2
OCR
PGC-1
ROS
Tg-tf-LC3
TI
oxygen consumption rate
peroxisome proliferator-activated receptor-gamma coactivator 1
reactive oxygen species
transgenic mice expressing mRFP-GFP-LC3
tamoxifen injection
INTRODUCTION
The heart muscle is characterized by a large volume of mitochondria due to its high energy demand
. Mitochondria produce ATP primarily by utilizing the electrochemical gradient formed by electron
transfer via the electron transport chain located on the inner mitochondrial membrane. However, electron
leakage from the electron transport chain and production of O2- and H2O2, which arises from dismutation
of O2-, occur constantly as byproducts of ATP synthesis, making mitochondria a major source of reactive
oxygen species (ROS) in cardiomyocytes (CMs). Although ROS at physiological levels act as signaling
molecules to induce adaptive responses 4, dysregulated ROS production in response to stress damages
mitochondrial proteins, stimulating a feed-forward mechanism for ROS production, mitochondrial
dysfunction, and cell death, including apoptosis triggered by cytochrome c release and necrosis triggered
by mitochondrial permeability transition pore (mPTP) opening. To protect against these catastrophic events,
cells have intrinsic quality control mechanisms to maintain the overall health of mitochondria, including
fusion, fission and mitochondrial autophagy 5.
1-3
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Mitochondria are dynamic organelles that constantly undergo fusion and fission 5 to adapt to
changes in the cellular environment. Whereas mitochondrial fusion allows mitochondria to maintain
membrane potential by fusing depolarized mitochondria to intact ones, fission allows the segregation of
unrecoverable mitochondria so that they can be eliminated by autophagy or mitophagy, a specialized form
of autophagy 6. Mitochondrial fusion is critically regulated by mitofusin 1 (Mfn1) and mitofusin 2 (Mfn2),
specialized proteins localized on the outer mitochondrial membrane, and by Opa1,a protein localized on
the inner mitochondrial membrane, whereas mitochondrial fission is regulated by mitochondrial fission 1
(Fis1) and mitochondrial fission factor (Mff), localized on the outer mitochondrial membrane, and by
recruitment of a cytoplasmic GTPase, Dynamin-related protein 1 (Drp1), to mitochondrial fission sites 7, 8.
Although the presence of fission and fusion has not been documented in adult ventricular myocytes
in an unequivocal manner, previous studies have suggested that mitochondrial quality control plays an
essential role in protecting the heart against stress 2. For example, downregulation of Mfn1 and Mfn2
promotes cardiac dysfunction at baseline and in response to stress due to the lack of mitochondrial fusion
9, 10
. In contrast, Drp1-mediated mitochondrial fission appears to promote cell death during
ischemia/reperfusion (I/R) 11. Since suppression of Drp1 induces mitochondrial fusion, these results have
led to a general belief that mitochondrial fusion is protective 12, 13. However, experiments investigating the
role of fission in the heart were conducted using mdivi-1, a chemical inhibitor of Drp1 14.
Drp1 plays an essential role in mediating Parkin-induced mitochondria selective autophagy, namely
mitophagy in MEF cells 15. Drp1 also mediates Bnip3-induced autophagy in adult CMs 16. However,
whether Drp1 is involved in general autophagy that can remove mitochondria (which we here referred to
as mitochondrial autophagy) at baseline and in response to stress in CMs awaits further investigation using
specific interventions. We reasoned that loss-of-function experiments should be conducted using shRNA
or a mouse model of genetic deletion of Drp1 17 in order to address the role of endogenous Drp1 in regulating
mitochondrial autophagy and consequent CM survival and death. In this study, we asked 1) whether
endogenous Drp1 plays a protective or detrimental role in the heart at baseline and in response to stress and
2) whether Drp1 mediates mitochondrial autophagy in response to energy stress in CMs.
DOI: 10.1161/CIRCRESAHA.116.303356 3
METHODS
An expanded Methods section is available in the online Data Supplement.
Mouse models.
Generation of Drp1 flox homo (fl/fl) mice has been described 17. Cardiac-specific conditional Drp1
knockout (Drp1-CKO) mice were generated by crossing Drp1 fl/fl mice and MHC-MerCreMer mice, and
expression of Drp1 was downregulated by tamoxifen injection (20 mg/kg, ip) for 5 days. Cardiac-specific
heterozygous Drp1 KO (Drp1-hetCKO) mice were generated by crossing Drp1 flox hetero (fl/+) mice and
MHC-Cre mice. Transgenic mice expressing mRFP-GFP-LC3 (Tg-tf-LC3) have been described 18. All
experiments involving animals were approved by the Rutgers–New Jersey Medical School’s Institutional
Animal Care and Use Committee.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Keima with mitochondrial localization signal (Keima-MLS).
Keima is a fluorescent protein that emits different colored signals at acidic and neutral pHs. Keima-MLS is
a mitochondrially localized pH-indicator protein described by Katayama et al 19. We generated adenovirus
harboring Keima-MLS. The method used to detect lysosomal delivery of Keima-MLS has been described
19
.
Statistical analysis.
Data are expressed as mean ± SEM. The difference in means between 2 groups was evaluated using the ttest. One-way ANOVA was used to compare multiple groups. Post-hoc comparisons of considered pairs
were performed using the Bonferroni post-hoc test. P values of <0.05 were considered statistically
significant. In figure legends, “n” indicates the number of experiments.
RESULTS
Drp1 downregulation stimulates apoptosis in CMs.
To evaluate the role of endogenous Drp1 in regulating mitochondrial morphology in CMs, we
constructed adenovirus harboring Drp1 shRNA (Ad-shDrp1) and confirmed that Ad-shDrp1 decreases
Drp1 in CMs compared to adenovirus harboring scramble shRNA (Ad-shScr) (Online Figure IA). To
observe the morphology of mitochondria, cultured CMs were co-transduced with adenovirus harboring
mitochondrially targeted DsRed2 (mt-DsRed2). Ninety-six hours after transduction, mitochondria in AdshDrp1-transduced CMs were elongated compared to those in Ad-shScr-transduced CMs (Figure 1A). The
proportion of CMs with elongated mitochondria, as defined by an average mitochondrion length greater
than two sarcomere units (Online Figure IB), was significantly greater in Ad-shDrp1-transduced CMs than
in Ad-shScr-transduced CMs (Figure 1A). On the other hand, CMs with foreshortened mitochondria, as
defined by an average mitochondrion length smaller than one sarcomere unit (Online Figure IB), were
markedly reduced in Ad-shDrp1-transduced CMs. These results suggest that Drp1 is required for
mitochondrial foreshortening in CMs at baseline.
Transduction with Ad-shDrp1 significantly increased the number of TUNEL-positive CMs (Figure
1B) and the amount of cleaved caspase 3 compared to transduction with Ad-shScr (Figure 1C), suggesting
that endogenous Drp1 is essential in protection against apoptosis in CMs. To exclude the possibility that
our timing prevented observation of a period during which Drp1 downregulation-induced fusion is
protective, we evaluated CM viability at various time points after transduction of Ad-shDrp1 and Ad-shScr.
Viability time-dependently decreased between 0 and 96 hours after transduction of Ad-shDrp1 into CMs,
DOI: 10.1161/CIRCRESAHA.116.303356 4
and was significantly lower in Ad-shDrp1-transduced CMs than in Ad-shScr-transduced CMs after 72 hours
(Figure 1D), indicating that Drp1 downregulation is persistently detrimental.
Endogenous Drp1 mediates autophagy and mitochondrial quality control.
We next examined the role of Drp1 in CM autophagy. Drp1 downregulation with Ad-shDrp1
significantly reduced LC3-II and increased p62/SQSTM1, a protein degraded by autophagy (Figure 2A).
To evaluate autophagic flux, CMs were treated with chloroquine, which inhibits fusion of autophagosomes
with lysosomes 20. Ad-shDrp1 significantly depressed chloroquine-induced accumulation of LC3-II and
accumulation of p62/SQSTM1 did not change significantly after chloroquine treatment in the presence of
Ad-shDrp1 (Figure 2A), suggesting that Drp1 downregulation suppresses autophagic flux in CMs at
baseline.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
To further evaluate the effect of Drp1 downregulation upon autophagosome formation, CMs were
transduced with Ad-GFP-LC3. The number of GFP-LC3 dots was low at baseline and there was no
statistically significant difference between Ad-shScr- and Ad-shDrp1-transduced CMs (Figure 2B).
However, there was significantly less chloroquine-induced accumulation of GFP-LC3 dots in Ad-shDrp1transduced CMs than in Ad-shScr-transduced CMs (Figure 2B), further supporting the idea that Drp1
downregulation suppresses autophagic flux at baseline.
There were significantly more mitochondria, as evaluated with real-time PCR of cytochrome b
DNA and immunoblotting of COX IV, in Ad-shDrp1-transduced CMs than in control CMs (Figure 2CD),
suggesting that suppression of autophagy due to Drp1 downregulation leads to accumulation of
mitochondria. Peroxisome proliferator-activated receptor-gamma coactivator 1 (PGC-1 expression was
not significantly altered in Ad-shDrp1-transduced CMs (Online Figure IC), suggesting that mitochondrial
biogenesis was not affected. Since suppression of autophagy may impair mitochondrial quality control, we
evaluated the effect of Drp1 downregulation upon mitochondrial function.
Mitochondrial ATP production was significantly lower in CMs transduced with Ad-shDrp1 than in
those with Ad-shScr (Figure 2E). The effect of Drp1 downregulation upon mitochondrial membrane
potential was evaluated with JC-1. Drp1 knockdown led to the appearance of green JC-1 staining, indicating
depolarization of the mitochondrial membrane potential in CMs (Figure 2F). Furthermore, decreases in
absorbance at 540 nm in mitochondrial swelling assays, indicative of mPTP opening, were significantly
greater in CMs with Drp1 knockdown than in control CMs (Figure 2G), suggesting that mPTP opening is
accelerated by Drp1 downregulation. Cyclosporin A (CsA) attenuated CM death as evaluated with CellTiter
Blue® assays, suggesting that mPTP opening contributes to CM death in response to Drp1 downregulation
(Figure 2H).
We also evaluated the rate of oxidative phosphorylation in CMs, using a Seahorse analyzer (Online
Figure IIA). We normalized the oxygen consumption rate (OCR) with either mtDNA content or cell
viability in order to compensate for potential cell loss due to cell death. The basal OCR was significantly
lower in Drp1-downregulated CMs than in control CMs (Online Figure IIB). The OCR-linked ATP
synthesis, as evaluated with oligomycin treatment, and the maximum respiratory rate, as determined by
FCCP uncoupling, were also significantly lower in Drp1-downregulated CMs than in control CMs (Online
Figure IICD). The level of proton leak, determined by subtracting OCR-linked ATP synthesis from basal
OCR, did not significantly differ between Drp1-downregulated and control CMs (Online Figure IIE).
Together, these data indicate that Drp1 downregulation induces accumulation of mitochondria accompanied
by mitochondrial dysfunction in CMs.
DOI: 10.1161/CIRCRESAHA.116.303356 5
Drp1 mediates mitochondrial foreshortening in response to glucose deprivation.
We next investigated the involvement of Drp1 in mitochondrial dynamics in response to energy
stress. Drp1 was localized primarily in the cytosol in control CMs (Figure 3A). Glucose deprivation (GD),
which is known to affect mitochondrial dynamics in other cell types 12, 13, induced modest mitochondrial
accumulation of Drp1 in cultured CMs within 4 hours (Figure 3A), accompanied by a slight decrease in
cytosolic Drp1, although the reduction did not reach statistical significance. GD-induced mitochondrial
expression of Drp1 was also observed with anti-Drp1 immunostaining in mt-DsRed2 expressing CMs
(Figure 3B). These results suggest that GD increases Drp1 translocation from the cytosol to mitochondria
in CMs.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
After 1 hour of GD, the proportion of CMs with elongated mitochondria was increased, but that of
CMs with foreshortened mitochondria was also increased slightly in Ad-shScr-transduced CMs (Figure 3C).
A similar result was obtained in CMs transduced with adenovirus harboring LacZ (Ad-LacZ) (not shown).
More than 50% of Ad-shDrp1-transduced CMs exhibited elongated mitochondria after 1 hour of GD. After
4 hours of GD, however, approximately 15% of Ad-shScr- or Ad-LacZ-transduced CMs exhibited
foreshortened mitochondria, whereas more than 50% still showed elongated mitochondria and less than 1%
exhibited foreshortened mitochondria in Ad-shDrp1-transduced CMs (Figure 3C).Thus, although GD
induces transient mitochondrial elongation followed by foreshortening, Drp1 downregulation induces
persistent increases in elongation irrespective of GD. These results suggest that Drp1 plays an essential role
in mitochondrial foreshortening at baseline and during GD. Transduction with Ad-shDrp1 significantly
increased TUNEL-positive CMs after 1 and 4 hours of GD compared to transduction with Ad-shScr (Figure
3D), suggesting that endogenous Drp1 protects CMs against apoptosis during GD.
We evaluated the role of endogenous Drp1 in mediating autophagy in response to GD. Four hours
of GD significantly increased the number of GFP-LC3 dots in Ad-shScr-transduced CMs, but this increase
was significantly attenuated in Ad-shDrp1-transduced CMs (Figure 3E). To evaluate autophagic flux, CMs
were co-transduced with adenovirus harboring tandem fluorescent mRFP-GFP-LC3 (Ad-tf-LC3) 21. mRFP
(monomeric red fluorescent protein) retains its fluorescence but GFP loses its fluorescence in the acidic
environment of lysosomes. In merged images, the red puncta that overlay green puncta and appear yellow
indicate autophagosomes, whereas free red puncta indicate autolysosomes. After 4 hours of GD, the
numbers of both yellow and free red dots were increased in Ad-shScr-transduced CMs, indicating
stimulation of autophagic flux. However, the GD-induced increases were attenuated in Ad-shDrp1transduced CMs (Figure 3F), suggesting that Drp1 downregulation inhibits GD-induced autophagic flux.
Atg7 increases autophagic flux in CMs 22, 23. Drp1 downregulation significantly reduced Atg7-induced
increases in autophagosomes and autolysosomes at baseline and in response to GD in CMs (Figure
3G).Together, the data indicate that endogenous Drp1 plays an essential role in mediating mitochondrial
foreshortening, autophagy, and cell survival during GD in CMs.
Since Drp1 physically interacts with Bcl-xL in neurons 24 and Bcl-xL inhibits autophagy through
its binding to Beclin1 {Pattingre, 2005 #3365}, we investigated the involvement of Bcl-xL in the
suppression of autophagy by Drp1. Using co-immunoprecipitation assays, we confirmed that Drp1
physically interacts with Bcl-2 and Bcl-xL in CMs in the presence of Drp1 overexpression (Online Figure
IIIA). Increased expression of Drp1 inhibited, whereas downregulation of Drp1 augmented, the physical
interaction between Beclin1 and Bcl-2 or Bcl-xL (Online Figure IIIB). Downregulation of Drp1 decreased
the number of GFP-LC3 dots at baseline and in response to 4 hours of GD. However, the number of GFPLC3 dots increased significantly when Drp1 was downregulated in the presence of Bcl-xL downregulation
with or without chloroquine {Iwai-Kanai, 2008 #3728} (Online Figure IIICD). These results suggests that
downregulation of Drp1 inhibits autophagy through a Bcl-xL-dependent mechanism, most likely by
enhancing interaction between Beclin1 and Bcl-xL.
DOI: 10.1161/CIRCRESAHA.116.303356 6
Prolonged treatment with mdivi-1 mimics the effect of Drp1 downregulation.
Since previous studies showed that suppression of Drp1 by mdivi-1 protects CMs from cell death
, we investigated the effect of mdivi-1 upon mitochondrial morphology and cell death. Single treatment
with mdivi-1 at 50 or 100 M for 1 hour significantly increased the number of CMs with elongated
mitochondria at baseline. Mdivi-1 at 100 M also prevented foreshortening of mitochondria after 4 hours
of GD (Online Figure IVA). To compare the effects of Drp1 downregulation and mdivi-1 upon survival of
CMs side-by-side, CMs were treated with chelerythrine (10 M), an inducer of apoptosis 25, in the presence
or absence of either mdivi-1 or Ad-shDrp1. Although Ad-shDrp1 transduction for 96 hours decreased CM
survival at baseline and in response to chelerythrine, mdivi-1 treatment for 1 hour increased CM survival
at baseline and in response to chelerythrine compared to vehicle treatment (Online Figure IVB). Mdivi-1
treatment at 50 M did not significantly affect CM viability in response to GD, but 100 M significantly
reduced it (Online Figure IVC). Taken together, these results suggest that a single treatment with mdivi-1
has direct cell-protective effects upon CMs independent of Drp1. However, a higher dose of mdivi-1
partially mimics the effect of Drp1 downregulation even after a single application.
11
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Treatment of CMs with 50 M mdivi-1 every 24 hours for 1 week induced elongation of
mitochondria at baseline and inhibited foreshortening of mitochondria even after 4 hours of GD (Online
Figure IVD). Prolonged treatment with mdivi-1 time-dependently decreased CM viability compared to
vehicle treatment (Online Figure IVE) and significantly suppressed GD-induced autophagic flux as
evaluated with mRFP-GFP-LC3 (Online Figure IVF). Thus, prolonged treatment with mdivi-1 mimics the
effect of Drp1 downregulation.
Drp1 mediates autophagic removal of mitochondria.
We investigated whether clearance of mitochondria is regulated by Drp1 using mitochondriatargeted Keima fluorescence 19. Keima has a bimodal excitation spectrum peaking at 438±12 and 550±15
nm, corresponding to neutral and acidic pH states, respectively 19. Because fusion of autophagosomes with
lysosomes exposes the autophagosome contents to acidic pH, the maturation of autophagosomes to
autolysosomes can be monitored by measuring Keima fluorescence 19. We confirmed that Keima with a
mitochondria-localization signal (Keima-MLS) is expressed in CM mitochondria (Figure 4A). Puncta with
a high ratio of excitation at 560/440 nm (high 560/440) colocalized with Alexa 488 Dextran, reflecting
increased lysosomal localization of Keima-MLS, after treatment with 25
M of cyanide 3chlorophenylhydrazone (CCCP), a mitochondrial uncoupler, for 16 hours to induce mitochondrial
autophagy 26 (Figure 4B, Online Figure VA), confirming that Keima-MLS works as expected in CMs.
Puncta with high 560/440, indicating the presence of mitochondria in lysosomes, were significantly
increased after 4 hours of GD in CMs transduced with Ad-shScr, but not in CMs transduced with AdshDrp1 (Figure 4C). This increase was abolished in the presence of Ad-shBeclin1-mediated Beclin1
downregulation (Online Figure VB), suggesting that it is mediated by autophagy and that Drp1 is necessary
for stimulating autophagic mitochondrial degradation. Interestingly, downregulation of Beclin1 did not
affect GD-induced increases in mitochondrial foreshortening (Online Figure VC) but significantly
increased GD-induced cell death (Online Figure VD). Thus, although evidence suggests that unopposed
fusion of mitochondria alone can induce cell death 27, suppression of autophagy alone may also induce CM
death even when mitochondrial foreshortening is not affected.
Atg7 overexpression, which is known to stimulate autophagy 22, 23, failed to increase puncta with
high 560/440 in Drp1-downregulated CMs (Figure 4D), even though it increased autophagosomes and
autolysosomes in this condition (Figure 3FG), nor did it inhibit Drp1 knockdown-induced cell death (Figure
4E).
DOI: 10.1161/CIRCRESAHA.116.303356 7
To further elucidate the role of endogenous Drp1 in autophagy, CMs were subjected to GD in the
presence or absence of Drp1 knockdown and electron microscopic (EM) analyses were conducted (Figure
4F). Drp1 downregulation significantly reduced the number of mitochondria and increased relative
mitochondria mass at baseline (Figure 4G). Drp1 downregulation also decreased the total number of
autophagosomes at baseline and in response to GD and decreased the number of autophagosomes
selectively containing mitochondria (Figure 4H). These results suggest that endogenous Drp1 is important
in mediating both general autophagy, including mitochondrial autophagy.
Forced Drp1 overexpression induces apoptosis in CMs.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Adenovirus-mediated overexpression of Drp1 by 5 fold, which is higher than the level induced by
GD, induced foreshortening of mitochondria in CMs at baseline (Online Figure VIAB). Under this
condition, increases in apoptosis, decreases in mitochondrial DNA, and decreases in mitochondrial
membrane potential were observed (Online Figure VIC-E). These results suggest that persistent and highlevel expression of Drp1 induces mitochondrial dysfunction and apoptosis in CMs. Drp1 overexpression
significantly increased Keima-MLS puncta with high 560/440 in CMs both at baseline and after 4 hours of
GD (Online Figure VIF), indicating stimulation of lysosomal removal of mitochondria. Interestingly,
suppression of autophagy by Ad-shAtg7 attenuated the increased cell death induced by Drp1
overexpression (Online Figure VIG), suggesting that excessive activation of autophagy by Drp1 may induce
cell death.
Basal characterization of Drp1-CKO mice.
To evaluate the role of endogenous Drp1 in vivo, we used loss-of-function mouse models. No
homozygous mice were born during attempts to generate cardiac-specific Drp1 knockout mice using
MHC-Cre mice. Therefore, in order to examine the effect of Drp1 on cardiac function in adult mice in
vivo, we generated cardiac-specific conditional Drp1 knockout (Drp1-CKO) mice by crossing Drp1 flox
homo (fl/fl) and MHC-MerCreMer mice, and Drp1 expression was downregulated in a tamoxifendependent manner. We used Drp1-CKO without tamoxifen injection (TI) and Drp1 fl/fl with or without TI
as controls. Fifteen-week-old male mice were subjected to TI (20 mg/kg, ip) for 5 days. Four and 8 weeks
after TI, we measured cardiac function and performed biochemical and histological analyses (Online Figure
VIIA). Immunoblot analyses confirmed that cardiac Drp1 levels were significantly lower in Drp1-CKO
mice than in control mice (Figure 5A) and that Drp1 was downregulated in a heart-specific manner in Drp1CKO mice (Online Figure VIIB). Cardiac levels of other proteins involved in mitochondrial dynamics, such
as Mfn1, Mfn2, OPA1, and Fis1, were unaltered in Drp1-CKO mice compared to in control mice (Online
Figure VIIC). Drp1-CKO mice started to die 8 weeks after TI and all died by 13 weeks after injection,
whereas no control mice died during the observation period of 16 weeks following TI. Kaplan–Meier
analysis revealed that the survival rate was significantly lower in Drp1-CKO mice than in control mice
(Online Figure VIID). Four weeks after TI, the hearts of Drp1-CKO mice were enlarged compared to
control hearts (Figure 5B). Postmortem assessment showed that both left ventricular (LV) weight/tibial
length, an index of LV hypertrophy, and lung weight/tibial length, an index of lung congestion, were
significantly greater in Drp1-CKO than in control mice 4 and 8 weeks after TI (Online Tables I and II).
Wheat germ agglutinin (WGA) staining of LV sections 4 and 8 weeks after TI showed that CM crosssectional area was significantly greater in Drp1-CKO than in control mice (Figure 5C and Online Figure
VIIE). Myocardial fibrosis, as evaluated with Picric Acid Sirius Red and Masson’s Trichrome staining, was
also significantly greater in Drp1-CKO mice than in control mice (Figure 5D and Online Figure VIIFG).
Echocardiographic measurements 4 and 8 weeks after TI showed that the LV diastolic dimension was
significantly greater, and the LV ejection fraction (LVEF), an indicator of LV systolic function, was lower
in Drp1-CKO mice than in control mice (Online Tables III and IV). Hemodynamic measurements at 4
weeks after TI showed that LV +dP/dt was decreased, whereas LV end-diastolic pressure was significantly
elevated in Drp1-CKO compared to in control mice (Online Table II). We confirmed that MHCDOI: 10.1161/CIRCRESAHA.116.303356 8
MerCreMer alone did not influence cardiac function or histology in either the presence or absence of
tamoxifen (Online Figure VIIF-H). Taken together, these results suggest that Drp1 downregulation induces
LV dysfunction and cardiac hypertrophy at baseline.
Drp1 downregulation induces mitochondrial elongation and dysfunction.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
To examine how Drp1 deletion affects mitochondrial morphology in the heart, EM analysis was
performed. At baseline, mitochondria in control mouse hearts were primarily rectangular or spherical in
shape, whereas tubular mitochondria were observed less frequently. On the other hand, mitochondria in
Drp1-CKO mice were mostly elongated/enlarged 4 and 8 weeks after TI (Figure 6A and Online Figure
VIIIA). After 48 hours of fasting, mitochondria in control mouse hearts became smaller and spherical. In
contrast, mitochondria in Drp1-CKO mice remained elongated even after fasting (Figure 6A).
Autophagosomes containing mitochondria were observed in control mouse hearts after 48 hours of fasting,
but not in Drp1-CKO mouse hearts (Figure 6A). Quantitative analysis revealed that mitochondrial mass
was significantly greater in Drp1-CKO mouse hearts than in control mouse hearts at baseline and after
fasting (Figure 6A and Online Figure VIIIA). These results suggest that endogenous Drp1 plays an essential
role in mediating mitochondrial foreshortening at baseline and during fasting in the mouse heart in vivo.
Four or 8 weeks after TI, Drp1 depletion increased the COX IV protein level (Figure 6B, Online
Figure VIIIB), and mitochondrial DNA content, evaluated with real-time PCR of cytochrome b DNA, was
significantly greater in Drp1-CKO mice than in control mice (Figure 6C). These results suggest that the
mitochondrial content is increased by Drp1 downregulation. Mitochondrial biogenesis was evaluated by
immunoblot analyses of PGC-1 and mitochondrial transcription factor A (TFAM). Cardiac protein
expression of PGC-1 and TFAM did not differ between Drp1-CKO and control mice 4 and 8 weeks after
TI, suggesting that Drp1 depletion did not affect mitochondrial biogenesis (Figure 6D and Online Figure
VIIIC). However, mitochondrial ATP production was significantly attenuated in Drp1-CKO mouse hearts
4 and 8 weeks after TI compared to in control mouse hearts (Figure 6E and Online Figure VIIID). The
activity of mitochondrial complexes I, II + III, and IV was also significantly attenuated in Drp1-CKO mouse
hearts 8 weeks after TI compared to in control mouse hearts (Online Figure VIIIE). The extent of mPTP
opening, as evaluated by the decrease in absorbance at 540 nm in mitochondrial swelling assays, was
significantly greater in Drp1-CKO mouse hearts 4 weeks after TI than in controls (Figure 6F), suggesting
that mPTP opening is accelerated in Drp1-CKO mice. The cardiac level of 4-Hydroxynonenal, a marker of
oxidative stress, and mitochondrial production of H2O2, evaluated with Amplex® Red assays, were also
significantly higher in Drp1-CKO mice 4 weeks after TI than in control mice (Figure 6GH). Thus, Drp1
depletion results in mitochondrial dysfunction and oxidative stress in the heart.
Since the initial assessment of mitochondrial function was conducted using hearts harvested 4-8
weeks after TI, when both hypertrophy and LV dysfunction are obvious in Drp1-CKO mice, mitochondrial
dysfunction could be secondary to pathological hypertrophy. We therefore also investigated an earlier time
point. Echocardiographic analyses revealed no significant difference in LVEF between control and Drp1CKO mice 10 days after TI (Online Table V), nor was there a significant difference in CM cross-sectional
area or cardiac fibrosis (Online Figure VIIIFG), confirming that this time point precedes the development
of pathological hypertrophy. Nevertheless, mitochondrial function, as assessed by ATP production and
mitochondrial swelling assays, was already severely attenuated in Drp1-CKO mice compared to in control
mice 10 days after TI (Figure 6EF). Together with the observation that Drp1 downregulation directly
induces mitochondrial dysfunction in cultured CMs (Figure 2E-J), these results suggest that Drp1 depletion
induces mitochondrial dysfunction in the heart even before manifestation of hypertrophy and LV
dysfunction.
We investigated whether Drp1 downregulation in the heart affects apoptosis. There were
significantly more TUNEL-positive nuclei in Drp1-CKO mouse hearts than in controls 4 and 8 weeks after
DOI: 10.1161/CIRCRESAHA.116.303356 9
TI (Figure 7A and Online Figure VIIIH). Cleaved caspase-3 and cytochrome c release into the cytosolic
fraction were also significantly elevated in Drp1-CKO mouse hearts 4 weeks after TI (Figure 7B), as was
the serum HMGB1 level, an indicator of necrosis (Online Figure VIII I). These results suggest that
endogenous Drp1 is required for protection against the mitochondrial mechanisms of apoptosis and necrosis
in CMs.
Autophagy is inhibited in Drp1-CKO mice.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
We next investigated the role of Drp1 in mediating autophagy in the heart in vivo. There was
significantly less LC3-II and significantly more p62 in Drp1-CKO mouse hearts than in controls 4 weeks
after TI (Figure 7C). To examine whether Drp1 downregulation attenuates autophagic flux, we evaluated
the effect of chloroquine injection upon autophagosome accumulation 20. LC3-II accumulation was
suppressed even in the presence of chloroquine, whereas p62/SQSTM1 accumulation did not change
significantly after chloroquine treatment in the Drp1-CKO mouse heart (Figure 7D). To further evaluate
the level of autophagic flux in CMs in vivo, we crossed Drp1-CKO and Drp1 fl/fl with cardiac-specific
mRFP-GFP-LC3 (tf-LC3) transgenic mice. Both Drp1-CKO X tf-LC3 and Drp1 fl/fl X tf-LC3 (control tfLC3) were injected with tamoxifen for 5 days. Fasting increased the number of LC3 dots with both green
and red color (appearing yellow in merged images), representing autophagosomes, as well as the number
of dots with only red color, representing autolysosomes, in control tf-LC3 mice, indicating increased
autophagic flux (Figure 7E). In contrast, the number of yellow and free red dots did not increase in response
to fasting in Drp1-CKO X tf-LC3 mice (Figure 7E). Taken together, these results suggest that Drp1
downregulation suppresses autophagic flux at baseline.
Drp1 depletion induces stress intolerance and enhances I/R injury.
Although mitochondrial fission and fusion are essential for maintaining mitochondrial quality
control, their role in cardiac development and stress resistance remains unknown. To address this question,
we crossed Drp1 fl/fl mice with MHC-Cre mice. Although no mice with cardiac-specific homozygous
Drp1 knockout were born, mice with cardiac-specific heterozygous Drp1 knockout (Drp1-hetCKO) were
viable at 12 weeks, suggesting that Drp1 is required for normal prenatal development but that one functional
allele is sufficient during this period. Cardiac Drp1 expression was 40% lower in Drp1-hetCKO mice than
in control (Drp1 flox/+) mice (Figure 8A). LV function, assessed by LVEF, in Drp1-hetCKO mice was
normal at 12 weeks of age (Figure 8B). Neither LV weight/tibial length nor lung weight/tibial length
differed between 12-week-old Drp1-hetCKO and control mice (Online Table VI). Histological analyses
showed that the CM cross-sectional area and myocardial fibrosis also did not differ between 12-week-old
Drp1-hetCKO and control mice (Online Figure IXAB). However, ATP production was significantly lower
in 12-week-old Drp1-hetCKO mice (Figure 8C), suggesting that mitochondrial dysfunction develops prior
to histological and hemodynamic changes in Drp1-hetCKO mice. The fact that LV function is maintained
at 12 weeks of age in Drp1-hetCKO mice allowed us to use these mice to examine the role of Drp1 during
stress in the heart.
Mitochondrial Drp1 was significantly increased in response to 48-hour fasting or I/R but not in
Drp1-hetCKO mice (Figure 8D). To evaluate the role of endogenous Drp1 in protection against stress in
vivo, 12-week-old Drp1-hetCKO and control mice underwent 48-hour fasting. The LVEF was significantly
lower in Drp1-hetCKO mice than in control mice after fasting, suggesting that endogenous Drp1 acts to
preserve LV function during fasting (Figure 8E). Similar results were observed in Drp1-CKO mice with
tamoxifen treatment (Online Figure IXC). To evaluate the role of endogenous Drp1 in protection against
I/R, 12-week-old Drp1-hetCKO and control mice were subjected to 30 minutes of myocardial ischemia
followed by 24 hours of reperfusion. EM analyses showed that I/R increased the number of smaller and
spherical mitochondria in control mice, suggesting that mitochondrial fission was induced. However, these
changes were significantly attenuated in Drp1-hetCKO mice (Figure 8F), suggesting that endogenous Drp1
DOI: 10.1161/CIRCRESAHA.116.303356 10
mediates mitochondrial fission after I/R. Autophagosomes containing mitochondria were observed in
control mouse hearts but not in Drp1-hetCKO mouse hearts after I/R (Figure 8F). There was also
significantly less LC3-II and more p62 in Drp1-hetCKO mouse hearts than in control hearts at baseline and
after I/R (Figure 8G), suggesting that autophagy is suppressed by heterozygous Drp1 downregulation. The
infarct size/area at risk (MI/AAR) after I/R, as evaluated with Alcian Blue and tetrazolium chloride staining,
was not affected by MHC-Cre alone (Online Figure IXD) but was significantly greater in Drp1-hetCKO
mice than in control mice (55.2 ± 3.0 vs. 40.2 ± 1.6%, p<0.05, n=3 per group, Figure 8H). Similar results
were observed in Drp1-CKO mice with tamoxifen treatment (Online Figure IXE).Taken together, these
results suggest that inhibition of mitochondrial fission through Drp1 downregulation enhances myocardial
injury in response to I/R.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
We also evaluated the effect of mdivi-1 upon I/R injury. One-time treatment with mdivi-1 just
before I/R significantly reduced the MI/AAR (Online Figure XA), confirming previous observations by
others 11. However, the same treatment also reduced the MI/AAR in Drp1-hetCKO mice, suggesting that
short term treatment with mdivi-1 protects the heart through Drp-1-independent mechanisms (Online Figure
XB). Although repetitive applications of mdivi-1 (1.2 mg/kg/day for 7 days) did not significantly reduce
LV systolic function (Online Figure XC and Online Table VI), it significantly increased mitochondrial mass,
as determined by EM (Online Figure XD), reduced mitochondrial function to a similar extent as
heterozygous Drp1 downregulation, as determined by mitochondrial swelling assays and ATP production
(Online Figures XEF), and significantly enhanced the MI/AAR after I/R (Online Figure XG), thereby
mimicking the effect of Drp1-hetCKO. Thus, while the effects of long-term treatment with mdivi-1 are
similar to those of Drp1 downregulation with regards to I/R injury enhancement, albeit weaker, one-time
treatment with mdivi-1 appears to have protective effects, which are most likely independent of Drp1.
DISCUSSION
Our results suggest that endogenous Drp1 induces mitochondrial foreshortening at baseline and in
response to stress in the heart and the CMs therein. Contrary to previous reports 11, 28, downregulation of
endogenous Drp1 in CMs induces mitochondrial dysfunction and apoptosis despite significant induction of
mitochondrial elongation, thereby inducing cardiac dysfunction at baseline and exacerbating myocardial
injury in response to I/R. Using Keima-MLS, we show that Drp1 plays an essential role in mediating
lysosomal removal of mitochondria in CMs. Thus, our results suggest that endogenous Drp1 contributes to
mitochondrial quality control.
Although it is generally believed that fused mitochondria function better, Drp1 downregulation
significantly increased the number of CMs with depolarized mitochondria even at baseline. Although Drp1
is localized primarily in the cytosol in unstimulated CMs, a low level of mitochondrial turnover mediated
by Drp1 appears essential to maintain mitochondrial function in CMs. Given that even heterozygous Drp1
downregulation induces mitochondrial dysfunction and heart failure in mice, it appears that endogenous
Drp1 plays an essential role in mitochondrial quality control in the heart in vivo as well.
Whether mitochondria undergo fusion or fission during stress may depend upon cell type and stress.
In MEF cells 12, 13, fasting induces mitochondrial fusion induced by phosphorylation of Drp1 at Ser637 by
protein kinase A and translocation of Drp1 to the cytoplasm, which allows mitochondria to maintain ATP
synthesis and escape autophagic destruction 12, 13. On the other hand, in HL1 cells in vitro and CMs in the
heart in vivo 11, 29, fasting and hypoxia stimulate mitochondrial fission. Regardless of whether fusion or
fission is stimulated by stress, these studies showed that suppression of fission and/or stimulation of fusion
through Drp1 downregulation, expression of dominant-negative Drp1, mdivi-1, or expression of Mfn1/2
promotes ATP production and cell survival. Here we show that mitochondria in CMs transiently undergo
DOI: 10.1161/CIRCRESAHA.116.303356 11
elongation during GD, but that the number of mitochondria with foreshortening also increases thereafter,
accompanied by accumulation of Drp1 in mitochondria. Drp1 downregulation in this scenario blunted
foreshortening of mitochondria and exacerbated cell death, suggesting that the induction of foreshortening
is adaptive in CMs.
Our results suggest that endogenous Drp1 is important in mediating autophagy in CMs. Drp1
controls autophagic flux at least at the level of autophagosome formation, since there were fewer GFP-LC3
puncta when Drp1 was downregulated in the presence of chloroquine, an inhibitor of autophagosomelysosome fusion or autophagic flux 20. The suppressive effect of Drp1 downregulation upon global
autophagy, rather than its specific effect upon mitochondria specific autophagy, was unexpected. We here
show that Drp1 physically interacts with Bcl-2/Bcl-xL and that downregulation of Drp1 promotes
interaction between Beclin1 and Bcl-2/Bcl-xL. Since Bcl-2 and Bcl-xL are endogenous inhibitors of
Beclin1 {Pattingre, 2005 #3365}, increased interaction between Beclin1 and Bcl-2/Bcl-xL in the presence
of Drp1 downregulation should lead to suppression of autophagy. In fact, the suppression of general
autophagy by Drp1 downregulation was rescued by downregulation of Bcl-xL, indicating the critical role
of the Bcl-2 family proteins in this process.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
We here show that a GD-induced increase in lysosomal localization of Keima-MLS 19 is attenuated
in the presence of Drp1 downregulation. Given the mitochondrial localization of Keima-MLS, and that
Keima-MLS puncta with high 560/440, indicating acidic pH, are localized in lysosomes and are abolished
when Beclin1 is downregulated, increases in Keima-MLS puncta with high 560/440 presumably reflect
autophagic degradation of mitochondria. Thus, the significant reduction in lysosomal Keima-MLS puncta,
together with EM images showing a significant reduction in autophagosomes primarily containing
mitochondria, in Drp1 knockdown CMs indicates that endogenous Drp1 plays an essential role in mediating
GD-induced increases in mitochondrial autophagy. The Keima-MLS analysis was not sensitive enough to
demonstrate a reduction in lysosomal removal of mitochondria at baseline when Drp1 is downregulated.
However, given that dysfunctional mitochondria accumulate in Drp1-downregulated CMs, it is likely that
Drp1 also mediates autophagic mitochondrial degradation at baseline.
In this work, we used the term “mitochondrial autophagy” to describe the clearance of mitochondria
by autophagy. Although our results suggest that Drp1 regulates mitochondrial clearance through general
autophagy, whether or not Drp1 also affects mitochondria-selective autophagy, namely mitophagy, could
not be evaluated due to technical limitations. To this end, specific assays to accurately evaluate the presence
of mitophagy and/or specific interventions to modulate mitophagy appear essential.
Conditional Drp1 downregulation leads to decreases in cardiac function within 4 weeks and all
animals died within 13 weeks due to heart failure. Histological analyses showed that the Drp1 deficiency
induces hypertrophy and fibrosis in the heart and increases CM apoptosis. The fact that conditional cardiacspecific combined downregulation of Mfn1 and Mfn2 (c-Mfn1/2-KO) also leads to rapid development of
cardiac dysfunction within 2 weeks 9 indicates that both unopposed fission and unopposed fusion of
mitochondria may cause cardiac dysfunction and suggests the critical importance of mitochondrial
remodeling in the heart.
There are some differences between the cardiac phenotypes of Drp1-CKO and c-Mfn1/2-KO mice
. For example, neither cardiac hypertrophy nor the increased CM apoptosis observed in Drp1-CKO were
apparent in c-Mfn1/2-KO mice. This suggests that ATP depletion may be more profound in the absence of
fusion than in the absence of fission.
9, 10
The reason for the opposite effects of Drp1 downregulation by genetic deletion and Drp1
suppression with mdivi-1 in response to I/R remains to be elucidated. One possibility is that our shRNA
treatment may have induced stronger, more prolonged suppression of mitochondrial fission than a single
DOI: 10.1161/CIRCRESAHA.116.303356 12
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
dose of mdivi-1 at 50 M, the concentration used by others10. We observed modest cell death-promoting
effects when CMs were treated with mdivi-1 at a higher concentration (100 M) or multiple times. A
second possibility is that Drp1 downregulation may induce more potent suppression of general autophagy
to even below physiological levels than mdivi-1. Although suppression of excessive autophagy may be
salutary, suppression below physiological levels may be harmful. Third, mdivi-1 may more strongly
suppress cell death than Drp1 downregulation by directly acting upon apoptosis mechanisms. Mdivi-1
blocks Bax/Bak-dependent release of both Smac/Diablo and cytochrome c in HeLa cells 14, and we found
that mdivi-1 inhibited chelerythrine-induced apoptosis in CMs, which Drp1 downregulation did not.
Furthermore, one-time treatment with mdivi-1 reduced I/R injury even in Drp1-hetCKO mice, suggesting
that mdivi-1 most likely has a Drp1-independent anti-apoptotic function. Along this same line, mdivi-1
affects other molecules besides Drp1, including delayed rectifier K+ channels 27, raising the issue of
specificity of the chemical inhibitor. Fourth, mitochondrial localization of Drp1 is positively regulated by
protein kinase A 30, calcineurin 30, PUMA 31, Bax/Bak 32, ceramide 33, and O-linked--N-acetylglucosamine
modification 34, and is negatively regulated by miR-499 35 and Pim1 36. Thus, some experimental conditions
may induce excessive Drp1 activation/upregulation, which may in turn induce deleterious effects in CMs.
In fact, Drp1 overexpression in CMs above the level caused by GD induced cell death. Drp1 suppression
by mdivi-1 may be protective under such experimental conditions.
We have shown previously that Beclin1 haploinsufficiency inhibits I/R injury and suppresses
autophagy 37. Here we show that Drp1 haploinsufficiency exacerbates I/R injury, but is also accompanied
by suppression of autophagy. Currently, mechanisms explaining the difference remain to be clarified. Drp1
downregulation may have a more pronounced effect upon general autophagy and/or mitochondrial
autophagy than Beclin1 downregulation, thereby suppressing autophagy below physiological levels.
Another possibility is that Drp1 downregulation may more globally affect mitochondrial quality control
mechanisms, including inducing unopposed mitochondrial elongation and suppression of global autophagy,
rather than being limited to suppression of autophagic mitochondrial degradation. Further investigation is
required to address this issue.
In summary, persistent Drp1 downregulation inhibits clearance of mitochondria by autophagy and
causes mitochondrial dysfunction and consequent cell death in the heart and in the CMs therein, both at
baseline and under stress conditions. Drp1 plays an important role in mediating mitochondrial
foreshortening and autophagic mitochondrial degradation in CMs.
ACKNOWLEDGEMENTS
The authors wish to thank Christopher D. Brady and Daniela Zablocki for critical reading of the manuscript
and Luke Fritzky for technical assistance.
SOURCES OF FUNDING
This work was supported in part by U.S. Public Health Service Grants HL102738, HL67724, HL69020,
HL91469, AG23039, and AG27211. This work was also supported by the Fondation Leducq Transatlantic
Networks of Excellence. IY has been supported by a Postdoctoral Fellowship from the Founders Affiliate,
American Heart Association, and by a grant from the Rotary Foundation Ambassadorial Scholarship.
DISCLOSURES
None.
DOI: 10.1161/CIRCRESAHA.116.303356 13
REFERENCES
1.
2.
3.
4.
5.
6.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
Gottlieb RA, Gustafsson AB. Mitochondrial turnover in the heart. Biochim Biophys Acta.
2011;1813:1295-1301.
Dorn GW, 2nd. Mitochondrial dynamics in heart disease. Biochim Biophys Acta. 2012;1833:233241.
Piquereau J, Caffin F, Novotova M, Lemaire C, Veksler V, Garnier A, Ventura-Clapier R, Joubert
F. Mitochondrial dynamics in the adult cardiomyocytes: which roles for a highly specialized cell?
Front Physiol. 2013;4:102.
Song M, Chen Y, Gong G, Murphy E, Rabinovitch PS, Dorn GW, 2nd. Super-suppression of
mitochondrial reactive oxygen species signaling impairs compensatory autophagy in primary
mitophagic cardiomyopathy. Circ Res. 2014;115:348-353.
Youle RJ, van der Bliek AM. Mitochondrial fission, fusion, and stress. Science. 2012;337:10621065.
Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh SE, Katz S,
Las G, Alroy J, Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, Shirihai OS. Fission and selective
fusion govern mitochondrial segregation and elimination by autophagy. Embo J. 2008;27:433-446.
Otera H, Mihara K. Molecular mechanisms and physiologic functions of mitochondrial dynamics.
J Biochem.149:241-251.
Okamoto K, Shaw JM. Mitochondrial morphology and dynamics in yeast and multicellular
eukaryotes. Annu Rev Genet. 2005;39:503-536.
Chen Y, Liu Y, Dorn GW, 2nd. Mitochondrial fusion is essential for organelle function and cardiac
homeostasis. Circ Res. 2011;109:1327-1331.
Papanicolaou KN, Kikuchi R, Ngoh GA, Coughlan KA, Dominguez I, Stanley WC, Walsh K.
Mitofusins 1 and 2 are essential for postnatal metabolic remodeling in heart. Circ Res.
2012;111:1012-1026.
Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial
fission protects the heart against ischemia/reperfusion injury. Circulation. 2010;121:2012-2022.
Gomes LC, Di Benedetto G, Scorrano L. During autophagy mitochondria elongate, are spared from
degradation and sustain cell viability. Nat Cell Biol. 2011;13:589-598.
Rambold AS, Kostelecky B, Elia N, Lippincott-Schwartz J. Tubular network formation protects
mitochondria from autophagosomal degradation during nutrient starvation. Proc Natl Acad Sci U
S A. 2011;108:10190-10195.
Cassidy-Stone A, Chipuk JE, Ingerman E, Song C, Yoo C, Kuwana T, Kurth MJ, Shaw JT,
Hinshaw JE, Green DR, Nunnari J. Chemical inhibition of the mitochondrial division dynamin
reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization. Dev Cell.
2008;14:193-204.
Tanaka A, Cleland MM, Xu S, Narendra DP, Suen DF, Karbowski M, Youle RJ. Proteasome and
p97 mediate mitophagy and degradation of mitofusins induced by Parkin. J Cell Biol.
2010;191:1367-1380.
Lee Y, Lee HY, Hanna RA, Gustafsson AB. Mitochondrial autophagy by Bnip3 involves Drp1mediated mitochondrial fission and recruitment of Parkin in cardiac myocytes. Am J Physiol Heart
Circ Physiol. 2011;301:H1924-1931.
Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda K, Otera H, Nakanishi Y, Nonaka
I, Goto Y, Taguchi N, Morinaga H, Maeda M, Takayanagi R, Yokota S, Mihara K. Mitochondrial
fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat Cell
Biol. 2009;11:958-966.
Hariharan N, Zhai P, Sadoshima J. Oxidative Stress Stimulates Autophagic Flux during
Ischemia/Reperfusion. Antioxid Redox Signal. 2010.
DOI: 10.1161/CIRCRESAHA.116.303356 14
19.
20.
21.
22.
23.
24.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
Katayama H, Kogure T, Mizushima N, Yoshimori T, Miyawaki A. A sensitive and quantitative
technique for detecting autophagic events based on lysosomal delivery. Chem Biol. 2011;18:10421052.
Iwai-Kanai E, Yuan H, Huang C, Sayen MR, Perry-Garza CN, Kim L, Gottlieb RA. A method to
measure cardiac autophagic flux in vivo. Autophagy. 2008;4.
Hariharan N, Maejima Y, Nakae J, Paik J, Depinho RA, Sadoshima J. Deacetylation of FoxO by
Sirt1 Plays an Essential Role in Mediating Starvation-Induced Autophagy in Cardiac Myocytes.
Circ Res. 2010;107:1470-1482.
Pattison JS, Osinska H, Robbins J. Atg7 induces basal autophagy and rescues autophagic deficiency
in CryABR120G cardiomyocytes. Circ Res. 2011;109:151-160.
Sciarretta S, Zhai P, Shao D, Maejima Y, Robbins J, Volpe M, Condorelli G, Sadoshima J. Rheb
is a Critical Regulator of Autophagy during Myocardial Ischemia: Pathophysiological Implications
in Obesity and Metabolic Syndrome. Circulation. 2012;125:1134-1146.
Li H, Alavian KN, Lazrove E, Mehta N, Jones A, Zhang P, Licznerski P, Graham M, Uo T, Guo J,
Rahner C, Duman RS, Morrison RS, Jonas EA. A Bcl-xL-Drp1 complex regulates synaptic vesicle
membrane dynamics during endocytosis. Nat Cell Biol. 2013;15:773-785.
Yamamoto S, Seta K, Morisco C, Vatner SF, Sadoshima J. Chelerythrine Rapidly Induces
Apoptosis through Generation of Reactive Oxygen Species in Cardiac Myocytes. J Mol Cell
Cardiol. 2001;33:1829-1848.
Matsuda N, Sato S, Shiba K, Okatsu K, Saisho K, Gautier CA, Sou YS, Saiki S, Kawajiri S, Sato
F, Kimura M, Komatsu M, Hattori N, Tanaka K. PINK1 stabilized by mitochondrial depolarization
recruits Parkin to damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol.
2010;189:211-221.
Bhandari P, Song M, Chen Y, Burelle Y, Dorn GW, 2nd. Mitochondrial contagion induced by
Parkin deficiency in Drosophila hearts and its containment by suppressing mitofusin. Circ Res.
2014;114:257-265.
Disatnik MH, Ferreira JC, Campos JC, Gomes KS, Dourado PM, Qi X, Mochly-Rosen D. Acute
inhibition of excessive mitochondrial fission after myocardial infarction prevents long-term cardiac
dysfunction. J Am Heart Assoc. 2013;2:e000461.
Kim H, Scimia MC, Wilkinson D, Trelles RD, Wood MR, Bowtell D, Dillin A, Mercola M, Ronai
ZA. Fine-tuning of Drp1/Fis1 availability by AKAP121/Siah2 regulates mitochondrial adaptation
to hypoxia. Mol Cell.44:532-544.
Cribbs JT, Strack S. Reversible phosphorylation of Drp1 by cyclic AMP-dependent protein kinase
and calcineurin regulates mitochondrial fission and cell death. EMBO Rep. 2007;8:939-944.
Wang JX, Li Q, Li PF. Apoptosis repressor with caspase recruitment domain contributes to
chemotherapy resistance by abolishing mitochondrial fission mediated by dynamin-related protein1. Cancer Res. 2009;69:492-500.
Wasiak S, Zunino R, McBride HM. Bax/Bak promote sumoylation of DRP1 and its stable
association with mitochondria during apoptotic cell death. J Cell Biol. 2007;177:439-450.
Parra V, Eisner V, Chiong M, Criollo A, Moraga F, Garcia A, Hartel S, Jaimovich E, Zorzano A,
Hidalgo C, Lavandero S. Changes in mitochondrial dynamics during ceramide-induced
cardiomyocyte early apoptosis. Cardiovasc Res. 2008;77:387-397.
Gawlowski T, Suarez J, Scott B, Torres-Gonzalez M, Wang H, Schwappacher R, Han X, Yates JR,
3rd, Hoshijima M, Dillmann W. Modulation of dynamin-related protein 1 (DRP1) function by
increased O-linked-beta-N-acetylglucosamine modification (O-GlcNAc) in cardiac myocytes. J
Biol Chem. 2012;287:30024-30034.
Wang JX, Jiao JQ, Li Q, Long B, Wang K, Liu JP, Li YR, Li PF. miR-499 regulates mitochondrial
dynamics by targeting calcineurin and dynamin-related protein-1. Nat Med. 2011;17:71-78.
Din S, Mason M, Volkers M, Johnson B, Cottage CT, Wang Z, Joyo AY, Quijada P, Erhardt P,
Magnuson NS, Konstandin MH, Sussman MA. Pim-1 preserves mitochondrial morphology by
inhibiting dynamin-related protein 1 translocation. Proc Natl Acad Sci U S A.110:5969-5974.
DOI: 10.1161/CIRCRESAHA.116.303356 15
37.
Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, Levine B, Sadoshima J. Distinct
Roles of Autophagy in the Heart During Ischemia and Reperfusion. Roles of AMP-Activated
Protein Kinase and Beclin 1 in Mediating Autophagy. Circ Res. 2007;100:914-922.
FIGURE LEGENDS
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Figure 1. Drp1 mediates mitochondrial fission and CM survival. A, Assessment of mitochondrial
morphology using mitochondria-targeted DsRed2 (mt-DsRed2). TnI: Troponin-I. TnI staining indicates
CM. The proportions of CMs with elongated and foreshortened mitochondria was quantified. Gray bar:
cells with elongated/total cell number, black bar: cells with foreshortened/total cell number, white bar: cells
with intermediate (mid) mitochondria/total cell number. * p<0.01 vs. foreshortened in Ad-shScr, # p<0.01
vs. elongated in Ad-shScr (n=4/group). Scale bar: 20 m. B, TUNEL staining of CMs. * p<0.01 vs. AdshScr (n=3/group). Scale bar: 200 m. C, Immunoblots for cleaved caspase 3 and -tubulin (n=3/group).
D, CM viability was evaluated at multiple time points after transduction with Ad-shScr or Ad-shDrp1 using
the CellTiter Blue assay. * p<0.01 vs. Ad-shScr 72 hours after transduction, # p<0.01 vs. Ad-shScr 96 hours
after transduction (n=4/group).
Figure 2. Drp1 plays an essential role in mediating autophagy and maintaining mitochondrial
function in CMs at baseline. A, CMs were transduced with Ad-shScr or Ad-shDrp1 for 96 hours and then
treated with or without chloroquine (10 μM) for 4 hours. Representative Immunoblots for Drp1, LC3 (long
and short exposures), p62 and -tubulin in shScr- or shDrp1-transduced CMs and quantitative analyses are
shown. Chl: chloroquine. * p<0.01 vs. Ad-shScr without chloroquine, # p<0.01 vs. Ad-shScr with
chloroquine, † p<0.01 vs. Ad-shDrp1 without chloroquine (n=3/group). B, CMs were transduced with AdGFP-LC3 for 48 hours and Ad-shScr or Ad-shDrp1 for 96 hours. Some CMs were incubated with
chloroquine (10 μM) for 4 hours. Representative images of fluorescent GFP-LC3 puncta in shScr- or
shDrp1-transduced CMs and quantitative analysis of the GFP-LC3 puncta. Ctr: control. * p<0.01 vs. AdshScr Ctr, # p<0.01 vs. Ad-shScr Chl (n=5/group). In C-J, CMs were transduced with Ad-shScr or AdshDrp1 for 96 hours (n=3/group). C, Relative mitochondrial DNA content in CMs with Drp1 knockdown,
evaluated by PCR for cytochrome b. * p<0.01 vs. Ad-shScr. D, Immunoblots for COX IV and -tubulin in
CMs with Drp1 knockdown. * p<0.05 vs. Ad-shScr. E, Relative mitochondrial ATP production. * p<0.01
vs. Ad-shScr. F, Mitochondrial membrane potential, evaluated with JC-1. Red indicates mitochondria in
which membrane potential is maintained, whereas green indicates depolarized mitochondria. The
quantification of CMs with depolarized mitochondria is shown. * p<0.01 vs. shScr. Yellow scale bar: 500
m; white scale bar: 100 m. G, Mitochondrial membrane potential assessment with TMRE. Red staining
indicates polarized mitochondria. Scale bar: 500 m. In H-I, some CMs were administered CsA (5 M)
after transduction. H, Mitochondrial swelling induced by Ca2+. Each data curve in the left panel represents
the average of 3 individual measurements. Right panel shows the decrease in optical density at 540 nm,
indicating the extent of mPTP opening. * p<0.01 vs. shScr without CsA, # p<0.01 vs. shDrp1 without CsA
(n=3/group). I, CM cell viability. * p<0.01 vs. shScr without CsA, # p<0.01 vs. shScr with CsA, † p<0.05
vs. shDrp1 without CsA (n=4/group). J, Assessment of oxidative stress using MitoSox Red. Scale bar: 200
m.
Figure 3. Drp1 mediates mitochondrial fission, autophagy, and cell survival during GD. A,
Immunoblots for Drp1, COX IV, and -tubulin in CM mitochondrial and cytosolic fractions. * p<0.01 vs.
Ctr (n=3/group). B, Immunohistochemistry for Drp1 and mt-DsRed2. Green: Drp1, Red: mt-DsRed2. Scale
DOI: 10.1161/CIRCRESAHA.116.303356 16
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
bar: 50 m. C, Assessment of mitochondrial morphology using mt-DsRed2. Insets show representative
mitochondria. Gray bar: cells with elongated/total cell number, black bar: cells with foreshortened/total cell
number, white bar: cells with intermediate (mid) mitochondria/total cell number. * p<0.01 vs. foreshortened
in Ad-shScr at baseline, # p<0.01 vs. foreshortened in Ad-shScr after 1 hour GD, † p<0.01 vs. foreshortened
in Ad-shScr after 4 hours GD, ** p<0.05 vs. elongated in Ad-shScr at baseline, ‡ p<0.01 vs. elongated in
Ad-shScr at baseline (n=4/group). Scale bar: 20 m. D, TUNEL staining of CMs with Drp1 knockdown. *
p<0.01 vs. shScr Ctr, # p<0.01 vs. Ad-shDrp1 Ctr, † p<0.01 vs. Ad-shDrp1 after 1 hour GD (n=3/group).
Scale bar: 200 m. E, Representative images of GFP-LC3 puncta. Scale bar: 50 μm. Bar graph indicates
mean number of autophagosomes per cell. * p<0.01 vs. Ad-shScr Ctr. # p<0.01 vs. Ad-shDrp1 Ctr. † p<0.01
vs. Ad-shScr after 4 hours GD (n=5/group). Scale bar: 50 μm. F, Representative images of mRFP-GFPLC3 puncta. Free red puncta indicate autolysosomes, and red and green (yellow) puncta indicate
autophagosomes. Scale bar: 50 μm. Bar graph indicates mean number of autophagosomes and
autolysosomes per cell. * p<0.01 vs. Ad-shScr at baseline, # p<0.01 vs. Ad-shDrp1 at baseline (n=5/group).
G, Representative images of mRFP-GFP-LC3 puncta. Scale bar: 50 m. Bar graph indicates mean number
of autophagosomes and autolysosomes per cell. a p<0.01 vs. Ad-shDrp1 at baseline, b p<0.05 vs. Ad-shScr
at baseline, c p<0.01 vs. Ad-shScr after 4 hours GD, d p<0.01 vs. Ad-shDrp1 at baseline, e p<0.01 vs. AdshDrp1 after 4 hours GD (n=3/group).
Figure 4. Drp1 mediates mitochondrial autophagy during GD. A, CMs were transduced with Ad-LacZ
or Ad-Keima-MLS and cytosolic and mitochondrial fractions were analyzed by immunoblot (n=3/group).
B, CMs were treated with CCCP (25 M) for 16 hours. Representative images of Keima-MLS. Lysosomes
visualized with Alexa 488 Dextran colocalized with puncta with a high ratio of red to green, detected at
560 nm and 440 nm, respectively (n=3/group). Scale bar: 20 μm. C-D, Representative images of KeimaMLS. Puncta with high 560/440 indicate mitochondrial autophagy. The ratio of the area of puncta with high
560/440 vs. the total cell area is shown. C, CMs were transduced with Ad-Keima-MLS and then with AdshScr or Ad-shDrp1. Some then underwent 4 hours of GD. * p<0.01 vs. Ad-shScr Ctr, # p<0.01 vs. AdshScr with GD (n=5/group). Scale bar: 20 μm. D, CMs were transduced with Ad-Keima-MLS and AdshDrp1, followed by Ad-LacZ or Ad-Atg7. Scale bar: 20 μm. E, CMs were transduced with Ad-shScr or
Ad-shDrp1 followed by Ad-LacZ or Ad-Atg7. Cell viability was evaluated with the CellTiter Blue assay.
* p<0.01 vs. Ad-shScr without Ad-Atg7, # p<0.01 vs. Ad-shScr with Ad-Atg7 (n=3/group). F,
Representative EM images of CMs in vitro. Asterisks indicate elongated mitochondria. Open arrows
indicate autophagic vacuoles without mitochondria. Closed arrows indicate autophagic vacuoles containing
mitochondria. Scale bar: 2 m. G, Left panel shows the number of mitochondria per cell. * p<0.01 vs. AdshScr without GD. Right panel shows relative mitochondrial mass per cell. Mitochondrial mass in CMs
transduced with Ad-shScr at baseline is expressed as 1. * p<0.01 vs. Ad-shScr at baseline, # p<0.01 vs. AdshScr with GD. H, Left panel shows the mean number of autophagic vacuoles per cell. * p<0.01 vs. AdshScr without GD, # p<0.01 vs. Ad-shScr with GD, † p<0.01 vs. Ad-shDrp1 without GD. Right panel
shows the number of autophagic vacuoles containing mitochondria per cell. * p<0.01 vs. Ad-shScr with
GD.
Figure 5. Drp1-CKO mice develop cardiac hypertrophy and fibrosis. A, Immunoblot analysis of
cardiac Drp1 in Drp1-CKO and control mice. MHC-MCM: Tg-MHC-MerCreMer. * p<0.01 vs. Ctr
(n=3/group). B, Gross morphology and sagittal sections of control and Drp1-CKO mouse hearts stained
with Hematoxylin-Eosin. Scale bar: 2 mm. C, Assessment of CM size using WGA staining. * p<0.01 vs.Ctr.
Scale bar: 200 m. D, Picric Acid Sirius Red staining to assess cardiac fibrosis. * p<0.01 vs.Ctr. Scale bar:
500 m. In C-D, n=4/group. In A-D, heart samples were harvested 4 weeks after TI.
Figure 6. Mitochondria are fused and dysfunctional in Drp1-CKO mice. A, EM images of Drp1-CKO
and control mouse hearts. The inset shows mitochondrial autophagy seen only in control mouse hearts after
48 hours fasting. Asterisks indicate elongated mitochondria. Mitochondrial mass in control mouse hearts at
DOI: 10.1161/CIRCRESAHA.116.303356 17
baseline is expressed as 1. * p<0.01 vs. Ctr without fasting, # p<0.01 vs. Ctr with fasting. Scale bar: 2 m.
B, Immunoblots for COX IV and -tubulin in Drp1-CKO and control mice. * p<0.01 vs. Ctr. C, Relative
mitochondrial DNA content in Drp1-CKO and control mouse hearts, evaluated by PCR for cytochrome b.
* p<0.01 vs. Ctr. D, Immunoblots for PGC-1, TFAM, and -tubulin in Drp1-CKO and control mice. E,
Relative cardiac ATP production in Drp1-CKO and control mice. * p<0.01 vs. Ctr 10 days after TI, # p<0.01
vs. Ctr 28 days after TI, † p<0.01 vs. Drp1-CKO 10 days after TI. F, Mitochondrial swelling induced by
Ca2+. Each data curve in the left panel represents the average of 3 individual measurements. Right panel
shows the decrease in optical density at 540 nm. * p<0.05 vs. Ctr 10 days after TI, # p<0.01 vs. Ctr 10 days
after TI, † p<0.01 vs. Ctr 28 days after TI, ** p<0.01 vs. Drp1-CKO 10 days after TI. G, ELISA of 4Hydroxynonenal. 4-HNE: 4-Hydroxynonenal. 4-HNE concentration in control mouse hearts is expressed
as 1. * p<0.05 vs Ctr. H, H2O2 production from isolated mitochondria was evaluated with the Amplex Red
Assay. * p<0.01 vs Ctr. In A-H, n=3/group. In A-E and G-H, samples were harvested 4 weeks after TI.
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Figure 7. Apoptosis was increased, whereas autophagy was suppressed, in Drp1-CKO mice. A,
TUNEL staining of hearts in Drp1-CKO and control mice. Arrow indicates TUNEL-positive nucleus. *
p<0.01 vs. Ctr. Scale bar: 50 μm. B, Immunoblots for cleaved caspase 3, cytochrome c, COX IV, and tubulin. Expression normalized with -tubulin or COX IV in Drp1 fl/fl (control) mice is expressed as 1. *
p<0.01 vs.Ctr. C and D, Immunoblots for LC3, p62, and -tubulin. * p<0.01 vs.Ctr. In D, some mice were
treated with chloroquine (10 mg/kg, i.p.) and evaluated 4 hours later. * p<0.01 vs. Ctr without chloroquine,
# p<0.01 vs. Ctr with chloroquine. E, Representative images of mRFP-GFP-LC3 puncta. Fst:fasting. Bar
graph indicates mean number of autophagosomes and autolysosomes per cell. * p<0.01 vs. yellow dots of
Ctr at baseline, # p<0.01 vs. free red dots of Ctr at baseline. Scale bar: 50 μm. In A-E, heart samples were
harvested 4 weeks after TI (n=3/group).
Figure 8. Drp1-hetCKO mice develop cardiac dysfunction and are more susceptible to I/R injury. A,
Immunoblots for Drp1 and -tubulin. * p<0.01 vs. Ctr (n=3/group). B, LVEF at 12 weeks of age. There
was no significant difference in LVEF between control and Drp1-hetCKO mice (n=4/group).C, Relative
ATP production. * p<0.01 vs. Ctr, (n=3/group). D, Immunoblots for Drp1, COX IV, and -tubulin in
mitochondrial and cytosolic fractions. Fst: fasting for 48 hours. I/R: 30 min ischemia and 24 hours
reperfusion. * p<0.01 vs.Ctr at baseline, # p<0.01 vs. Ctr at baseline and Drp1-hetCKO at baseline and after
Fst, † p<0.01 vs. Ctr sham, ** p<0.01 vs. Ctr sham and Drp1-hetCKO sham and after Fst (n=3/group). E,
LVEF, as assessed by echocardiography. * p<0.01 vs. Ctr at baseline, Ctr after Fst and Drp1-hetCKO at
baseline (n=3/group). F, EM images of Drp1-hetCKO and control mouse hearts. The inset shows
mitochondrial autophagy seen only in control mouse hearts after I/R. Asterisks indicate elongated
mitochondria. Mitochondrial mass in control mouse hearts without I/R is expressed as 1. * p<0.01 vs. Ctr
without I/R, # p<0.01 vs. Ctr with I/R (n=3/group). Scale bar: 2 m. G, Immunoblots for LC3, p62, and tubulin. * p<0.01 vs. Ctr without I/R, # p<0.01 vs. Ctr with I/R (n=3/group). H, Representative images of
tetrazolium chloride/Alcian Blue staining of LV sections after I/R. Statistical analyses of % area at risk
(AAR) and MI/AAR are shown. * p<0.05 vs. control (n=3/group).
DOI: 10.1161/CIRCRESAHA.116.303356 18
Novelty and Significance
What Is Known?

Combined genetic downregulation of mitofusin 1 and mitofusin 2, mitochondrial outer membrane
proteins regulating mitochondrial fusion, causes mitochondrial fragmentation and dysfunction and heart
failure in mice.

Drp1 is a GTPase that mediates mitochondrial fission in non-cardiac cells.

Pharmacological suppression of Drp1 with mdivi-1 attenuates myocardial injury in response to
ischemia/reperfusion.
What New Information Does This Article Contribute?
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017

Chronic downregulation of Drp1 induces elongation of mitochondria, mitochondrial dysfunction, heart
failure and premature death in mice.

Downregulation of Drp1 inhibits general autophagy and movement of mitochondrial proteins into
lysosomes in cardiomyocytes.

Chronic downregulation of Drp1 enhances myocardial injury in response to ischemia/reperfusion.
Mitochondria have the ability to remove damaged parts through a process of fission and fusion and
consequent degradation through autophagy. Using a loss-of-function mouse model, we show that chronic
downregulation of Drp1, a GTPase known to induce mitochondrial fission, causes mitochondrial
dysfunction, myocardial cell death, heart failure and the death of the animal. In vitro analyses show that
genetic downregulation of Drp1 directly inhibits general autophagy in cardiomyocytes through Bcl-xLdependent mechanisms. Furthermore, using mito-Keima, a pH-sensitive protein, we show that endogenous
Drp1 is essential for mitochondrial autophagy in response to glucose starvation in cardiomyocytes.
Downregulation of Drp1 in turn causes accumulation of dysfunctional mitochondria and increased cell
death. Furthermore, downregulation of Drp1 exacerbates ischemia/reperfusion injury in the mouse heart in
vivo. These results suggest that endogenous Drp1 plays an important role in mediating mitochondrial
autophagy and maintaining mitochondrial function in response to stress.
DOI: 10.1161/CIRCRESAHA.116.303356 19
Figure 1
shDrp1
TnI
100
Elongated
Mid
Foreshortened
80
#
60
10
*
0
shScr shDrp1
t
shScr
Cells with elongated,
foreshortened
/ total cell number (%)
no
C
mtDsRed2
shScr shDrp1
o
A
ul
di at
st io
rib n
TUNEL positive
ut Re
nuclei (%)
e. se
D ac
es h
tr Pe
oy e
af r R
Relative
tecell viability
e
r u vie
se w.
. D
caspase3
17kDa
D-tubulin
shScr
TUNEL
shDrp1
DAPI
D
15
10
5
0
*
(%)
100
䕔
䕦
rC
irc
䕔
䕦
䕔
*
60
40
20
䕔
䕦
80
䕔
䕦
䕦
#
shScr
shDrp1
0
0
Fo
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
B
50kDa
24
48 72
(hours)
96
Figure 2
Chl
-
sh shScr sh
Drp1
Drp1
-
+
+
Drp1
LC3 I
LC3 II
15kDa
Short LC3 II
p62
62kDa
D-tubulin
50kDa
*
1.6
1.2
0.8
#†
0.4
#
0.8
0.4
0
-
+
C
*
80
60
2
0
E
F
1.0
*
0.5
0
shScr
shDrp1
*
20
0
shScr
shDrp1 + CsA
0.22
0.20
0.18
0
shDrp1
0
4
8 12 16 20
Time (min)
20
H
*
16
12
8
#
4
0
CsA
5PM
*
-
#
+
-
+
shScr shDrp1
Relative cell viability
shScr + CsA
0.24
% Decrease in OD540
Fo
shScr shDrp1
0.26
1.2
#†
1.0
0.8
*
0.6
0.4
0.2
0
CsA
5PM
-
+
-
50kDa
4
*
3
2
1
0
shScr shDrp1
40
G
17kDa
D-tubulin
shDrp1
60
rC
irc
Relative ATP
production
COX IV
Ctr Chl Ctr Chl
shScr
*
D
#
#
0
+
shScr shDrp1
Relative Cox-IV
expression
20
Chl
10PM
4Hr
-
shDrp1
1
40
OD540nm
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Ctr
+
shScr
t
shDrp1
Relative mt DNAo
shDrp1
-
Chl
+
no
-
ul
di at
st io
rib n
ut Re
e. se
D ac LC3 dots / cell
es h
tr Pe
Green fluorescent
oy e
cell number
/ total cell number (%)
af r R
te ev
r u ie
se w.
. D
shScr
*
*#
1.2
0
Chl
shScr
B
*
1.6
content
Long
80kDa
p62 / D-tubulin
expression
shScr
LC3II / D-tubulin
expression
A
+
shScr shDrp1
Figure 3
Ctr GD
4Hr
Drp1
COX IV
D-tubulin
80kDa
17kDa
50kDa
C
Base
mt-DsRed2
80
60
40
20
0
Fo
TUNEL
shScr
Ctr GD
4Hr
GD4Hr
GD1Hr
mt-DsRed2
TnI
TnI
E
GD4Hr
mt-DsRed2
Ctr
GD4Hr
shScr
**
‡
*#
†
*#
‡
‡
#†
†
Base GD1Hr GD4Hr
Base GD1Hr GD4Hr
shScr
shDrp1
GD1Hr GD4Hr
shDrp1
†
#
30
*
*
25
#*
20
15
10
*
5
0
LC3 dots / cell
100
rC
irc
Cells with elongated,
foreshortened
/ total cell number (%)
DAPI
0
Elongated
Mid
Foreshortened
Ctr
shDrp1
0.2
Ctr GD
4Hr
TUNEL positive nuclei (%)
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
shDrp1
TUNEL
0
Base
0.4
ul
di at
st io
rib n
ut Re
e. se
D ac
es h
tr Pe
oy e
af r R
te ev
r u ie
se w.
. D
shScr
D
1
0.6
Drp1 mt-DsRed2 Merge
o
TnI
2
B
t
GD
4Hr
*
0.8
no
Ctr
3
Drp1 / COX IV
Cytosol Mitochondria
Drp1 / D-tubulin
A
30
20
#†
10
0
DAPI
shScr shDrp1
shScr shDrp1
Figure 3
F
shScr
Baseline
shDrp1
GD4Hr
Baseline
*
100
GD4Hr
Free red puncta
Yellow puncta
80
60
*
8
6
t
RFP
40
no
LC3 dots / cell
GFP
2
#
0
Merge
Free red
puncta
G
shScr + Atg7
Baseline
GD
shDrp1 + Atg7
Baseline
Base GD4Hr
Base GD4Hr
shScr
shDrp1
de
120
Free red puncta
de
GD
Yellow puncta
100
GFP
80
60
40
RFP
a
b
a
c
Merge
rC
irc
20
Fo
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
ul
di at
st io
rib n
ut Re
e. se
D ac
es h
tr Pe
oy e
af r R
te ev
LC3 dots / cell
r u ie
se w.
. D
Yellow puncta
o
4
0
Base GD4Hr Base GD4Hr
shScr
shDrp1
Atg7
A
cytosol mitochondria
B
Ratio
Alexa 488
560 nm 560/440 nm Dextran Merge
440 nm
E
CCCP
25 PM
Keima
COX IV
D-tubulin
28kDa
17kDa
50kDa
Relative cell viability
Figure 4
1.0
*#
0.8
0.6
*#
0.4
0.2
0
Atg7 -
-
+
+
shScr shDrp1
C
*
8
6
440 nm
560 nm
4
2
560 nm
# #
0
GD - +
High (560/440) signal area
/ cell area (%)
shDrp1 shDrp1
+ LacZ + Atg7
no
GD4H䡎
ul
di at
st io
rib n
ut Re
e. se
D ac
es h
tr Pe
oy e
af r R
te ev
r u ie
se w.
. D
440 nm
0.8
0.4
0
shDrp1
- +
+
+
Ratio
Ratio
F
Baseline
rC
irc
shScr
*
shDrp1
*
GD 4Hr
*
*
*
100
*
80
*
*
60
40
20
0
GD
Relative
mitochondrial mass
2
120
*
1
0
-
+
shScr
-
+
shDrp1
GD
-
+
-
+
shScr shDrp1
Autophagic vacuole / cell
H
* #
*
14
12
10
8
6
4
#
2
†
*#
0
GD
-
+
-
+
shScr shDrp1
Autophagic vacuole
containing mitochondria / cell
Fo
*
G
Number of
mitochondria / cell
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
High (560/440) signal area
/ cell area (%)
shDrp1
Ctr
GD4H䡎
o
shScr
Ctr
t
D
4
3
2
1
*
0
GD
-
*
+
-
*
+
shScr shDrp1
Figure 5
B
50kDa
Tamoxifen
-
+
-
+
Drp1 fl/fl
+
+
+
+
D-MHC MCM
-
-
+
+
0.8
0.4
0
Tamoxifen
Drp1 fl/fl
Ctr
Drp1-CKO
D-MHC MCM
*
+
-
+
+
-
+
+
+
+
+
t
80kDa
Ctr
1.2
no
Drp1
α-tubulin
Relative Drp1
expression
A
C
(Pm2)
-
Tamoxifen
*
300
+
200
Drp1 fl/fl
㽢
D-MHC MCM
Drp1 fl/fl
100
0
Tamoxifen
-
+
-
+
Drp1 fl/fl
+
+
+
+
D-MHC MCM
-
-
+
+
Ctr
D
-
rC
irc
Drp1 fl/fl
㽢
D-MHC MCM
Drp1 fl/fl
Fo
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
ul
di at
st io
rib n
ut Re
e. se
D ac
es h
tr Pe
oy e
area
rR
% Fibrosis
aCross-sectional
fte e
r u vie
se w.
. D
o
Ctr
Tamoxifen
+
*
12
8
4
0
Tamoxifen
-
+
-
+
Drp1 fl/fl
+
+
+
+
D-MHC MCM
-
-
+
+
Ctr
Figure 6
*
48Hr
fasting
3
0
Tamoxifen
2
*
1
-
Fasting
+
PGC-1D
TFAM
α-tubulin
+
+
+
+
+
+
Drp1 fl/fl
D-MHC MCM
-
-
+
+
D-MHC MCM
COX IV
17kDa
D-tubulin
+
-
*
E
27kDa
+
+
-
Drp1-CKO
3
+
+
1.2
1.0
* #
0.8
2
0.6
†
1
0.4
* #
+
Drp1 fl/fl
+
+
D-MHC MCM
-
-
-
Ctr
+
+
+
+
+
0
Tamoxifen
Drp1 fl/fl
D-MHC MCM
rC
irc
**
Ctr (28D)
Fo
0.50
Drp1-CKO (10D)
Drp1-CKO (28D)
0.45
10
8
*†
6
4
2
0
0
0
4
8
+
+
+
+
+
0.2
0
12
16
20
1.8
1.6
1.4
1.2
1.0
0.8
0.6
0.4
0.2
0
Tamoxifen
Drp1 fl/fl
Time (min)
D-MHC MCM
Ctr
Drp1
CKO
Ctr
G
#†
Ctr (10D)
0.55
+
+
-
Ctr
F
0.60
+
-
+
-
+
+
Ctr
+
+
Drp1-CKO
H
*
(Pmol/Pg)
2.5
H2O2 concentration
-
+
+
+
Ctr
50kDa
Tamoxifen
OD540nm
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Drp1 fl/fl
4
90kDa
50kDa
Tamoxifen
ul
di at
st io
rib n
% Decrease in OD540
ut Re
e. se Relative COX IV
D acexpression
es h
tr Pe
oy e
Relative 4-HNE content
af r R
te ev
r u ie
se w.
. D
Relative ATP production
+
Ctr
B
D-MHC MCM
+
+
+
D
+
+
Tamoxifen
*
+
+
-
Drp1 fl/fl
0
*
2
no
Base
#
4
*
4
t
Relative
mitochondrial mass
*
C
#
*
Drp1-CKO
o
Ctr
Relative mtDNA
content
A
+
+
+
*
2.0
1.5
1.0
0.5
0
Tamoxifen
+
+
+
+
D-MHC MCM +
Ctr Drp1
CKO
Drp1 fl/fl
B
mitochondria
TUNEL
DAPI
17kDa
50kDa
Tamoxifen -
+
-
+
Drp1 fl/fl +
+
+
+
D-MHC MCM -
-
+
+
Cytochrome c
COX IV
D-tubulin
Tamoxifen Drp1 fl/fl +
D-MHC MCM -
0.2
0.1
+
+
*
12
8
4
0
Tamoxifen Drp1 fl/fl +
D-MHC MCM -
+
+
+
+
+
-
+
+
+
+
+
p62
D-tubulin
D-MHC MCM
+
+
-
+
+
15kDa
1.2
62kDa
0.8
+
+
+
+
-
+
+
+
+
+
Drp1 fl/fl D-MHC MCM -
Ctr
+
+
-
Chl
-
LC3 I
-
Ctr
rC
irc
p62
E
+
Drp1
CKO
+
+
+
+
+
-
Ctr
Baseline Fasting
62kDa
50kDa
*#
0
Chl -
*
3
2
1
+
Yellow puncta
10
*
0
Base Fst Base Fst
Free red
puncta
1
0
Chl -
+
Free red puncta
5
Merge
*
Yellow puncta
15
Ctr
Drp1-CKO
+
+
+
*
2
Ctr
20
+
+
-
*
#
25
+
+
Ctr
Drp1-CKO
Baseline Fasting
LC3 dots / area
RFP
-
+
+
+
4
Ctr Drp1-CKO
30
GFP
+
+
+
+
Ctr
Drp1 fl/fl D-MHC MCM -
#
1
+
+
0
Tamoxifen +
2
+
15kDa
LC3 II
D-tubulin
Drp1
CKO
+
+
*
0
Tamoxifen Drp1 fl/fl +
D-MHC MCM -
expression
Ctr
+
+
Ctr
Ctr
D
+
-
1
*
0
Tamoxifen +
+
+
+
2
0.4
Drp1 fl/fl
50kDa
Ctr
50kDa
Fo
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
LC-3 I
LC-3 II
+
-
17kDa
Ctr
Ctr
Tamoxifen
14kDa
ul
di at
st io
rib n
ut Re
e. se
D ac LC3II / D-tubulin
LC3II / D-tubulin
es h expression
expression
tr Pe
oy e
af r R
te ev
r u ie
se w.
D
p62 / D-tubulin .
p62 / D-tubulin
+
+
-
Cytochrome c / D-tubulin
expression
% TUNEL positive
nuclei
*
0
Tamoxifen Drp1 fl/fl +
D-MHC MCM -
C
Caspase3 / D-tubulin
expression
Ctr
cytosol
t
Caspase3
D-tubulin
no
Drp1-CKO
o
Ctr
expression
Figure 7
A
-
+
Drp1-CKO
80
80kDa
50kDa
1.0
60
40
20
*
0.5
0
Ctr
0
Ctr Drp1
hetCKO
Drp1
hetCKO
0.8
*
60
40
20
0.4
0
0
Ctr
(12W) (12W)
48Hr
Fst
Drp1
hetCKO
Ctr
(12W) (12W)
5
G
* *
p62
Drp1
80kDa
17kDa
48Hr
Fst
Drp1
hetCKO
#
1.0
0.8
0.6
0.4
0.2
0
3
2
1
0
D-tubulin
† †
*
* #
2
*
1
#
*#
*#
1
*
62kDa
0
I/R
(12W)
Infarct / AAR (%)
(12W)
I/R
Drp1
hetCKO
40
AAR (%)
Drp1
hetCKO
0
- + - +
Ctr
Ctr
Drp1
hetCKO
Sham
*
**
10
8
6
4
2
0
2
50kDa
Ctr
50kDa
15kDa
Fo
LC3 I
LC3 II
I/R
F
Drp1-hetCKO
COX IV
D tubulin
4
rC
irc
Sham
Drp1 / COX IV
Drp1 / D-tubulin
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Drp1
COX IV
D tubulin
1.0
0.8
0.6
0.4
0.2
0
Ctr
Drp1-hetCKO
30
20
10
0
Ctr
Drp1
hetCKO
- +
- +
Ctr
Drp1
hetCKO
*
60
40
20
0
Ctr
Drp1
hetCKO
I/R
Relative
mitochondrial mass
Ctr
ul
di at
LC3II / D-tubulin
i
expression s
tr on
ib R
ut e Drp1 / D-tubulin
e. se
D ac
es h
tr Pe
p62 / D-tubulin
o COX IVe
expression
Drp1 /y
af r R
te ev
r u ie
se w.
. D
D
H
*
o
Relative Drp1
Expression
Drp1
α-tubulin
80
1.2
t
(12W)
E
C
no
(12W)
B
LVEF (%)
Drp1
hetCKO
Relative ATP production
Ctr
LVEF(%)
Figure 8
A
#
*
2
#
*
1
0
I/R
*
-
Ctr
+
-
+
Drp1
hetCKO
Downloaded from http://circres.ahajournals.org/ by guest on May 3, 2017
Endogenous Drp1 Mediates Mitochondrial Autophagy and Protects the Heart Against Energy
Stress
Yoshiyuki Ikeda, Akihiro Shirakabe, Yasuhiro Maejima, Peiyong Zhai, Sebastiano Sciarretta, Jessica
Toli, Masatoshi Nomura, Katsuyoshi Mihara, Kensuke Egashira, Mitsuru Ohishi, Maha Abdellatif
and Junichi Sadoshima
Circ Res. published online October 20, 2014;
Circulation Research is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231
Copyright © 2014 American Heart Association, Inc. All rights reserved.
Print ISSN: 0009-7330. Online ISSN: 1524-4571
The online version of this article, along with updated information and services, is located on the
World Wide Web at:
http://circres.ahajournals.org/content/early/2014/10/20/CIRCRESAHA.116.303356
Data Supplement (unedited) at:
http://circres.ahajournals.org/content/suppl/2014/10/20/CIRCRESAHA.116.303356.DC1
Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in
Circulation Research can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial
Office. Once the online version of the published article for which permission is being requested is located, click
Request Permissions in the middle column of the Web page under Services. Further information about this process is
available in the Permissions and Rights Question and Answer document.
Reprints: Information about reprints can be found online at:
http://www.lww.com/reprints
Subscriptions: Information about subscribing to Circulation Research is online at:
http://circres.ahajournals.org//subscriptions/
Supplemental Materials and Methods
Primary culture of neonatal rat ventricular myocytes
Primary cultures of ventricular cardiomyocytes (CMs) were prepared from 1-day-old
Crl:(WI) BR-Wistar rats (Harlan). A CM-rich fraction was obtained by centrifugation
through a discontinuous Percoll gradient as described.1
Glucose deprivation
To obtain starvation conditions, CMs were washed three times with phosphate-buffered
saline (PBS) and incubated in glucose-free serum-free DMEM (11966-025, Invitrogen)
at 37 °C as described.1
Chloroquine treatment
To inhibit autophagic flux in vivo, chloroquine was injected (10 mg/kg) intraperitoneally
as previously described.2 Four hours later, animals were euthanized for immunoblot
detection of autophagy markers. To inhibit autophagic flux in vitro, cultured CMs were
treated with 10 mM chloroquine for 4 hours.
Adenovirus construction and transduction
Adenoviruses harboring GFP-LC3 (Ad-GFP-LC3),2 tandem fluorescent mRFP-GFP-LC3
(Ad-tf-LC3),2 mt-DsRed2,3 wild-type Atg7,3 LacZ (Ad-LacZ),3 hemagglutinin (HA)-tagged
Drp1,4 Beclin1 short hairpin (sh) RNA (Ad-shBeclin1),1 flag-tagged Beclin1,5 Bcl-xL
shRNA (Ad-shBcl-xL),5 and Scramble shRNA (Ad-shScr)1 have been described.
Adenoviruses harboring shRNA for Drp1 (Ad-shDrp1) and for Atg7 (Ad-shAtg7) were
generated using the Admax system (Microbix) as previously described1 using the
following hairpin-forming oligos: 5’CGGCAATTGAGCTAGCATATTTCAAGAGAATATGCTAGCTCAATGCCTTTTTA-3’ for
1 Ad-shDrp1 and 5’CGCGTCACAGCCCTGCCATATTCAAGAGATATGGCAGGGCTGTGACGCTTTTTA-3’
for Ad-shAtg7. Transductions with Ad-Drp1, Ad-Keima-MLS, Ad-mt-DsRed2, Ad-Atg7,
Ad-tf-LC3, Ad-GFP-LC3, and Ad-LacZ were carried out for 48 hours. Knockdown
adenoviruses, including Ad-shScr, Ad-shDrp1, Ad-shBeclin1, and Ad-shAtg7, were
transduced for 96 hours. Adenoviruses were transduced at 15 MOI.
Evaluation of fluorescent LC3 puncta
The method of evaluating tandem fluorescent LC3 puncta using Ad-tf-LC3 has been
described previously.2 CMs cultured on cover slips were transduced with Ad-GFP-LC3
or Ad-tf-LC3 at 15 MOI. Forty-eight hours after adenovirus transduction, the cells were
washed with PBS, fixed with 4% paraformaldehyde (PFA), mounted with a reagent
containing 4’,6-diamidino-2-phenylindole (DAPI) (Vectashield, Vector Laboratories), and
viewed under a fluorescence microscope (Nikon Eclipse E800). The number of GFP
and mRFP dots was determined by manual counting of fluorescent puncta from at least
4 different myocyte preparations with a 60X objective. At least 50 cells were scored in
each experiment. The nuclear number was evaluated by counting the number of DAPIstained nuclei in the same field. The number of dots/cell was obtained by dividing the
total number of dots by the number of nuclei in each microscopic field. For in vivo
determination of the number of fluorescent LC3 dots, fresh heart slices were embedded
in Tissue-Tek OCT compound (Sakura Finetechnical Co.) and frozen at -80°C. Sections
10 μm thick were obtained from the frozen tissue samples using a cryostat (CM3050 S;
Leica), air-dried for 30 min, fixed by washing in 95% ethanol for 10 min, mounted using
a reagent containing DAPI, and viewed under a fluorescence microscope.
2 Histological analysis
Histological analysis was performed as described.1 In brief, heart specimens were fixed
with formalin, embedded in paraffin, and sectioned at 6 m thickness. Interstitial fibrosis
was evaluated by Masson’s Trichrome and Picric Acid Sirius Red (PASR) staining. The
myocyte cross-sectional area was measured from images captured from wheat germ
agglutinin (WGA)-stained sections. The outlines of 100–200 myocytes were traced in
each section using NIH ImageJ.
Immunohistochemistry
The method of immunostaining has been described.3 CMs were stained with anti-Drp1
mouse monoclonal antibody (BD Transduction, 611112), anti-Troponin I rabbit
polyclonal antibody (Santa Cruz, 15368), Alexa Fluor 488-conjugated goat anti-mouse
IgG (Invitrogen), Alexa Fluor 488-conjugated goat anti-rabbit IgG (Invitrogen), and
Vectashield mounting medium with DAPI (Vector Laboratories). Analyses were
performed using fluorescence microscopy (Zeiss).
Electron microscopy
Conventional electron microscopy was performed as described previously.1 In brief,
CMs were fixed in Karnofsky’s fixative and then postfixed in 1% osmium tetraoxide,
dehydrated in a graded series of acetone concentrations, and embedded in Sparr resin.
Sections of 98 nm thickness were placed on copper grids that were double-stained with
uranyl acetate and lead citrate. Discs were examined with a JEOL 1200 electron
microscope. Mitochondrial mass was analyzed using Image J. The average
mitochondrial mass was calculated from 50 mitochondria per slide on three different
slides.
3 Evaluation of mitochondrial morphology
Mitochondrial morphology was examined according to the modified methods described
previously.6 At least 100 CMs transduced with mitochondria-targeted DsRed2 and
immunostained with Troponin I were examined using confocal microscopy. Mitochondria
whose length is shorter than one sarcomere (the distance between consecutive Zbands) are defined as foreshortened, those whose length is longer than 2 sarcomeres
are defined as elongated, and those whose length is longer than one sarcomere and
shorter than 2 sarcomeres are defined as intermediate (mid). Cells displaying either
predominantly (>50%) elongated or (>50%) foreshortened mitochondria were classified
as cells with elongated or foreshortened mitochondria, respectively. Cells containing
<50% elongated and <50% foreshortened mitochondria were classified as intermediate
(mid).
Quantitative real-time PCR for mitochondrial DNA
Total DNA was extracted from mouse hearts using the Quick-gDNA MiniPrep kit (ZYMO
RESEARCH) according to the manufacturer’s protocol. The mtDNA content was
quantified by real-time PCR of cardiac DNA as described.7 Primer sequences used for
cytochrome b and β-actin are as follows: 5’-CCACTTCATCTTACCATTTATTATCGC-3’
(forward primer) and 5’-TTTTATCTGCATCTGAGTTTAA-3’ (reverse primer) for
cytochrome b, and 5’-CTGCCTGACGGCCAGG-3’ (forward primer) and 5’CTATGGCCTCAGGAGTTTTGTC-3’ (reverse primer) for genomic β-actin.
Subcellular fractionation
Mitochondrial and cytosolic fractions were purified through a previously described
procedure.7 Briefly, isolated mouse hearts were homogenized in 10 volumes of ice-cold
4 Buffer A [200 mM mannitol, 50 mM sucrose, 10 mM KCl, 1 mM EDTA, 10 mM HepesKOH (pH 7.4), 0.1% BSA, and a mixture of protease inhibitors]. Homogenates were
centrifuged at 600 × g for 5 min at 4 °C. Supernatants were then centrifuged at 3,500 ×
g for 15 min at 4 °C. The pellets were resuspended in Buffer A and centrifuged at 1,500
× g for 5 min. The supernatants were centrifuged at 5,500 × g for 10 min at 4 °C, and
then the pellets were suspended as the mitochondrial fraction in PBS containing
protease inhibitors. The supernatant was further centrifuged at 100,000 × g for 60 min,
and the resultant pellet and supernatant were used as microsomal and cytosolic
fractions, respectively.
ATP production assay
The mitochondrial fraction of mouse hearts was prepared as described above. ATP
production was measured with an ATP Bioluminescent Assay kit (Sigma). 25mg of
mitochondria is incubated with ATP assay mix and MSH buffer containing 625M ADP
and substrate (10mM pyruvate and 10mM malate).
Mitochondrial complex activity assay
The mitochondrial fraction was prepared from mouse hearts as described above.
Electron transport chain complex activities were measured, using MitoCheck Complex I,
II-III and IV Activity Assay Kit (Cayman Chemical Company, USA) according to the
method described previously.8 Briefly, complex I was assayed by monitoring the
rotenone-sensitive ubiquinone-1 (Q1)-stimulated NADH oxidation, complex II+III by
measuring the rate of reduced cytochrome c formation using succinate as substrate,
and complex IV by measuring ferrocytochrome c oxidation with or without KCN.
Mitochondrial complex activities were normalized by the weight of mitochondria.
5 Mitochondrial swelling assay
The mitochondrial swelling assay was performed as described.7 In brief, 50 g of
isolated mitochondria from mice or 30 g of isolated mitochondria from CMs were
suspended in a swelling buffer [250 mM sucrose, 10 mM MOPS, 5  EGTA, 2 mM
MgCl2, 5 mM KH2PO4, 5 mM pyruvate, and 5 mM malate] and incubated with 150 M
calcium chloride (CaCl2) in a final volume of 200 L in a 96-well plate for 20 min.
Absorbance was read at 540 nm.
Evaluation of mitochondrial membrane potential
In order to evaluate mitochondrial membrane potential/integrity, cultured CMs were
stained with tetramethylrhodamine ethyl ester (TMRE) and 5,5’,6,6’- tetrachloro1,1’,3,3’-tetraethylbenzimidazolocarbocyanine iodide (JC-1) using MitoPT® TMRE and
MitoPT® JC-1 (ImmunoChemistry Technologies), respectively, according to the
manufacturer’s instructions.
Mitochondrial flux analyses using the Seahorse system
To measure the rate of oxidative phosphorylation in intact CMs, a Seahorse XF24
Extracellular Flux Analyzer (Seahorse Bioscience, Billerica, MA, USA) was used
according to the methods described previously.9 CMs were plated at a density of
120,000 cells/well in 24-well Seahorse assay plates. CMs were transduced with AdshScr or Ad-shDrp1 for 96 hours prior to measurement. One hour prior to the beginning
of measurements, the medium was replaced with XF medium supplemented with 17.5
mM glucose and 1 mM pyruvate and incubated for 1 hour in a 37°C incubator without
CO2. Oxygen consumption rate (OCR) was measured three times at baseline, followed
by injection with oligomycin (1 μM) to measure the ATP-linked OCR. Carbonylcyanide6 p-triflouromethoxyphenylhydrazone (FCCP, 3 μM), an uncoupler, was used to
determine maximal respiration, and rotenone (1 μM) and antimycin A (1 μM) were
injected to determine the non-mitochondrial respiration. Three independent experiments
were performed. OCR was normalized by the amount of mtDNA or the rate of CM cell
viability in each well.
H2O2 measurement
H2O2 production was measured with an Amplex Red H2O2 assay kit (Molecular Probes;
Invitrogen) as described.10
Assays for measurement of oxidative stress
Cardiac tissue homogenates were assessed for 4-Hydroxynonenal (4-HNE) content (Cell
Biolabs, Inc, San Diego, CA, USA) as described.11
Cell viability
Cell viability was measured by CellTiter-Blue (CTB) assays (Promega) as described.1 In
brief, CMs (1 X 105 per 100 μl) were seeded onto 96-well dishes. After 24 hours, the
cells were incubated with complete medium or glucose-free medium in the presence or
absence of chelerythrine chloride (10 mM) or adenovirus vectors. Viable cell numbers
were measured on the indicated days by the CTB assay. The CTB assays were
performed according to the supplier's protocol. The experiments were conducted in
triplicate at least three times.
Evaluation of apoptosis
DNA fragmentation was detected in situ with the use of terminal deoxynucleotidyl
transferase dUTP nick end labeling (TUNEL), as described.1 Nuclear density was
determined by manual counting of DAPI-stained nuclei in 6 fields for each animal with
7 the 40x objective, and the number of TUNEL-positive nuclei was counted by examining
the entire section with the same power objective.
Immunoblot analysis
The methods used for preparation of cell lysates from in vitro and in vivo samples and
for immunoblot analyses have been described previously.2 For in vitro samples, protein
lysates were prepared from myocytes cultured in 6 cm culture dishes using boiled
(95 °C for 2 min) 2X SDS sample buffer containing 4% SDS, 20% glycerol, 120 mM
Tris-HCl (pH 6.8), 0.01% bromophenol blue, and 5% beta-mercaptoethanol. The protein
samples were immediately boiled again at 95 °C for 3 min. Heart tissue homogenates
were prepared using RIPA buffer containing 50 mM Tris-HCl (pH 8.0), 150 mM NaCl,
0.1% SDS, 1% Igepal CA-630, and 0.5% sodium deoxycholate with protease inhibitors
(Sigma, P8340) at a 1:400 dilution. The antibodies used include Drp1 (BD Transduction,
611112), Mfn1 (abcam, ab57602), Mfn2 (Sigma, M6319), OPA1 (BD Transduction,
612608), Fis1 (Santa Cruz, sc98900), LC3 (BML, M186-3), p62 (ORIGENE, TA307334),
PGC-1 (Santa Cruz, sc-13067), TFAM (Sigma, SAB1401383), COX IV (Cell Signaling,
4844S), cytochrome c (Cell Signaling, 4272S), cleaved caspase 3 (Cell Signaling,
9664S), Keima (BML, M182-3), and -tubulin (Sigma, T6199). Densitometric analyses
were performed using Scion Image software (Scion).
Immunoprecipitation
Immunoprecipitation was performed according to the methods described previously.5 In
brief, CMs were lysed with IGEPAL CA-630 buffer (50 mM Tris-HCl (pH 7.4), 1%
IGEPAL CA-630, 10 mM EDTA, 150 mM NaCl, 50 mM NaF, 1 μM leupeptin and 0.1 μM
aprotinin). Primary antibody was covalently immobilized on protein A/G agarose using
8 the Pierce Crosslink Immunoprecipitation Kit according to the manufacturer's
instructions (Thermo Scientific). Samples were incubated with immobilized antibody
beads for at least 2 h at 4 °C. After immunoprecipitation, the samples were washed with
TBS five times. They were then eluted with glycine-HCl (0.1 M, pH 3.5) and the
immunoprecipitates were subjected to immunoblotting using specific primary antibodies
and a conformation-specific secondary antibody that recognizes only native IgG (Cell
Signaling).
Hemodynamic analysis
Echocardiography and measurement of LV +dP/dt were performed as described,12
using ultrasonography (Acuson Sequoia C256; Siemens Medical Solutions) and a highfidelity microtip pressure transducer catheter (1.4 Fr, Model SPR-839; Millar
Instruments, Houston, TX), respectively.
I/R surgery and assessment of area at risk and infarct size
Myocardial I/R was achieved by temporarily occluding the left anterior descending
coronary artery (LAD) and then releasing the occlusion as described.1 The duration of
ischemia was 30 min, and that of reperfusion was 24 hours. To demarcate the ischemic
area at risk (AAR), Alcian blue dye (1%) was perfused into the aorta and coronary
arteries. Hearts were excised, and LVs were sliced into 1 mm thick cross sections. The
heart sections were then incubated with a 1% triphenyltetrazolium chloride solution at
37 °C for 10 min. The infarct area (pale), the AAR (not blue), and the total LV area from
both sides of each section were measured using Adobe Photoshop (Adobe Systems
Inc.), and the values obtained were averaged. The percentage of area of infarction and
AAR of each section were multiplied by the weight of the section and then totaled from
9 all sections. AAR/LV and infarct area/AAR were expressed as a percentage as
previously reported.1
Mdivi1 treatment
CMs or mice were administered with mdivi1 (Sigma) as described.6
10 References
1. Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, Levine B, Sadoshima J.
Distinct roles of autophagy in the heart during ischemia and reperfusion: roles of
AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ Res.
2007;100:914-922.
2. Hariharan N, Maejima Y, Nakae J, Paik J, Depinho RA, Sadoshima J. Deacetylation
of FoxO by Sirt1 Plays an Essential Role in Mediating Starvation-Induced Autophagy
in Cardiac Myocytes. Circ Res. 2010;107:1470-1482.
3. Sciarretta S, Zhai P, Shao D, Zablocki D, Nagarajan N, Terada LS, Volpe M,
Sadoshima J. Activation of NADPH Oxidase 4 in the Endoplasmic Reticulum
Promotes Cardiomyocyte Autophagy and Survival During Energy Stress Through the
Protein Kinase RNA-Activated-Like Endoplasmic Reticulum Kinase/Eukaryotic
Initiation Factor 2α/Activating Transcription Factor 4 Pathway. Circ Res.
2013;113:1253-1264.
4. Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda K, Otera H, Nakanishi
Y, Nonaka I, Goto Y, Taguchi N, Morinaga H, Maeda M, Takayanagi R, Yokota S,
Mihara K. Mitochondrial fission factor Drp1 is essential for embryonic development
and synapse formation in mice. Nat Cell Biol. 2009;11:958-966.
5. Maejima Y, Kyoi S, Zhai P, Liu T, Li H, Ivessa A, Sciarretta S, Del Re DP, Zablocki
DK, Hsu CP, Lim DS, Isobe M, Sadoshima J. Mst1 inhibits autophagy by promoting
the interaction between Beclin1 and Bcl-2. Nat Med. 2013;19:1478-88. Ong SB,
Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting
mitochondrial fission protects the heart against ischemia/reperfusion injury.
11 Circulation. 2010;121:2012-2022.
6. Kuroda J, Ago T, Matsushima S, Zhai P, Schneider MD, Sadoshima J. NADPH
oxidase 4 (Nox4) is a major source of oxidative stress in the failing heart. Proc Natl
Acad Sci U S A. 2010;107:15565-15570.
7. Montaigne D, Marechal X, Coisne A, Debry N, Modine T, Fayad G, Potelle C, El Arid
JM, Mouton S, Sebti Y, Duez H, Preau S, Remy-Jouet I, Zerimech F, Koussa M,
Richard V, Neviere R, Edme JL, Lefebvre P, Staels B. Myocardial contractile
dysfunction is associated with impaired mitochondrial function and dynamics in type
2 diabetic but not in obese patients. Circulation. 2014;130:554-564.
8. Yu H, Tigchelaar W, Koonen DP, Patel HH, de Boer RA, van Gilst WH, Westenbrink
BD, Silljé HH. AKIP1 expression modulates mitochondrial function in rat neonatal
cardiomyocytes. PLoS One. 2013;8:e80815.
9. Matsushima S1, Kuroda J, Ago T, Zhai P, Ikeda Y, Oka S, Fong GH, Tian R,
Sadoshima J. Broad suppression of NADPH oxidase activity exacerbates
ischemia/reperfusion injury through inadvertent downregulation of hypoxia-inducible
factor-1α and upregulation of peroxisome proliferator-activated receptor-α. Circ Res.
2013 12;112:1135-49.
10. Ikeda Y, Fujita S, Miyata M, Shinsato T, Kubozono T, Kuwahata S, Hamada N,
Miyauchi T, Yamaguchi T, Torii H, Hamasaki S, Tei C. Effect of Waon therapy on
oxidative stress in chronic heart failure. Circ J. 2011;75:348-356.
11. Maejima Y, Galeotti J, Molkentin JD, Sadoshima J, Zhai P. Constitutively active
MEK1 rescues cardiac dysfunction caused by overexpressed GSK-3α during aging
and hemodynamic pressure overload. Am J Physiol Heart Circ Physiol.
12 2012;303:H979-88.
13 Online table I. Measurement of organ weight parameters in Drp1-CKO mice 4 weeks
after tamoxifen injection
Drp1 flox/flox
MHC-MerCreMer
Tamoxifen ip
n
BW (g)
Tibia length (mm)
LV weight (mg)
LV weight/Tibia length (mg/mm)
RV weight (mg)
RV/Tibia length (mg/mm)
Lung weight (mg)
Lung weight/Tibia length (mg/mm)
Liver weight (mg)
Liver weight/Tibia length (mg/mm)
+
4
26.0±3.4
19.6±0.3
84±4
4.3±0.2
15±3
0.8±0.2
166±4
8.5±0.3
983±111
50.3±5.8
+
+
4
28.2±3.9
19.4±0.1
86±3
4.4±0.2
14±3
0.7±0.1
162±12
8.4±0.7
945±73
48.8±3.7
+
+
4
24.1±0.5
19.6±0.4
83±2
4.2±0.2
12±3
0.6±0.1
163±16
8.3±0.8
935±123
47.9±6.7
+
+
+
8
26.4±4.1
19.5±0.6
105±13 *
5.4±0.8 *
21±4 *
1.1±0.2 *
191±16 *
9.8±0.8 *
1089±294
56.3±16.5
BW: Body weight, LV: Left ventricle, RV: Right ventricle. * p<0.01 vs. Drp1 flox/flox
without Tamoxifen ip, Drp1 flox/flox with Tamoxifen ip or Drp1 flox/flox X MHCMerCreMer without Tamoxifen ip.
Online table II. Measurement of organ weight parameters in Drp1-CKO mice 8 weeks
after tamoxifen injection
Drp1 flox/flox
MHC-MerCreMer
Tamoxifen ip
n
BW (g)
Tibia length (mm)
LV weight (mg)
LV weight/Tibia length (mg/mm)
RV weight (mg)
RV/Tibia length (mg/mm)
Lung weight (mg)
Lung weight/Tibia length (mg/mm)
Liver weight (mg)
Liver weight/Tibia length (mg/mm)
Ctr
+
+
4
26.3±2.6
20.2±0.9
87.5±7.6
4.34±0.44
13.0±2.4
0.64±0.09
158.0±13.9
7.9±1.0
1079±141
53.6±8.7
Drp1-CKO
+
+
+
4
27.5±2.3
20.4±0.4
112.3±5.3 #
5.51±0.32 #
25.0±5.1 #
1.23±0.26 #
199.0±21.0 *
9.8±1.1 *
1340±91 *
65.7±4.1 *
BW: Body weight, LV: Left ventricle, RV: Right ventricle. * p<0.05 vs. Ctr, # p<0.01 vs.
Ctr.
Online table III. Measurement of hemodynamic parameters in Drp1-CKO mice 4 weeks
after tamoxifen injection
Drp1 flox/flox
MHC-MerCreMer
Tamoxifen ip
n
HR (beat/min)
LVEDP (mmHg)
LVSP (mmHg)
dP/dt Max (mmHg/sec)
dP/dt Min (mmHg/sec)
LVDd (X10-2mm)
LVDs (X10-2mm)
LVEF (%)
IVS (X10-2mm)
LVPW (X10-2mm)
+
4
495±34
2.5±1.0
88.0±5.7
7250±100
7200±650
308±28
169±26
83.7±3.3
80.5±1.3
79.8±1.7
+
+
4
495±34
2.5±1.0
89.5±4.4
7500±890
7500±380
328±39
179±22
82.0±1.9
79.5±1.7
80.5±2.1
+
+
4
488±30
3.5±1.9
88.5±4.1
7400±520
7500±500
317±21
176±11
82.8±1.9
79.8±1.7
80.0±0.8
+
+
+
8
484±61
12.0±2.1 *
75.5±8.7 *
5200±520 *
5000±1100 *
393±44 *
307±38 *
52.4±5.9 *
84.0±2.6 *
84.5±3.3 *
HR: Heart rate, LVEDP: Left ventricular end diastolic pressure, LVSP: Left ventricular
systolic pressure, LVDd: Left ventricular diastolic dimension, LVDs: Left ventricular
systolic dimension, LVEF: Left ventricular ejection fraction, IVS: Interventricular septum,
LVPW: Left ventricular posterior wall. * p<0.01 vs. Drp1 flox/flox without Tamoxifen ip,
Drp1 flox/flox with Tamoxifen ip or Drp1 flox/flox X MHC-MerCreMer without
Tamoxifen ip.
Online table IV. Measurement of hemodynamic parameters 8 weeks after tamoxifen
injection
Drp1 flox/flox
MHC-MerCreMer
Tamoxifen ip
n
HR (beats/min)
LVDd (X10-2mm)
LVDs (X10-2mm)
LVEF (%)
Ctr
+
+
4
498±79
319±15
175±15
83.2±4.4
Drp1-CKO
+
+
+
4
493±54
409±11 *
339±24 *
41.9±12.9 *
HR: Heart rate, LVDd: Left ventricular diastolic dimension, LVDs: Left ventricular systolic
dimension, LVEF: Left ventricular ejection fraction. * p<0.01 vs. Ctr.
Online table V. Measurement of hemodynamic parameters 10 days after tamoxifen
injection
Drp1 flox/flox
MHC-MerCreMer
Tamoxifen ip
n
HR (beats/min)
LVDd (X10-2mm)
LVDs (X10-2mm)
LVEF (%)
Ctr
+
+
5
504±14
318±11
178±15
81.9±4.8
Drp1-CKO
+
+
+
6
502±12
319±25
177±16
82.0±5.9
HR: Heart rate, LVDd: Left ventricular diastolic dimension, LVDs: Left ventricular systolic
dimension, LVEF: Left ventricular ejection fraction.
Online table VI. Measurement of organ weight parameters in Drp1-hetCKO mice
Drp1 flox/+
MHC-Cre
Age (Week)
n
BW (g)
Tibia length (mm)
LV weight (mg)
LV weight/Tibia length (mg/mm)
RV weight (mg)
RV/Tibia length (mg/mm)
Lung weight (mg)
Lung weight/Tibia length (mg/mm)
Liver weight (mg)
Liver weight/Tibia length (mg/mm)
+
12
4
22.1±2.4
18.2±0.3
73.5±5.7
4.04±0.27
13.0±3.8
0.71±0.21
147.0±7.4
8.1±0.5
882±88
48.5±4.9
BW: Body weight, LV: Left ventricle, RV: Right ventricle.
+
+
12
4
22.4±2.5
18.2±0.6
74.0±9.5
4.06±0.40
13.0±3.8
0.73±0.26
146.5±7.0
8.0±0.2
883±94
48.4±3.9
Online table VII. Measurement of organ weight parameters in mdivi-1-treated mice
Age (Week)
n
BW (g)
Tibia length (mm)
LV weight (mg)
LV weight/Tibia length (mg/mm)
RV weight (mg)
RV/Tibia length (mg/mm)
Lung weight (mg)
Lung weight/Tibia length (mg/mm)
Liver weight (mg)
Liver weight/Tibia length (mg/mm)
Ctr
12
4
26.4±1.3
18.3±0.2
88.8±7.7
4.84±0.40
18.3±5.1
1.00±0.28
145.0±18.6
7.9±1.0
858124
48.3±5.1
BW: Body weight, LV: Left ventricle, RV: Right ventricle.
mdivi-1
12
4
25.1±2.3
18.8±0.6
88.0±2.7
4.69±0.23
18.3±3.8
0.97±0.18
1461.5±11.5
7.5±0.6
878±152
47.2±8.4
Online Figure I
A
B
shScr shDrp1
80kDa
a-tubulin
50kDa
Relative Drp1
Expression
Drp1
TnI
Foreshortened
mitochondria
1.0
0.5
0
*
shscr shDrp1
Elongated
mitochondria
mt-DsRed2 Merge
C
shScr shDrp1
PGC1-a
90kDa
a-tubulin
50kDa
Online Figure I. Experimental validations. A, Construction of Ad-shDrp1.
Representative immunoblots for Drp1 and α-tubulin are shown. * p<0.01 vs. Ad-shScr.
The experiment was repeated 3 times. B, Definition of foreshortened and elongated
mitochondria. Mitochondria whose length is shorter than one sarcomere (the distance
between consecutive Zbands) are defined as foreshortened, those whose length is
longer than 2 sarcomeres are defined as elongated, and those whose length is longer
than one sarcomere and shorter than 2 sarcomeres are defined as intermediate (mid).
TnI: Troponin-I, mt-DsRed2: mitochondria-targeted DsRed2. C, Immunoblot for PGC-1α
and α-tubulin in CMs transduced with Ad-LacZ or Ad-shDrp1. The experiment was
repeated 3 times.
E
300
100
*
0
C
400
300
200
100
*
0
600
400
200
0
90
60
30
0
*
Relative basal OCR /
mt DNA
400
43.5
1.2
1.0
0.8
0.6
0.4
0.2
1.2
1.0
0.8
0.6
0.4
0.2
0.5
0
1.5
1.0
0.5
0
54.4
65.3
*
0
#
0
1.5
1.0
*
76.2
Relative basal OCR /
cell viability
32.6
Relative ATP linked OCR /
cell viability
21.8
FCCP
Relative maximum OCR /
cell viability
200
10.9
Relative ATP linked OCR /
mt DNA
OCR (pMol/min)
Oligomycin
Relative proton leak /
cell viability
Basal OCR (pMol/min)
0
Relative maximum OCR /
mt DNA
ATP linked OCR (pMol/min)
A
Relative proton leak /
mt DNA
Maximum OCR (pMol/min)
D
Proton leak (pMol/min)
Online Figure II
Rotenone
Antimycin A
600
shScr
500
shDrp1
400
300
200
100
0
Time (min)
87.0
97.9
B
1.2
1.0
0.8
0.6
0.4
0.6
0.4
0.5
0
1.5
1.0
0.5
0
*
0.2
0
1.2
1.0
0.8
*
0.2
0
1.5
1.0
*
Online Figure II. Downregulation of Drp1 inhibits mitochondrial function in CMs
as assessed with a Seahorse system. A, Representative changes in OCR in CMs in
response to oligomycin, FCCP, and rotenone plus antimycin A. Each data point
represents the mean of 4 replicates. B-E, Bar graphs showing basal OCR (B), ATPlinked OCR (C), maximum OCR (D) and proton leak (E). Experimental data were
normalized with mtDNA (middle columns) or cell viability (left columns). In all graphs,
data from CMs transduced with Ad-shScr is expressed as 1. *p<0.05, #p<0.01 vs. Ctr,
The experiment was repeated 3 times.
Online Figure III
A
B
Input
Input
Drp1
Drp1
Beclin1
Bcl-2
Bcl-2
Bcl-xL
Ad-HA-Drp1
Ad-LacZ
–
+
Bcl-xL
+
–
IP: HA
Ad-Flag-Beclin1
+
+
+
Ad-HA-Drp1
–
+
–
Ad-sh-Drp1
–
–
+
IP: Flag
Bcl-2
Bcl-2
Bcl-xL
Ad-HA-Drp1
–
Bcl-xL
+
Ad-HA-Drp1
+
–
+
+
+
–
Ad-shDrp1
–
–
+
Ad-Flag-Beclin1
C
shScr
shDrp1
shDrp1
+
shBcl-xL
D
shBcl-xL
shScr
Ctr
GD
4Hr
Chl
10mM
4Hr
†
60
50
40
†
†
* #
* #
*
#
†
* #
30
20
10
0
†
* #
†
Number of GFP-LC3 dots / cell
Number of GFP-LC3 dots / cell
GD
(-)
GD4Hr
#
*
170
150
130
*
40
*
20
0
Chl
-
+
shScr
Baseline
shDrp1
+
shBcl-xL
-
+
shDrp1
+
shBcl-xL
Online Figure III. Drp1 physically interacts with Bcl-2/Bcl-xL, thereby inhibiting
interaction between Beclin1 and Bcl-2/Bcl-xL. Downregulation of Drp1 inhibits
autophagy by stimulating interaction between Beclin1 and Bcl-2/Bcl-xL. A, CMs
were transduced with Ad-HA-Drp1 or Ad-LacZ. Forty-eight hours after transduction,
lysates were extracted for immunoprecipitation with HA antibody, followed by probing
with Bcl-2 or Bcl-xL antibodies. Representative images are shown. B, CMs were
transduced with Ad-Flag-Beclin1 either in the absence or presence of Ad-HA-Drp1 or
Ad-sh-Drp1. Seventy-two hours after transduction, lysates were extracted for
immunoprecipitation with Flag antibody, followed by probing with Bcl-2 or Bcl-xL
antibodies. Representative images are shown. In A and B, experiments were repeated 3
times. C, Representative images of GFP-LC3 puncta. Scale bar: 50 μm. Bar graph
indicates mean number of autophagosomes per cell. * p<0.01 vs. shScr without GD, #
p<0.01 vs. shDrp1 without GD, † p<0.01 vs. shDrp1 with GD. D, Representative images
of GFP-LC3 puncta in CMs incubated with chloroquine (10 μM) or vehicles for 4 hours.
Scale bar: 50 μm. Chl: chloroquine. Bar graph indicates mean number of
autophagosomes per cell. * p<0.01vs. shScr without Chl, # p<0.01 vs. shScr with Chl, †
p<0.01 vs. shDrp1+shBcl-xL without Chl. In C and D, 50 myocytes per group were
evaluated in each experiment and experiments were repeated 5 times.
A
Base
TnI
GD1Hr
mt-DsRed2
TnI
GD4Hr
mt-DsRed2
TnI
mt-DsRed2
B
Ctr
mdivi-1
(50 mM)
100
80
a
b
60
40
Elongated
Mid
Foreshortened
g
g
ab
20
c
c
0
Ctr
gh
e
ad
cf
mdivi-1
g
gh
cf
cf
*
0.8
0.6
0.4
0.2
0
1.2
1.0
*
0.8
0.6
* †
*#
0.4
0.2
0
Chelerythrine - + - +
10mM
shScr shDrp1
mdivi-1
(50 mM)
(DMSO)
(100 mM)
D
1.2
Ctr
*
1.0
(DMSO 1W)
0.8
0.6
0.4
mdivi-1
0.2
(50mM, 1W)
0
GD -
-
+
+
-
+
Relative cell viability (%)
DMSO mdivi-1 mdivi-1
50mM 100mM
(Ctr)
120
100
80
60
40
20
■
▲
*
Ctr
#
mdivi-1
0
Base 1
24
48 72 96
(hours)
Cells with elongated,
foreshortened
/ total cell number (%)
Relative cell viability
1.0
Relative cell viability
Cells with elongated,
foreshortened
/ total cell number (%)
(100 mM)
E
*††
1.2
Chelerythrine - +
- +
10mM
DMSO mdivi-1
(Ctr) 50 mM
mdivi-1
C
Relative cell viability
Online Figure IV
100
Elongated
Mid
Foreshortened
80
f
60
e
i
eh
i
eh
d
ac
d
bc
cd
Base
GD1Hr
GD4Hr
i
eg
40
20
ac
a
0
Base
GD1Hr
GD4Hr
Ctr
mdivi-1
(DMSO, 1W)
(50mM, 1W)
Online Figure IV
F
Ctr
Baseline
mdivi-1
4Days
GD
Baseline
Free red puncta
120
Yellow puncta
GD
d
100
GFP
LC3 dots / cell
RFP
80
60
d
e
f
a
40
a
b
c
20
Merge
e
0
Base
Ctr
GD
b
Base
GD
mdivi-1
Online Figure IV. GD induces mitochondrial fission in CMs treated with mdivi-1.A,
Assessment of mitochondrial morphology using mt-DsRed2. CMs were treated with
mdivi-1 (50 M) or DMSO (vehicle) as control. Insets show typical mitochondrial
morphology. Gray bar: cells with elongated/total cell number; black bar: cells with
foreshortened/total cell number; white bar: cells with intermediate (mid)/total cell
number . Base: baseline, Ctr: DMSO (control). a p<0.01 vs. Ctr foreshortened at
baseline, b p<0.01 vs. Ctr foreshortened after 1 hour GD, c p<0.01 vs. Ctr foreshortened
after 4 hours GD, d p<0.01 vs. mdivi-1 foreshortened at baseline, e p<0.01 vs. mdivi-1
after 1 hour GD, f p<0.01 vs. mdivi-1 foreshortened after 4 hours GD, g p<0.01 vs. Ctr
elongated at baseline, h p<0.01 vs. Ctr elongated after 4 hours GD (n=4/group). Scale
bar: 20 m. B, Upper panel: Cell viability of CMs with DMSO as a control or 50 M
mdivi-1. * p<0.01 vs. DMSO without chelerythrine, † p<0.01 vs. DMSO with 10 M
chelerythrine (n=4/group). Lower panel: Cell viability of CMs in Ad-shScr- or Ad-shDrp1transduced CMs. * p<0.01 vs. Ad-shScr without chelerythrine, # p<0.01 vs. Ad-shScr
with 10 M chelerythrine, † p<0.01 vs. Ad-shDrp1 without chelerythrine (n=4/group). C,
Cell viability of CMs with DMSO as a control or mdivi-1 at 2 different doses (50 M and
100 M). *p<0.05 vs Ctr or 50 M mdivi-1 with GD. D, Assessment of mitochondrial
morphology using mt-DsRed2. CMs were treated with mdivi-1 or the same volume of
DMSO as control for 4 days. Insets show representative mitochondria. Gray bar: cells
with elongated/total cell number; black bar: cells with foreshortened/total cell number;
white bar: cells with intermediate (mid)/total cell number. a p<0.01 vs. Ctr foreshortened
at baseline, b p<0.05 vs. Ctr foreshortened at baseline,.c p<0.01 vs. Ctr foreshortened
after 1 hour GD, d p<0.01 vs. Ctr foreshortened after 4 hours GD, e p<0.01 vs. Ctr
elongated at baseline, f p<0.05 vs. Ctr elongated at baseline, g p<0.01 vs. Ctr elongated
after 1 hour GD, h p<0.05 vs. Ctr elongated after 1 hour GD, i p<0.01 vs. Ctr elongated
after 4 hours GD (n=4/group). Scale bar: 20 m. E, Time course of cell viability in CMs
treated daily with DMSO or 50 M mdivi-1, as evaluated with the CellTiter Blue assay
(n=4/group). * p<0.01 vs. Ctr 72 hours after transduction, # p<0.01 vs. Ctr 96 hours after
transduction. F, Representative images of mRFP-GFP-LC3 puncta. Scale bar: 50 m.
Bar graph indicates mean number of autophagosomes and autolysosomes per cell. a
p<0.01 vs. Ctr at baseline, b p<0.01 vs. Ctr with 4 hours GD, c p<0.01 vs. mdivi-1 at
baseline, d p<0.01 vs. Ctr at baseline, e p<0.01 vs. Ctr with 4 hours GD, f p<0.01 vs.
mdivi-1 at baseline (n=3/group).
B
CCCP
25 mM
440 nm
shScr
+ GD
440 nm
560 nm
560 nm
Ratio
560/440 nm
Ratio
560/440 nm
C
shBeclin1
+ GD
TnI
Base
mt-DsRed2
TnI
GD1Hr
mt-DsRed2
TnI
High (560/440) signal area
/ cell area (%)
Online Figure V
A
Ctr
12
9
6
*
3
0
GD4Hr
mt-DsRed2
shScr
100
80
e
60
f
f
e
h
eg
40
ab
ab
20
cd
cd
Base
GD1Hr
0
Base
GD1Hr
shScr
GD4Hr
GD4Hr
shBeclin1
Elongated
Mid
Foreshortened
D
Relative cell viability
Cells with elongated,
foreshortened
/ total cell number (%)
shBeclin1
1.2
1.0
0.8
0.6
0.4
0.2
0
*
Online Figure V. Detection of CCCP-induced mitophagy using mitochondriatargeted Keima. A, Representative images of fluorescent Keima puncta in CMs with or
without 25 M CCCP treatment after transduction with adenovirus harboring
mitochondria-targeted Keima (Ad-Keima-MLS). Inset shows punctum with high 560/440
ratio. Scale bar: 20 m. B, CMs were transduced with Ad-Keima-MLS and either AdshScr or Ad-shBeclin1. Some were then subjected to 4 hours of GD. Inset shows
foreshortened mitochondria. * p<0.01 vs. Ad-Scr with GD (n=5/group). Scale bar: 20
μm. C, Assessment of mitochondrial morphology using mt-DsRed2. CMs were
transduced with Ad-shBeclin1 or Ad-shScr as control for 4 days. Insets show
representative mitochondria. Gray bar: cells with elongated/total cell number; black bar:
cells with foreshortened/total cell number; white bar: cells with intermediate (mid)/total
cell number. a p<0.01 vs. shScr foreshortened at baseline, b p<0.01 vs. shScr
foreshortened after 1 hour GD, c p<0.01 vs. shScr foreshortened after 4 hours GD, d
p<0.01 vs. shBeclin1 elongated after 4 hours GD, e p<0.01 vs. shScr elongated at
baseline, f p<0.01 vs. shScr elongated after 1 hour GD, g p<0.01 vs. shScr elongated
after 4 hours GD, h p<0.01 vs. shBeclin1 elongated at baseline (n=4/group). Scale bar:
20 m. D, Cell viability of CMs transduced with either Ad-shScr or Ad-shBeclin1 for 4
days followed by 4 hours GD. * p<0.01 vs. shScr with 4 hours GD (n=4/group).
B
LacZ
a-tubulin
50kDa
Relative Drp1
Expression
Drp1
80kDa
Drp1
TnI
*
4
mtDsRed2
2
0
Cells with elongated,
foreshortened
/ total cell number (%)
Online Figure VI
A
LacZ Drp1
100
60
*
40
20
0
LacZ Drp1
LacZ Drp1
LacZ
D
Drp1
LacZ
Drp1
E
Relative mt DNA
content
C
Elongated
Mid
Foreshortened
#
80
TUNEL
DAPI
0.8
*
0.4
0
*
8
4
0
LacZ Drp1
*
60
40
20
0
LacZ Drp1
Relative cell viability
12
Green fluorescent
cell number
/ total cell number (%)
% TUNEL positive
nuclei
LacZ Drp1
G
†
1.0
*#
0.8
0.6
0.4
0.2
0
shAtg7
-
+
LacZ
Ctr
440 nm
560 nm
Ratio
GD4Hr
+
Drp1
Drp1
Ctr
†
GD4Hr
High (560/440) signal area
/ cell area (%)
LacZ
F
-
* #
10.0
8.0
*
*
6.0
4.0
2.0
0
Ctr GD Ctr GD
LacZ
Drp1
Online Figure VI. Forced overexpression of Drp1 induces mitochondrial
dysfunction and apoptosis in CMs. A, Construction of Ad-Drp1. Representative
immunoblots for Drp1 and -tubulin are shown. * p<0.01 vs. Ad-LacZ. B, Assessment of
mitochondrial morphology using mt-DsRed2. The proportions of CMs with elongated
and foreshortened mitochondria were quantitated. TnI staining indicates CM. Gray bar:
cells with elongated /total cell number; black bar: cells with foreshortened /total cell
number; white bar: cells with intermediate (mid)/total cell number. * p<0.01 vs.
foreshortened in Ad-LacZ, # p<0.01 vs. elongated in Ad-LacZ (n=4/group). Scale bar:
20 m. C, TUNEL staining of CMs with overexpression of Drp1. * p<0.01 vs. Ad-LacZ
(n=3/group). Scale bar: 200 m. D, Mitochondrial membrane potential was evaluated
with JC-1. Red color indicates mitochondria in which membrane potential is maintained,
whereas green color indicates depolarized mitochondria. Quantification of CMs with
depolarized mitochondria is shown. * p<0.01 vs. Ad-LacZ (n=3/group). Yellow scale bar:
500 m; white scale bar: 100 m. E, Relative mitochondrial DNA content in CMs with
Drp1 overexpression, as evaluated by PCR for cytochrome b. * p<0.01 vs. Ad-LacZ
(n=3/group). F, Representative images of Keima fluorescent puncta after transduction
with Ad-Keima-MLS. A high 560/440 ratio indicates mitophagy. Ctr: control. The
proportions of the high ratio (560/440) signal area to the total cellular area are shown. *
p<0.01 vs. Ctr of Ad-LacZ, # p<0.01 vs. GD of Ad-LacZ, † p<0.01 vs. Ctr of Ad-Drp1
(n=5/group). Scale bar: 20 μm. G, Cell viability in Ad-LacZ- or Ad-Drp1-transduced
CMs, as evaluated with the CellTiter Blue assay. * p<0.01 vs. Ad-LacZ without AdshAtg7, # p<0.01 vs. Ad-LacZ with Ad-shAtg7, † p<0.01 vs. Ad-Drp1 without Ad-shAtg7
(n=8/group).
Online Figure VII
15 weeks
old
A
16 weeks
old
Tamoxifen
(20mg / ip)
for 5 days
Drp1-CKO mice
20 weeks
old
24 weeks
old
4 weeks
4 weeks
Drp1 MCM
Drp1 fl/fl
Control mice
Drp1 MCM
Drp1 fl/fl
B
Measured hemodynamics
and harvested tissue samples
C
Drp1
80kDa
Mfn-1
75kDa
α-tubulin
50kDa
Mfn-2
86kDa
100kDa
OPA1
D
86kDa
Survival rate (%)
100
17kDa
Fis1
80
α-tubulin
60
40
Drp1-CKO
20
Control
0
0
10
50kDa
Tamoxifen
-
+
-
+
Drp1 fl/fl
+
+
+
+
a-MHC MCM
-
-
+
+
30 (weeks)
20
Ctr
Tamoxifen ip
(15-16 weeks old)
E
Drp1 fl/fl
a-MHC MCM
Tamoxifen
Ctr
+
+
Drp1-CKO
+
+
+
F
-
Tamoxifen
+
Drp1 fl/fl
×
a-MHC MCM
(mm2)
*
400
300
200
100
0
Drp1 fl/fl
a-MHC MCM
Tamoxifen
+
+
+
+
+
8 weeks after
tamoxifen ip
*
% Fibrosis
Cross-sectional
area
Drp1 fl/fl
16
12
8
4
0
Tamoxifen
Drp1 fl/fl
a-MHC MCM
+
-
+
+
Ctr
+
+
+
+
+
Online Figure VII
G
Ctr
+
+
Drp1 fl/fl
a-MHC MCM
Tamoxifen
H
Drp1-CKO
+
+
+
-
Tamoxifen
+
a-MHC
MCM
WT
*
Cross-sectional
area
% Fibrosis
20
15
10
5
0
Drp1 fl/fl
+
+
a-MHC MCM
-
+
Tamoxifen
+
+
8 weeks after
tamoxifen ip
I
100
0
-
+
-
+
a-MHC MCM
-
-
+
+
J
Tamoxifen
+
80
a-MHC
MCM
WT
2
% Fibrosis
200
Tamoxifen
LVEF (%)
-
(mm2)
1
0
Tamoxifen
-
+
-
+
a-MHC MCM
-
-
+
+
60
40
20
0
Tamoxifen
-
+
-
+
a-MHC MCM
-
-
+
+
Online Figure VII. Basal characterization of Drp1-CKO mice. A, A scheme of the
experimental protocol. MCM: MerCreMer B, Immunoblots for Drp1 and -tubulin in
Drp1-CKO mice (n=3/group). C, Immunoblots for factors related to mitochondrial
dynamics in Drp1-CKO and control mice (n=3/group). D, Kaplan–Meier curve of Drp1CKO and control mice. p<0.05 vs. control (n=8/group). E, Assessment of CM crosssectional area in control and Drp1-CKO mice using WGA staining 8 weeks after
tamoxifen injection. Scale bar: 200 m. * p<0.01 vs. Ctr (n=3/group). F, Masson’s
trichrome staining of sections from Drp1-CKO and control mice. * p<0.01 vs. controls
(n=4/group). Scale bar: 500 m. G, Assessment of CM fibrosis in control and Drp1-CKO
mice using Picric Acid Sirius Red (PASR) staining 8 weeks after tamoxifen injection.
Scale bar: 500 m. * p<0.01 vs. Ctr (n=3/group). H, Assessment of CM size in TgMHC-MerCreMer and wild-type mice with or without tamoxifen using WGA staining.
Scale bar: 200 m. There was no significant difference between the 4 groups of mice
(n=4/group). WT: wild-type. I, Assessment of fibrosis in Tg-MHC-MerCreMer and wildtype mice with or without tamoxifen using PASR staining. Scale bar: 500 m. There was
no significant difference between the 4 groups (n=4/group). J, LV ejection fractions
(LVEF), as evaluated with echocardiography 4 weeks after tamoxifen injection
(n=4/group). In B, C and F, heart samples were harvested 4 weeks after tamoxifen
injection. In E and G, heart samples were harvested 8 weeks after tamoxifen injection.
B
C
tamoxifen ip
Ctr
Drp1-CKO
Ctr
Tamoxifen ip
*
*
*
Drp1-CKO
4W
8W
4W
PGC1-a
90kDa
a-tubulin
50kDa
8W
COX IV
17kDa
a-Tubulin
50kDa
Ctr
*
6
Relative COX IV
expression
Relative
Mitochondrial
mass
D
4
2
0
+
+
Drp1 fl/fl
a-MHC MCM
Tamoxifen
+
+
+
*# *#
3
2
Drp1-CKO
1.2
Relative ATP
production
Online Figure VIII
A
8 weeks after
0.8
*
0.4
0
1
Tamoxifen
+
+
-
+
+
+
Ctr
Drp1
CKO
Drp1 fl/fl
0
4W
Ctr Drp1
CKO
8W
Ctr
4W
a-MHC MCM
8W
Drp1-CKO
*
Tamoxifen
+
+
-
Drp1 fl/fl
a-MHC MCM
1.2
1.0
0.8
0.6
0.4
0.2
0
*
Tamoxifen
+
+
+
Drp1 fl/fl
a-MHC MCM
Ctr Drp1
CKO
+
+
-
+
+
+
Ctr
Drp1
CKO
1.2
1.0
0.8
0.6
0.4
0.2
0
*
Ctr
Drp1-CKO
TUNEL
Tamoxifen
Drp1 fl/fl
a-MHC MCM
+
+
-
+
+
+
Ctr
Drp1
CKO
DAPI
8 weeks after
tamoxifen ip
8 weeks after
tamoxifen ip
8 weeks after
tamoxifen ip
H
% TUNEL positive
nuclei
1.2
1.0
0.8
0.6
0.4
0.2
0
Relative complex IV
activity
Relative complex I
activity
E
Relative complex II+III
activity
8 weeks after
tamoxifen ip
*
2.0
1.5
1.0
0.5
0
+
+
+
+
+
Ctr
Drp1
CKO
Drp1 fl/fl
F
Ctr
Drp1-CKO
+
+
+
+
+
Drp1 fl/fl
a-MHC MCM
Tamoxifen
G
Drp1 fl/fl
a-MHC MCM
Tamoxifen
Ctr
Drp1-CKO
+
+
+
+
+
a-MHC MCM
Tamoxifen
8 weeks after
tamoxifen ip
I
Serum HMGB1
concentration
(mm2)
200
% Fibrosis
Cross-sectional
area
(ng/ml)
100
0
Drp1 fl/fl
a-MHC MCM
Tamoxifen
+
+
+
+
+
10 days after
tamoxifen ip
1.5
1
0.5
0
Drp1 fl/fl
a-MHC MCM
Tamoxifen
+
+
+
+
+
10 days after
tamoxifen ip
*
12
8
4
0
Drp1 fl/fl
a-MHC MCM
Tamoxifen
+
+
Ctr
+
+
+
Drp1
CKO
4 weeks after
tamoxifen ip
Online Figure VIII. Hypertrophy and mitochondrial dysfunction in Drp1-CKO mice.
A, Electron microscope images of Drp1-CKO and control mouse hearts 8 weeks after
tamoxifen injection. Asterisks indicate elongated mitochondria. Mitochondrial mass in
control mouse hearts is expressed as 1. * p<0.01 vs. Ctr (n=3/group). Scale bar: 2 m.
B, Immunoblots for COX IV and -tubulin in Drp1-CKO and control mice. * p<0.01 vs.
Ctr 4weeks after tamoxifen ip, # p<0.01 vs. Ctr 8weeks after tamoxifen ip (n=3/group).C,
Immunoblot for PGC-1 and -tubulin in Drp1-CKO and control mice whose hearts
were harvested 4 or 8 weeks after tamoxifen injection. D, Relative cardiac ATP
production in Drp1-CKO and age-matched control mice 8 weeks after tamoxifen
injection. * p<0.01 vs. Ctr (n=3/group). E, Respiratory chain complex activity in
mitochondria from mouse hearts. Relative respiratory chain complex I, II+II, and IV
activities are shown. The activity in mitochondria from Ctr mouse hearts is expressed as
1. *p<0.05 vs. Ctr (n=4/group). F, Assessment of CM size in control and Drp1-CKO
mice using WGA staining 10 days after tamoxifen injection. Scale bar: 200 m. There
was no significant difference between the 2 groups (n=3/group). G, Assessment of CM
fibrosis in control and Drp1-CKO mice using PASR staining 10 days after tamoxifen
injection. Scale bar: 500 m. There was no significant difference between the 2 groups
(n=3/group). H, TUNEL staining of hearts in Drp1-CKO and control mice 8 weeks after
tamoxifen injection. Arrows indicate TUNEL-positive nuclei. * p<0.01 vs. Ctr. Scale bar:
50 μm. I, Serum HMGB1 concentration in Drp1-CKO and age-matched control mice 4
weeks after tamoxifen injection was assessed by ELISA. * p<0.05 vs. Ctr (n=3/group).
In A-E and H, samples were harvested 8 weeks after tamoxifen injection. In B and I,
samples were harvested 4 weeks after tamoxifen injection.
Ctr
B
Drp1
hetCKO
Ctr
Drp1
hetCKO
(mm2)
200
100
0
% Fibrosis
*
#
40
20
WT
6000
*
*#
4000
40
2000
20
0
Ctr
E
Drp1
CKO
AAR (%)
40
30
20
10
Ctr
Drp1
CKO
Ctr
Drp1-CKO
Infarct / AAR (%)
Ctr
60
a-MHC Cre
8000
0
0
Drp1
hetCKO
0.4
AAR (%)
LVEF (%)
*
Ctr
0.8
D
LV +dP/dt (mmHg/sec)
C
60
Drp1
hetCKO
1.2
0
80
Ctr
*
40
20
0
Ctr Drp1
CKO
Drp1
CKO
Infarct / AAR (%)
A
Cross-sectional area
Online Figure IX
40
20
0
Online Figure IX. Basal characterization of Drp1-hetCKO mice. A, Assessment of
CM size using WGA staining. Ctr: control. (n=3/group). Scale bar: 200 m. B, PASR
staining to assess cardiac fibrosis. (n=3/group). Scale bar: 500 m. C, LV ejection
fraction, as evaluated with echocardiography, and +dP/dt, as evaluated by
hemodynamic measurement. * p<0.01 vs. Ctr at baseline, # p<0.01 vs. Drp1-hetCKO at
baseline. Fst: fasting (n=4/group). D, Representative images of heart sections with I/R
injury. WT: wild-type. Statistical analysis of % area at risk (AAR) and ratio of infarct size
to AAR are shown. There was no significant difference between Tg-MHC-Cre and
wild-type mice (n=3/group). E, Representative images of TTC/Alcian Blue staining of LV
sections after I/R. All of the mice underwent I/R surgery 10 days after tamoxifen
injection. Statistical analyses of AAR and infarct size/AAR are shown. * p<0.05 vs. Ctr
(n=3/group).
Online Figure X
A
Ctr
(DMSO)
B
mdivi-1
50mM
Drp1-hetCKO
mdivi-1
-
C
+
10
0
30
20
10
0
30
20
10
0
mdivi-1 -
+
Ctr
mdivi-1
Ctr
(DMSO
1week)
(1 week)
(Drp1 fl/+)
LVEF (%)
0
20
0
Drp1hetCKO
OD540nm
Ctr (DMSO 1W)
0.55
Drp1-hetCKO
0.50
mdivi1 (1W)
0.45
0
0
4
8 12 16 20
Time (min)
AAR (%)
40
30
20
10
0
Ctr
mdivi-1
(DMSO, 1W)
(50mM, 1W)
Infarct / AAR (%)
G
50
40
30
20
10
0
*
% Decrease in OD540
*
Ctr (Drp1 fl/+)
*
* †
6
4
#
2
0
+
50mM
*
0.60
40
40
*
E
60
20
*
mdivi-1 -
50mM
D
60
Relative
mitochondrial mass
20
40
*
F
Relative ATP production
30
40
Infarct / AAR (%)
AAR (%)
40
AAR (%)
Infarct / AAR (%)
80
2
*
#
1
0
1.2
1.0
0.8
0.6
0.4
0.2
0
*
#
Online Figure X. Chronic treatment with mdivi-1 leads to mitochondrial
dysfunction and exacerbates I/R injury. A, Representative images of TTC/Alcian
Blue staining of LV sections after I/R. Mice were injected with mdivi-1 at 1.2 mg/kg or
the same volume of DMSO once 30 minutes prior to myocardial ischemia. Statistical
analyses of % area at risk (AAR) and infarct size/AAR are shown. * p<0.05 vs. Ctr
(DMSO ip once) (n=3/group). B, Representative images of TTC/Alcian Blue staining of
LV sections after I/R. Drp1-hetCKO mice were treated with mdivi-1 at 1.2 mg/kg or the
same volume of DMSO once 30 minutes prior to myocardial ischemia. Statistical
analyses of AAR and infarct size/AAR are shown. * p<0.05 vs. Drp1-hetCKO without
mdivi-1 (n=3/group). C, LVEF of mice treated with mdivi-1 at 1.2 mg/kg or the same
volume of DMSO. There was no significant difference in LVEF between control and
mdivi-1-treated mice (n=3/group). D, Electron microscope images of hearts from mdivi1-treated, Drp1-hetCKO and control mice. Mdivi-1 at 1.2 mg/kg or the same volume of
DMSO was injected intraperitoneally for 1 week in the mdivi-1-treated and control
groups, respectively. Asterisks indicate elongated mitochondria. Mitochondrial mass in
control (injected with DMSO) mouse hearts is expressed as 1. * p<0.05 vs. Ctr (DMSO),
# p<0.01 vs. Ctr (Drp1 fl/+) (n=3/group). Scale bar: 2 m. E, Mitochondrial swelling
induced by Ca2+. Each data curve in the left panel represents the average of 3 individual
measurements. Right panel shows the decrease in optical density at 540 nm. * p<0.01
vs. Ctr (DMSO ip for 1 week), # p<0.01 vs. mdivi-1 ip for 1 week, † p<0.01 vs. Ctr (Drp1
fl/+) (n=3/group). F, Relative cardiac ATP production in mdivi-1 treated, Drp1-hetCKO
and control mice. Mdivi-1 at 1.2 mg/kg or the same volume of DMSO was injected
intraperitoneally for 1 week in the mdivi-1-treated and control groups, respectively. *
p<0.01 vs. Ctr (DMSO), # p<0.01 vs. Ctr (Drp1 fl/+) at 12 weeks of age (n=3/group). G,
Representative images of TTC/Alcian Blue staining of LV sections after I/R. Mice
underwent treatment with mdivi-1 at 1.2 mg/kg or the same volume of DMSO for 1
week. Statistical analyses of % area at risk (AAR) and infarct size/AAR are shown. *
p<0.05 vs. Ctr (DMSO ip for 1 week) (n=3/group).