cardiovascular biology copyright © 2018 … › content › sigtrans › 11 › 556 › eaat...gets...

15
Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018 SCIENCE SIGNALING | RESEARCH ARTICLE 1 of 14 CARDIOVASCULAR BIOLOGY Hypoxia-induced interaction of filamin with Drp1 causes mitochondrial hyperfission–associated myocardial senescence Akiyuki Nishimura 1,2,3,4 , Tsukasa Shimauchi 1,2,5 , Tomohiro Tanaka 1,2 , Kakeru Shimoda 1,2,3 , Takashi Toyama 1,4,6 , Naoyuki Kitajima 1,4 , Tatsuya Ishikawa 4,7 , Naoya Shindo 4 , Takuro Numaga-Tomita 1,2,3 , Satoshi Yasuda 8 , Yoji Sato 4,8 , Koichiro Kuwahara 9 , Yoshito Kumagai 6 , Takaaki Akaike 10 , Tomomi Ide 5 , Akio Ojida 4 , Yasuo Mori 11 , Motohiro Nishida 1,2,3,4 * Defective mitochondrial dynamics through aberrant interactions between mitochondria and actin cytoskeleton is increasingly recognized as a key determinant of cardiac fragility after myocardial infarction (MI). Dynamin-related protein 1 (Drp1), a mitochondrial fission–accelerating factor, is activated locally at the fission site through interactions with actin. Here, we report that the actin-binding protein filamin A acted as a guanine nucleotide exchange factor for Drp1 and mediated mitochondrial fission–associated myocardial senescence in mice after MI. In peri-infarct regions characterized by mitochondrial hyperfission and associated with myocardial senescence, filamin A colocalized with Drp1 around mitochondria. Hypoxic stress induced the interaction of filamin A with the GTPase domain of Drp1 and increased Drp1 activity in an actin-binding–dependent manner in rat cardiomyocytes. Expression of the A1545T filamin mutant, which potentiates actin aggregation, promoted mitochondrial hyperfission under normoxia. Furthermore, pharmacological perturbation of the Drp1–filamin A interaction by cilnidipine suppressed mitochon- drial hyperfission–associated myocardial senescence and heart failure after MI. Together, these data demonstrate that Drp1 association with filamin and the actin cytoskeleton contributes to cardiac fragility after MI and suggests a potential repurposing of cilnidipine, as well as provides a starting point for innovative Drp1 inhibitor development. INTRODUCTION Mitochondria are dynamic organelles that interchange their mor- phology between two distinct arrangements by undergoing fission to generate an elongated interconnected mitochondrial network or fusion to give rise to a fragmented discrete phenotype (1). Although mitochondrial fission and fusion networks are absent and mobility is not normally observed in healthy adult cardiomyocytes, mitochon- drial fusion and fission proteins are abundant and indispensable for physiological functions of the heart (2). Defects in mitochondrial dynamics are implicated in the development of cardiovascular diseases (3), as well as neurodegenerative diseases. Although many studies using genetically manipulated mice and/or chemical re- agents have highlighted the potential of mitochondrial fusion and fission proteins as therapeutic targets for combating cardiovascular diseases (49), drugs targeting these proteins have not been devel- oped, under the assumption that direct inhibition of fission or fusion proteins will eventually impair the balance between mito- chondrial fission and fusion (10). Originally defined as terminal cell growth arrest, cellular senes- cence is now appreciated to participate in various biological processes such as development, aging, and age-related diseases (11). Cells undergoing senescence show distinctive functional alterations includ- ing telomere shortening, secretome changes, increases in senescence- associated -galactosidase (SA--gal) activity, and changes in the expression of various genes. Although cardiomyocytes are terminally differentiated, SA--gal–positive cardiomyocytes are increased in aged heart (12). Senescent cardiomyocytes show decreased contract- ile performance due to altered abundances of contractile proteins and Ca 2+ -handing proteins, leading to maladaptive changes in myocardial structure and function (13). Moreover, p53, a key medi- ator of senescence, accumulates in aged heart and promotes mito- chondrial dysfunction by inhibiting Parkin-mediated mitophagy, leading to cardiac dysfunction (14). Premature myocardial senes- cence, which is observed in several cardiac disease models including myocardial infarction (MI), has been suggested as a major cause of cardiac dysfunction (121516). The senescence-associated microRNA (miRNA) miR-34a, which is transcriptionally activated by p53 (17), is increased after MI, and miRNA-34a inhibition reduces DNA dam- age responses and improves myocardial function through its target phosphatase-1 nuclear targeting subunit (PNUTS) after MI (16). How- ever, the mechanisms underlying defect in mitochondrial dynamics for the induction of premature myocardial senescence are obscure. Mitochondrial fission is specifically mediated by the dynamin- related guanosine triphosphatase (GTPase) protein 1 (Drp1) (18), which translocates from the cytosol to the outer mitochondrial membrane where it interacts with receptor proteins (19), including fission protein-1 (Fis1), mitochondrial fission factor (Mff), and mito- chondrial dynamics proteins of 49 and 51 kDa (MiD49 and MiD51, respectively). Translocated Drp1 oligomerizes and encircles a mito- chondrion to constrict the mitochondrial membrane, after which dynamin-2 triggers membrane scission as the final step of mitochon- drial division (20). Translocation of Drp1 is regulated by several 1 National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan. 2 Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan. 3 SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan. 4 Graduate School of Pharmaceutical Sciences, Kyushu Uni- versity, Fukuoka 812-8582, Japan. 5 Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan. 6 Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan. 7 EA Pharma Co. Inc., Tokyo 104-0042, Japan. 8 National Institute of Health Sciences, Kanagawa 210-9501, Japan. 9 Shinshu University School of Medicine, Nagano 390-8621, Japan. 10 Tohoku University Graduate School of Medi- cine, Sendai 980-8575, Japan. 11 Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan. *Corresponding author. Email: [email protected] Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works on July 15, 2020 http://stke.sciencemag.org/ Downloaded from

Upload: others

Post on 28-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

1 of 14

C A R D I O V A S C U L A R B I O L O G Y

Hypoxia-induced interaction of filamin with Drp1 causes mitochondrial hyperfission–associated myocardial senescenceAkiyuki Nishimura1,2,3,4, Tsukasa Shimauchi1,2,5, Tomohiro Tanaka1,2, Kakeru Shimoda1,2,3, Takashi Toyama1,4,6, Naoyuki Kitajima1,4, Tatsuya Ishikawa4,7, Naoya Shindo4, Takuro Numaga-Tomita1,2,3, Satoshi Yasuda8, Yoji Sato4,8, Koichiro Kuwahara9, Yoshito Kumagai6, Takaaki Akaike10, Tomomi Ide5, Akio Ojida4, Yasuo Mori11, Motohiro Nishida1,2,3,4*

Defective mitochondrial dynamics through aberrant interactions between mitochondria and actin cytoskeleton is increasingly recognized as a key determinant of cardiac fragility after myocardial infarction (MI). Dynamin-related protein 1 (Drp1), a mitochondrial fission–accelerating factor, is activated locally at the fission site through interactions with actin. Here, we report that the actin-binding protein filamin A acted as a guanine nucleotide exchange factor for Drp1 and mediated mitochondrial fission–associated myocardial senescence in mice after MI. In peri-infarct regions characterized by mitochondrial hyperfission and associated with myocardial senescence, filamin A colocalized with Drp1 around mitochondria. Hypoxic stress induced the interaction of filamin A with the GTPase domain of Drp1 and increased Drp1 activity in an actin-binding–dependent manner in rat cardiomyocytes. Expression of the A1545T filamin mutant, which potentiates actin aggregation, promoted mitochondrial hyperfission under normoxia. Furthermore, pharmacological perturbation of the Drp1–filamin A interaction by cilnidipine suppressed mitochon-drial hyperfission–associated myocardial senescence and heart failure after MI. Together, these data demonstrate that Drp1 association with filamin and the actin cytoskeleton contributes to cardiac fragility after MI and suggests a potential repurposing of cilnidipine, as well as provides a starting point for innovative Drp1 inhibitor development.

INTRODUCTIONMitochondria are dynamic organelles that interchange their mor-phology between two distinct arrangements by undergoing fission to generate an elongated interconnected mitochondrial network or fusion to give rise to a fragmented discrete phenotype (1). Although mitochondrial fission and fusion networks are absent and mobility is not normally observed in healthy adult cardiomyocytes, mitochon-drial fusion and fission proteins are abundant and indispensable for physiological functions of the heart (2). Defects in mitochondrial dynamics are implicated in the development of cardiovascular diseases (3), as well as neurodegenerative diseases. Although many studies using genetically manipulated mice and/or chemical re-agents have highlighted the potential of mitochondrial fusion and fission proteins as therapeutic targets for combating cardiovascular diseases (4–9), drugs targeting these proteins have not been devel-oped, under the assumption that direct inhibition of fission or fusion proteins will eventually impair the balance between mito-chondrial fission and fusion (10).

Originally defined as terminal cell growth arrest, cellular senes-cence is now appreciated to participate in various biological processes

such as development, aging, and age-related diseases (11). Cells undergoing senescence show distinctive functional alterations includ-ing telomere shortening, secretome changes, increases in senescence- associated -galactosidase (SA--gal) activity, and changes in the expression of various genes. Although cardiomyocytes are terminally differentiated, SA--gal–positive cardiomyocytes are increased in aged heart (12). Senescent cardiomyocytes show decreased contract-ile performance due to altered abundances of contractile proteins and Ca2+-handing proteins, leading to maladaptive changes in myocardial structure and function (13). Moreover, p53, a key medi-ator of senescence, accumulates in aged heart and promotes mito-chondrial dysfunction by inhibiting Parkin-mediated mitophagy, leading to cardiac dysfunction (14). Premature myocardial senes-cence, which is observed in several cardiac disease models including myocardial infarction (MI), has been suggested as a major cause of cardiac dysfunction (12, 15, 16). The senescence-associated microRNA (miRNA) miR-34a, which is transcriptionally activated by p53 (17), is increased after MI, and miRNA-34a inhibition reduces DNA dam-age responses and improves myocardial function through its target phosphatase-1 nuclear targeting subunit (PNUTS) after MI (16). How-ever, the mechanisms underlying defect in mitochondrial dynamics for the induction of premature myocardial senescence are obscure.

Mitochondrial fission is specifically mediated by the dynamin- related guanosine triphosphatase (GTPase) protein 1 (Drp1) (18), which translocates from the cytosol to the outer mitochondrial membrane where it interacts with receptor proteins (19), including fission protein-1 (Fis1), mitochondrial fission factor (Mff), and mito-chondrial dynamics proteins of 49 and 51 kDa (MiD49 and MiD51, respectively). Translocated Drp1 oligomerizes and encircles a mito-chondrion to constrict the mitochondrial membrane, after which dynamin-2 triggers membrane scission as the final step of mitochon-drial division (20). Translocation of Drp1 is regulated by several

1National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan. 2Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan. 3SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan. 4Graduate School of Pharmaceutical Sciences, Kyushu Uni-versity, Fukuoka 812-8582, Japan. 5Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan. 6Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan. 7EA Pharma Co. Inc., Tokyo 104-0042, Japan. 8National Institute of Health Sciences, Kanagawa 210-9501, Japan. 9Shinshu University School of Medicine, Nagano 390-8621, Japan. 10Tohoku University Graduate School of Medi-cine, Sendai 980-8575, Japan. 11Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan.*Corresponding author. Email: [email protected]

Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

2 of 14

different posttranslational modifications, including phosphoryl ation (7), SUMOylation (21), ubiquitination (22), S-nitrosylation (23), and O-GlcNAcylation (24). In addition, we have reported that sul-fur deprivation-polysulfidation cycling of cysteine residues in Drp1 regulates its GTPase activity (25). Mitochondrial fusion is also regulated by other mitochondrial membrane–localized GTPases, namely, mitofusins (Mfn1 and Mfn2) in the outer mitochondrial membrane and optic atrophy 1 (Opa1), an inner membrane protein that governs inner mitochondrial membrane fusion (3, 18). These GTPases play important roles in maintaining mitochondrial quality control and the functional integrity of the heart (4, 5, 26, 27).

Drp1 maturation has been suggested to require the interactions of mitochondria with other organelles such as the endoplasmic reticulum (ER) (28), lysosome (29), and cytoskeleton (30–33). For example, mitochondria-ER contact precedes Drp1 oligomerization on mitochondrial membranes (28). Cytoskeletal proteins, including myosin II and actin filaments, promote Drp1 assembly at fission sites, leading to efficient mitochondrial fragmentation (31–33). However, it remains unclear whether interactions of Drp1 with actin or myo-sin are sufficient to promote Drp1 activity and assembly at fission sites. It also remains unclear why basal Drp1 activity and mitochon-drial fission are not enhanced in actin- and myosin-enriched muscle cells and actin-reorganized cells compared with nonmuscle cell lines. Cells may express a protein that anchors Drp1 to the actomyosin cytoskeleton under pathological conditions. In this study, we identi-fied the actin-binding protein filamin A (FLNa) as a binding partner of Drp1 and demonstrated that FLNa acted as a guanine nucleotide exchange factor (GEF) for Drp1 in coordination with actin cyto-skeleton. FLNa overexpression promoted protein complex formation between Drp1 and the actin cytoskeleton at mitochondrial fission sites, leading to progression of hypoxia-induced mitochondrial hyper-fission in cardiomyocytes and fibroblasts. Mitochondrial fusion and fission proteins have been suggested to be potential therapeutic tar-gets for combating cardiovascular disease, but clinically applicable drugs have not been identified to date. In this study, we identified cilnidipine as a specific inhibitor of hypoxia-induced mitochondrial hyperfission, a drug initially characterized as a dihydropyridine (DHP)–derivative voltage-dependent L/N-type Ca2+ channel blocker. Using this drug, we demonstrated that cilnidipine disrupted hypoxia- induced Drp1-filamin interaction and improved cardiac function after MI and suggested the potential repurposing of cilnidipine for the treatment of ischemic diseases.

RESULTSPremature myocardial senescence associated with Drp1-dependent mitochondrial fission is induced in the peri-infarct region of mouse hearts after MIWe have previously reported that formation of reactive oxygen species (ROS)–dependent electrophiles, such as 8-nitroguanosine 3′5′-cyclic monophosphate (8-NO2-cGMP), mediates H-Ras–dependent myo-cardial early senescence at the late stage of heart failure after MI (34). Here, we identified the region of the left ventricular (LV) myocardium that was the predominant cause of myocardial senescence after MI. The SA--gal–positive cell area was significantly increased in size the infarct zone and peri-infarct zone of the LV myocardium at 4 weeks after MI (Fig. 1A and fig. S1A). To identify the molecular event upstream of cardiac senescence, we analyzed MI heart sam-ples at an earlier time point. Sarcomere structures in the infarct zone

were almost absent, and myocardium was eventually replaced by myofibroblasts by 1 week after MI. In contrast, sarcomere structures in the peri-infarct zone were not disrupted, and almost all striated cardiomyocytes were still healthy (Fig. 1B). Electron microscopic analysis also revealed that mitochondria in peri-infarct zone myocar-dium were smaller and more rounded than those in sham-operated myocardium (Fig. 1, B and C, and fig. S1B). Although blood flow was apparently supplied properly to the peri-infarct zone, the pro-tein abundances of hypoxia-inducible factor 1 (HIF-1) and heme oxygenase-1 (HO-1) were significantly increased in the peri-infarct zone myocardium at 1 but not 4 weeks after MI (fig. S2A), indicating that hypoxic stress was transiently induced at 1 week after MI. Because the abundances of mitochondrial proteins, such as Mn- superoxide dismutase and tafazzin, did not change (fig. S2A), moder-ate hypoxic stress may cause mitochondrial fragmentation but not decreased mitochondrial protein quantity. Hypoxic stress increases Drp1 activity (8, 35), and accordingly, the GTP-binding activity of Drp1 was significantly increased, although Drp1 protein abundance was slightly reduced in the peri-infarct zone myocardium at 1 week after MI (Fig. 1D). Exposing neonatal rat cardiomyocytes (NRCMs) to 1% hypoxia significantly increased the GTP-binding activity of Drp1 in vitro (Fig. 1E). The GTP-binding activities of the fusion- accelerating proteins Mfn1, Mfn2, and Opa1 were not changed in the peri-infarct zone myocardium compared with sham-operated myocardium (fig. S2B) or NRCMs with hypoxia (fig. S2C). Consist-ent with previous report about reduced Opa1 protein levels in human heart with ischemic cardiomyopathy (36), Opa1 protein abundance was slightly reduced after MI (fig. S2B). These results suggest that Drp1 activity is the main determinant of mitochondrial morphology after MI. Depending on the mitochondrial fragment length, we classified the mitochondrial morphology of NRCMs into three forms: tubule, intermediate, and vesicle (fig. S3, A and B). Con-sistent with Drp1 activation (Fig. 1E), hypoxia treatment significantly increased the number of NRCMs with vesicle-type mitochondrial fragmentation compared to those with tubule-type fragmentation, which was the predominant mitochondrial morphology in normoxic NRCMs (Fig. 1F). Hypoxic stress was reduced at 4 weeks after MI (fig. S2A), which is also when myocardial senescence occurred (Fig. 1A). ROS are critical to inducing cellular senescence (37), and hypoxia/reoxygenation but not hypoxia alone increased ROS production in NRCMs (fig. S4A). Hypoxia itself did not induce NRCM senescence (fig. S4B), whereas hypoxia/reoxygenation treatment sig-nificantly increased the number of SA--gal–positive cardiomyocytes (Fig. 1G). We also used p53 as an alternative senescence marker because cytosolic p53 is increased in abundance in aged hearts (14). The popu-lation of cells containing p53 puncta in - actinin–positive NRCMs was significantly increased in cells subjected to hypoxia/ reoxygenation (fig. S4C). Hypoxia/reoxygenation- induced myocardial senescence was attenuated by application of the mitochondria- targeted super-oxide scavenger (2-(2,2,6,6- tetramethylpiperidin- 1-oxyl- 4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mito- TEMPO) (fig. S4D). These results suggest that hypoxia-induced mitochondrial fission after mitochondrial ROS production is involved in the induction of senescence, although it remains unclear whether this cascade is suf-ficient for senescence. Knockdown of Drp1 by one of two independent siRNAs (fig. S5A) prevented hypoxia-induced mitochondrial fragmen-tation (fig. S5B) and hypoxia/reoxygenation-induced senescence of NRCMs (Fig. 1G). In addition, the hypoxia/reoxygenation- induced cardiomyocyte senescence and mitochondrial ROS production were

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

3 of 14

significantly suppressed by the Drp1 inhibitor Mdivi-1 (fig. S5, C and D) (38). These results suggest that hypoxic stress–induced Drp1 activation par-ticipates in mitochondrial fragmentation–associated premature senes-cence of cardiomyocytes in the peri-infarct zone myocardium after MI.

Drp1 associates with FLNa under hypoxiaTo investigate how Drp1 is activated under hypoxic conditions, we screened for proteins that bound to Drp1 in a hypoxia-dependent manner. Proteomic analyses revealed that endogenous cytoskeletal proteins, such as the actin-binding protein FLNa (39) and -actin,

were cross-linked and coprecipitated with FLAG-tagged Drp1 in HeLa cells and that these interactions were increased after exposure to hypoxic stress (Fig. 2A and fig. S6). To examine whether hypoxic stress induced the direct interaction of Drp1 with these cytoskeletal proteins, we performed immunoprecipitation assays without a cross- linker. Hypoxic stress did not promote the interaction of FLAG-Drp1 with endogenous FLNa but not with endogenous -actin (Fig. 2B), suggesting that only FLNa directly associates with Drp1. Dephos-phorylation of Drp1 at Ser637 participates in mitochondrial fission, and hypoxic stress induces dephosphorylation of Drp1 Ser637 in

GTP-Drp1

3

GTP

-Drp

1/in

put-D

rp1

(fold

)

2

1

0

Input-Drp1

D

MI

Sham

Sham MI (peri-IZ)

IF

PN

B MI (IZ)

NN

N

A Sham

MI

1: Sham 2: MI (IZ)

3: MI (peri-IZ) 4: MI (RZ)

Normoxia Hypoxia

Nor

mox

iaH

/R

F G

80

60

40

20

0% o

f tot

al c

ells

HypoxiaNormoxia

Vesicl

eInt

er-

mediat

eTub

ule

∗∗

siNC siDrp1 #1

− +H/R0

10

20

30

SA

- β-g

al–p

ositi

ve c

ells

(%)

siNC

∗∗siDrp1 #240

− + − +#1 #2siDrp1

∗∗∗∗

4Sham MI∗∗ E

GTP-Drp1

N H

N H012345

Input-Drp1

GTP

-Drp

1/in

put-D

rp1

(fold

) 6∗

C

GAPDH

1

23

4

1.5

Drp

1 ex

pres

sion

/GA

PD

H (f

old)

1.0

0.5

0 MI

Sham

2.0∗∗

MI

ShamM

itoch

ondr

ial a

rea

(µm

2 )

3

2

1

0 MI

Sham

Circ

ular

ity

0

0.5

1.0∗∗ ∗∗

Fig. 1. Mitochondrial fission–associated premature myocardial senescence in peri-infarct zone myocardium after MI. (A) Myocardial senescence determined by SA--gal staining in mouse heart 4 weeks after MI (n = 5 mice per group). IZ, infarct zone; RZ, remote zone. Scale bars, 500 m (left panels) and 50 m (middle and right panels). (B) Representative electron micrographs of peri-infarct zone myocardium 1 week after MI depicting two subpopulations of mitochondria: interfibrillar (IF) and perinuclear (PN) mitochondria. N, nucleus (n = 5 mice per group). Scale bars, 1 m. (C) Quantification of mitochondrial area and circularity (n > 350 or 125 perinuclear or interfibrillar mitochondria from 5 mice per group). (D) Guanosine 5′-triphosphate (GTP)–binding activity and protein abundance of Drp1 in the peri-infarct zone myocar-dium at 1 week after MI (n = 5 mice per group). GAPDH, glyceraldehyde-3-phosphate dehydrogenase. (E) GTP-binding activity of Drp1 in NRCMs under normoxia (N) or hypoxia (H) for 16 hours (n = 3 independent experiments). (F) Hypoxia-induced change in mitochondrial morphology. Left: Representative images of mitochondrial morphologies of NRCMs loaded with MitoTracker Green. Right: Mitochondrial morphologies of NRCMs classified into three groups: vesicle, intermediate, and tubule (n = 3 independent experiments). Scale bars, 10 m (white) and 5 m (yellow). (G) Effects of Drp1 knockdown on hypoxia/reoxygenation (H/R)–induced cardiomyocyte senes-cence. NRCMs transfected with negative control small interfering RNA (siRNA) (siNC) or two different siRNAs for Drp1 (siDrp1 #1 and #2) were incubated for 16 hours of hypoxia, followed by 12 hours of reoxygenation (n = 3 independent experiments). Left: Representative images of SA--gal staining of NRCMs. The arrowheads show SA--gal–positive cells. Right: Quantification of SA--gal–positive cells. Scale bars, 50 m. Data are means ± SEM. *P < 0.05, **P < 0.01 by unpaired t test (D to F) or one-way analysis of variance (ANOVA) (G).

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

4 of 14

endothelial progenitor cells (40). We investigated whether the phosphoryl-ation state at Ser637 on Drp1 alters the interaction with FLNa. However, Drp1 phospho-deficient S637A and phospho- mimic S637D mutants associated with FLNa to the same extent (Fig. 2C). Green fluorescent protein (GFP)–Drp1 was mainly localized in the cytoplasm, whereas mCherry-FLNa showed punc-tate structures in normoxic H9c2 cardiac myoblasts. After hypoxia, GFP-Drp1 formed punctate structures and colo-calized with mCherry-FLNa (Fig. 2D). To investigate whether Drp1 and FLNa interaction occurred in mitochondria, we performed subcellular fractionation. The Drp1-FLNa interaction was mainly observed in the mitochondrial fraction (Fig. 2E). Moreover, FLNa was diffusely detected in cardiomyocytes of the hearts of sham-operated mice, whereas Drp1 was detected as punctate dots. In the peri-infarct zone myocardium 4 weeks after MI, the signals for FLNa and Drp1 were increased and were colocalized (Fig. 2F). These results suggest that FLNa binds to Drp1 under hypoxic conditions.

The GTPase domain of Drp1 interacts with the rod-2 segment of FLNaNext, we identified the protein domains re-quired for the interaction of Drp1 and FLNa. Drp1 contains a GTPase, middle domain (MID), variable (Var), and GTPase effector domain (GED) domain (41). The GTPase domain of Drp1 was sufficient for the inter-action with FLNa (Fig. 3A). FLNa consists of an N-terminal actin- binding domain and 24 immunoglobulin (Ig) repeats. The rod-1 segment (Ig1-15) is the secondary actin-binding region. Mutants lacking the actin-binding regions (Ig1-24 and Ig16-24) are still associated with Drp1 (Fig. 3B). The Ig24 dimerization domain did not asso-ciate with Drp1 (fig. S7), indicating that the rod-2 segment (Ig16-23) of FLNa is sufficient for the interaction with Drp1.

FLNa is indispensable for hypoxia-induced mitochondrial fissionActin and myosin filaments recruit Drp1 to mitochondrial fission sites and induce ring-like oligomeric assembly of Drp1 (30–33), but the mechanism for how Drp1 accumulates in these cytoskeletal filaments remains obscure. Consistent with previous reports, disruption of the

FLNa

Drp1

Actin

Keratin

Hypox

ia

197

112

84

60

47

(kDa) Normox

iaA B

D

Hyp

oxia

Nor

mox

ia

GFP-Drp1mCherry-FLNa

F

Hypoxia + − +FLAG-Drp1 − + +

FLNa

FLAG

FLNa

FLAGIP

: FLA

GLy

sate

Sham LV

Actin

Actin

Drp1

FLNa

Drp1

FLNa

Myc

FLAG

Myc

FLAG

VDAC1

α-Tubulin

Myc-FLNaFLAG-Drp1

+−

+−

++

++

Mito CytoFraction

IP: F

LAG

Lysa

te

E

Drp1 FLNa

MI peri-IZ

Drp1 FLNa 2nd Ab only

FLNa

Drp1

Sham MI peri-IZ

FLNa

Drp1

C

FLAG-Drp1 −

Myc

FLAG

Myc

FLAG

IP: F

LAG

Lysa

te

SD SAMyc-FLNa

−− + + +

Fig. 2. FLNa is a hypoxia-dependent binding partner of Drp1 in peri-infarct zone myocardium after MI. (A) Coomassie brilliant blue–stained images of SDS–polyacrylamide gel electrophoresis (PAGE) gels to identify proteins interacting with FLAG-Drp1. HeLa cells expressing FLAG-Drp1 were exposed to normoxia or hypoxia. (B) Hypoxia- dependent interaction of FLAG-Drp1 with endogenous FLNa in HeLa cells. (n = 3 independent experiments). (C) Inter-action of Myc-FLNa with FLAG-Drp1 S637D (SD) and S637A (SA) mutants in HeLa cells under hypoxia (n = 3 independent experiments). (D) Colocalization of GFP-Drp1 with mCherry-FLNa in H9c2 cells under normoxia or hypoxia (n = 3 inde-pendent experiments). Scale bars, 10 m. (E) Interaction of Myc-FLNa with FLAG-Drp1 in mitochondrial (Mito) or cyto-solic (Cyto) fractions of HeLa cells (n = 3 independent experiments). (F) Representative images and distribution of Drp1 (red) and FLNa (green) in mouse LV myocardium 4 weeks after MI. Lower panels display high magnification images. The boxed regions in peri-infarct zone myocardium of MI-treated heart or sham-treated hearts were magnified and color separated (n = 3 mice per group). Scale bars, 60 m (white) and 10 m (yellow).

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

5 of 14

actin cytoskeleton by latrunculin B (LatB; Fig. 4A) prevented hypoxia- induced mitochondrial fission in H9c2 myoblasts (Fig. 4B). To exam-ine whether FLNa was required for mitochondrial fission, we tested the effect of FLNa knockdown on hypoxia-induced mitochondrial fission, using two different siRNAs for FLNa (siFLNa #1 and #2; fig. S8A). Each siRNA for FLNa inhibited hypoxia-induced mitochon-drial fragmentation (Fig. 4C) in H9c2 cells. In addition, FLNa knockdown prevented hypoxia-induced mitochondrial fission in nonmuscle cardiac fibroblasts (fig. S8B). We evaluated the mito-chondrial function of NRCMs by analyzing mitochondrial membrane potential and lactate production. Knockdown of FLNa suppressed not only the hypoxia-induced reduction in mitochondrial mem-brane potential and lactate production (fig. S8, C and D) but also the hypoxia/reoxygenation-induced senescence of NRCMs (Fig. 4D). Drp1 is mainly localized in the cytosol but forms punctate foci at fission sites during mitochondrial fission (31). FLNa knockdown pre-vented the targeting of Drp1 punctate structures to mitochondria under hypoxia (Fig. 4E). These results suggest a critical role of FLNa in Drp1-mediated mitochondrial fission under hypoxia in both muscular and nonmuscular cells.

FLNa acts as a GEF for Drp1 in coordination with an actin cytoskeletonHypoxic stress increases FLNa abundance in neurons (42). The protein abundance of -actin and FLNa was increased in the peri-infarct zone myocardium after MI (Fig. 5A). To examine whether the increased abundance of these proteins affected Drp1 function, we analyzed the GTP-binding activity of Drp1 in human embryonic kidney–293 (HEK293) cells overexpressing FLNa or -actin. Overexpression of FLNa promoted the GTP-binding activity of Drp1 (Fig. 5B), whereas -actin overexpression did not affect Drp1 activation, although actin filaments reportedly promote Drp1 GTPase activity (31). GEFs increase the steady-state GTPase activity

of G protein by accelerating guanosine diphosphate (GDP)/GTP exchange. We evaluated the GEF activity of FLNa by measuring the steady-state GTP hydrol-ysis mediated by Drp1. The GTP hydrol-ysis of recombinant Drp1 was increased by the addition of EDTA, which enhances spontaneous GDP/GTP exchange, as ex-pected, and by the addition of FLNa pu-rified from mammalian cells (Fig. 5C). In general, GEFs preferentially bind to the GDP-bound form or the nucleotide- free form of G protein. FLNa is asso-ciated with the Drp1 K38A mutant, which is deficient in GTP binding (Fig. 5D) (43). These results suggest that FLNa acts as a GEF for Drp1. Drp1 was not activated by the overexpression of the actin-binding–deficient FLNa mutants (Ig1-24 and Ig16-24; Fig. 5E) that could still interact with Drp1 (Fig. 3B), indicat-ing that FLNa mediates actin-dependent Drp1 activation in cells. Consistent with its ability to increase the GTP-binding activity of Drp1, FLNa overexpression induced mitochondrial fission in H9c2

cells (Fig. 5F). Some FLNc protein variants, including A1539T, cause familial hypertrophic cardiomyopathy, and overexpression of FLNc A1539T induces the aggregation of actin filaments (44). Over-expression of the FLNa A1545T mutant, which corresponds to the FLNc A1539T mutant, caused more severe mitochondrial fragmen-tation than overexpression of wild-type FLNa (Fig. 5F). These re-sults suggest that hypoxia-induced ternary complex formation between Drp1, FLNa, and actin filaments underlies Drp1 activation, followed by mitochondrial fission.

Cilnidipine prevents hypoxia-induced mitochondrial fission by disrupting the Drp1-FLNa interactionDrp1-mediated mitochondrial fission is an attractive target for combating cardiovascular diseases. Mdivi-1 was originally identified as a pharmacological inhibitor of Drp1 and blocks Drp1-mediated mitochondrial fission (38). Although Mdivi-1 treatment improves cardiac function in several cardiac disease models (7–9, 45–47), the selectivity of Mdivi-1 for Drp1 has been challenged (48, 49). Alter-natively, drug repositioning is an attractive strategy for the applica-tion of known drugs to new treatment indications while reducing the risk of failure because of adverse toxicology. For these reasons, we screened compounds that inhibit hypoxia-induced mitochon-drial fission among antihypertensive drugs approved by the Japanese government. We used cardiac fibroblasts for drug screening because their mitochondrial morphology enables more sensitive evaluation of mitochondrial fission in response to hypoxic stress (compare fig. S9, A and B, to fig. S3, A and B). Moreover, FLNa is involved in hypoxia- induced mitochondrial fission in cardiac fibroblasts (fig. S8B) as it is in H9c2 cells (Fig. 4C). Among the antihypertensive drugs we tested, we found that cilnidipine, an antihypertensive drug with a DHP-based structure that targets voltage-dependent L/N-type Ca2+ channels, specifically suppressed hypoxia-induced mitochondrial frag-mentation (Fig. 6A). Cilnidipine also suppressed hypoxia-mediated

A

GTPase MID Var GEDDrp1

FLAG- GTPase

–FLNa

FLAG

B

FLNaABD 1 2415

Ig1-24

Ig16-24

16

Rod-1 Rod-2

FLAG-Drp1 − + − +

Myc

− +

Myc

FLAG

FLAG

IP: F

LAG

Lysa

te

FLNa

Ig1-24

Ig16-2

4

Myc-

GTPase

FLNa

FLAG

IP: F

LAG

Lysa

te

Drp1

GTPase

Drp1

GTPase

FLNaIg1-24Ig16-24

FLNaIg1-24

Ig16-24

Drp1

Fig. 3. The interacting regions of Drp1 and FLNa. (A) Interaction of truncated FLAG-Drp1 (GTPase) with endogenous FLNa under hypoxia (n = 3 independent experiments). (B) Interaction of FLAG-Drp1 with actin-binding–deficient Myc-FLNa truncated mutants (Ig1-24 or Ig16-24) under hypoxia (n = 3 independent experiments).

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

6 of 14

mitochondrial fission in NRCMs (Fig. 6B). Other drugs, including DHP-derived L-type Ca2+ channel blockers and other antihypertensive drugs, did not affect hypoxia-induced mitochondrial hyper-fission, suggesting that pharmacological inhibition of L/N-type Ca2+ channels did not contribute to mitochondrial fission. Cilnidipine also suppressed hypoxia/reoxygenation-induced cardiomyocyte senescence and ROS production (Fig. 6C and figs. S4C and S5C), whereas amlodip-ine, an L-type Ca2+ channel blocker, did not affect hypoxia/reoxygenation-induced myocardial senescence (Fig. 6C).

Next, we examined the effect of cil-nidipine on hypoxia-induced interaction between Drp1 and FLNa. The hypoxia- induced formation of Drp1-FLNa com-plexes was suppressed by cilnidipine, but not by Mdivi-1 in HeLa cells (Fig. 6, D and E). HeLa cells do not express voltage- dependent Ca2+ channels, suggest-ing that this cilnidipine effect is indepen-dent of blockage of voltage-dependent L/N-type Ca2+ channels. These results in-dicate that cilnidipine prevents hypoxia- induced mitochondrial fragmentation by disrupting Drp1-FLNa complex formation.

Cilnidipine improves cardiac function after MI by inhibiting Drp1-mediated myocardial senescenceCa2+ channel blockers, especially cardiac- specific non-DHP Ca2+ channel blockers, generally exacerbate cardiac dysfunction in patients with chronic heart failure (50). We investigated whether inhibition of Drp1 by cilnidipine had beneficial effects on heart failure after MI in mice. Because pharmacological pretreatment or early posttreatment (3 hours to 3 days) with amlodipine reportedly improves cardiac function after MI in rodents (51–54), we compared the cardioprotective effects of cilnidipine and amlodipine starting 7 days after infarction. Continuous treatment of mice with cilnidipine (20 mg/kg per day) from 7 days after MI improved cardiac function compared with vehicle- treated mice at 4 weeks after MI (Fig. 7A). Because the same dose of amlodipine treatment increased mortality (fig. S10A), we analyzed the effect of a low daily dose of amlodipine (2.5 mg/kg per day). Post-treatment of mice with amlodipine did not have a cardioprotective effect (Fig. 7A),

siNC siFLNa #1 siFLNa #2

Hyp

oxia

Nor

mox

ia

C

E

GFP

-Drp

1M

itoch

ondr

iaE

nlar

ge

siNC normoxia siNC hypoxia siFLNa #1 normoxia siFLNa #1 hypoxia

siFLNa #2siFLNa #1siNC

100

80

60

40

20

0

% o

f tot

al c

ells

N H N H N HVesicle Intermediate Tubule

∗∗∗

B DMSO LatB

Hyp

oxia

Nor

mox

ia

100

80

60

40

20

0

% o

f tot

al c

ells

N H N H N HVesicle Intermediate Tubule

LatBDMSO

∗∗ ∗∗

∗∗ ∗∗

∗∗∗∗∗∗

siNC siFLNa #1 siFLNa #2

H/R

N

D

α-actinin p53 DAPI

40

30

20

10

0% o

f p53

+ α-a

ctin

in+ c

ells

N H/R N H/R N H/RsiNC siFLNa

#1siFLNa

#2

∗∗ ∗∗∗∗

AD

MS

OLa

tB

Fig. 4. FLNa is required for hypoxia-mediated Drp1 activation and mitochondrial fission–associated myocardial senescence. (A) Morphological changes in actin cytoskeleton of H9c2 cells in response to the actin-depolymerizing drug LatB (20 nM, 18 hours). Actin cytoskeleton was labeled with Alexa 594–phalloidin (n = 3 independent experi-ments). (B) Effects of LatB on hypoxia-induced mitochondrial fission. H9c2 cells were treated with 20 nM LatB for 1 hour before being exposed to hypoxia (n = 3 independent experiments). Scale bars, 50 m. DMSO, dimethyl sulfoxide. (C) Effect of FLNa knockdown on hypoxia-induced mitochondrial fission. H9c2 cells were treated with negative control siRNA (siNC) or two different siRNAs for FLNa (siFLNa #1 and #2) (n = 3 independent experiments). Scale bars, 50 m. (D) Effect of FLNa knockdown on hypoxia/reoxygenation-induced accumulation of p53 in NRCMs. The population of senescent cells near p53 punctae in -actinin–positive NRCMs was quantified (n = 3 independent experiments). Scale bars, 20 m. (E) Effects of FLNa siRNA on the localization of GFP-Drp1 and mitochondria in cardiac fibroblasts under normoxia or hypoxia. Mitochondria were stained with MitoTracker Red (n = 3 independent experiments). Scale bars, 10 m. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01 by one-way ANOVA.

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

7 of 14

suggesting that cilnidipine exerts cardioprotection through a different molecular mechanism than amlodipine. Moreover, echocardiographic and Millar catheter analyses showed that cilnidipine treatment from 1 day after MI significantly protected LV function compared with vehicle-treated mice at 4 weeks after MI (tables S1 and S2). We used heart samples from mice treated with cilnidipine 1 day after infarction for further analyses. Cilnidipine treatment signifi-cantly reduced LV hypertrophy compared with vehicle-treated mice at 4 weeks after MI (Fig. 7, B and C). The mortality and LV hyper-trophy after MI in N-type Ca2+ channel Cav2.2-deficient (Cav2.2−/−) mice were similar to those in wild-type (Cav2.2+/+) mice (Fig. 7D and fig. S10B). Cilnidipine treatment significantly improved LV hyper-trophy and function after MI in both Cav2.2+/+ and Cav2.2−/− mice (Fig. 7D and table S3). These results suggest that inhibition of L/N-type Ca2+ channel activity does not participate in the ability of cil-nidipine to prevent heart failure. Furthermore, cilnidipine significantly improved mitochondrial morphology (Fig. 7E) and decreased Drp1 activity (Fig. 7F) and SA--gal–positive areas (Fig. 7G) in the peri- infarct zone in the LV myocardium, as well as LV remodeling, in-

cluding the increase in interstitial fibrosis (Fig. 7H) and expression of hypertrophy- and fibrosis-related genes (Fig. 7I). Cilnidipine failed to suppress HO-1 protein induction in the peri-infarct zone myocar-dium after MI (Fig. 7J), indicating that cilnidipine may not reduce hypoxic stress. Collectively, these results suggest that cilnidipine im-proves cardiac function after MI by suppressing Drp1-mediated myocardial cell senescence, although Ca2+ channel blockers are be-lieved to be contraindicated for treatment of heart failure.

DISCUSSIONStructural connections between mitochondria and the cytoskeleton play key roles in various mitochondrial functions, including trans-port and fission. Microtubules are important for mitochondrial mo-tility and transport (55). The GTPase mitochondrial Rho (Miro) and the kinesin adaptor Milton complex drives kinesin-dependent mito-chondrial transport among microtubules (55). The Miro-Milton complex regulates mitochondrial morphology in a Ca2+-dependent manner (55). In the case of mitochondrial fission, ER tubules wrap

A

FLNa

GAPDH

2.01.5

Act

in/G

AP

DH

(fo

ld)

0.50

1.0

MI − +

MI − +

Drp1

Actin

43

FLN

a/G

AP

DH

(fo

ld)

10

∗∗

2

MI − +

E

GTP-Drp1

input-Drp1

Myc

Ig16-24Ig1-24FLNaMockMyc-

GTP

-Drp

1/in

put-D

rp1

(fold

)

3

1

0

2

Ig16-24Ig1-24FLNaMockMyc-

∗∗∗∗∗∗

B

GTP-Drp1

input-Drp1

MockMyc-FLNa

mCherry-β-actin

Myc-FLNamCherry-β-actin

−−

+−

−+

GTP

-Drp

1/in

put-D

rp1

(fold

)

43

10

2

∗∗ ∗∗

Myc

mCherry

FLNa WTmCherry

FLNa A1545T

FLNa A1545TFLNa WTmCherry

F

100

80

60

40

20

0

% o

f tot

al c

ells

Vesicle Inter-mediate

Tubule

∗∗

C

0.8

0.6

0.4

0.2

0

Drp1Drp1 + FLNa

Pi r

elea

sed

(nm

ol)

0 20 40 60Time (min)

1.5

1.0

0.5

0

Drp1

Drp1 +

FLNa

GTP

hyd

roly

sis

rate

(pm

ol/m

in/p

mol

Drp

1)

∗∗Drp1 + EDTA1.0

Drp1 +

EDTA

WT KA–Myc

FLAG-Drp1Myc-FLNa

FLAG

Myc

FLAG

IP:

FLA

GLy

sate

D

Fig. 5. FLNa drives Drp1 activation and mitochondrial fission in coordination with actin filaments. (A) Protein abundances of actin and FLNa in peri-infarct zone myocardium at 4 weeks after MI (n = 3 mice per treatment). (B) Effect of FLNa or -actin overexpression on Drp1 activation in HEK293 cells (n = 3 biological replicates). (C) The time course of GTP hydrolysis by Drp1 (600 nM) in the absence or presence of FLNa (200 nM) or EDTA. Right: Calculated GTP hydrolysis rate of Drp1 (n = 3 independent experiments). (D) Interaction of Myc-FLNa with FLAG-Drp1 WT or K38A (KA) under hypoxia (n = 3 independent experiments). (E) Effect of actin-unbound FLNa mutant on Drp1 activation in HEK293 cells (n = 3 biological replicates). (F) Effect of FLNa overexpression on mitochondrial fission. H9c2 cells expressing mCherry-FLNa wild-type (WT) or A1545T were incubated under normoxia and stained with MitoTracker Green (n = 3 independent experiments). Scale bars, 50 m. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01 by unpaired t test (A) or one-way ANOVA (B, C, E, and F).

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

8 of 14

around mitochondria to facilitate constriction. This ER-mitochondria communication through contact sites known as mitochondria- associated ER membranes (MAMs) is important for mitochondrial fission. Actin filament formation at this region through ER-localized inverted formin 2 (INF2) and mitochondrial Spire1C is important for the accumulation and oligomeric ring formation of Drp1 (31, 56). Myosin II is also involved in this process (33). In this study, we iden-tified FLNa as a binding partner of Drp1. FLNa is a scaffold for more than 90 binding proteins involved in cell adhesion and migration, includ-ing channels, receptors, intracellular signaling molecules, and transcrip-tional factors (39). Our results showed that FLNa acted as a protein scaffold to functionally couple Drp1 with actin at fission sites and activated Drp1 through GEF activity under hypoxic conditions. How-ever, it is still unclear how Drp1-FLNa interaction is regulated under hypoxic conditions. The mitochondrial outer membrane protein FUN14

domain-containing 1 (FUNDC1) accumulates at MAMs and recruits Drp1 to MAMs and mitochondrial fission in response to hypoxia (57). Other binding proteins such as FUNDC1 or posttranslational modi-fications of Drp1 may regulate its interaction with FLNa.

Humans have three filamin genes, which encode FLNa, FLNb, and FLNc proteins. These proteins share the same domain structures and have about 70% sequence similarity overall. FLNa is ubiquitously distributed, and mutations in FLNa cause an X-linked form of famil-ial cardiac valvular dystrophy (58, 59). FLNa gene deletion in mice leads to a severe defect in cardiac morphogenesis (60), suggesting a critical role of FLNa in cardiac systems. FLNc is expressed in both cardiac and striated muscle. Various mutations in the FLNc gene have been identified in patients with familial hypertrophic cardiomyopathy, and the FLNc mutants affect actin and FLNc organization (44). Future clinical studies are necessary to determine whether FLNc participates

BDMSOCIL

0

20

40

60

80

100

% o

f tot

al c

ells

Vesicle TubuleInter-mediate

N H N H N H

∗∗ ∗ ∗

N

H/R

DMSO

− −+ +H/R

C

0

10

20

30

SA

- β-g

al–p

ositi

ve c

ells

(%)CIL 40

DMSO CIL

A

% o

f ves

icle

cel

ls

60

40

20

0

HN

DM

SO

DM

SO

CIL

Am

lodi

pine

Nife

dipi

neN

icar

dipi

neN

isol

dipi

neN

itren

dipi

neV

erap

amil

Dilt

iaze

mS

ilden

afil

Losa

rtan

Ate

nolo

lP

ropr

anol

olM

etop

rolo

l

∗∗ ∗∗

Cilnidipine (CIL)

N

H

DMSO CIL

HypoxiaCIL

−− −

+ ++

0

1

2

3

Drp

1-bi

ndin

g FL

Na

/tota

l FLN

a

FLAG-Drp1

FLNa

HypoxiaCIL

−− −

+ ++

FLAG-Drp1

−+

+ + + −∗ ∗ Hypoxia

Mdivi-1+− −

− +−

FLAG-Drp1

++

− + + +

FLNa

FLAG-Drp1

FLNa

FLAG-Drp1

IP: F

LAG

Lysa

te

HypoxiaMdivi-1

−− −

+ ++

0

1

23

4

Drp

1-bi

ndin

g FL

Na

/tota

l FLN

a ∗∗

D E

FLAG-Drp1

FLNa

IP: F

LAG

Lysa

te

∗∗

− +AML

∗∗ ∗∗AML

Fig. 6. Identification of cilnidipine as an inhibitor of hypoxia-induced mitochondrial fission by disrupting Drp1-FLNa complex. (A) Effects of antihypertensive drugs on mitochondrial fission. Cardiac fibroblasts were treated with each drug (1 M) for 1 hour before being exposed to hypoxia. The percentages of cells with vesicle-type mitochondrial fragmentation were quantified (n = 3 to 6 independent experiments). Cilnidipine, CIL. (B) Effects of cilnidipine on hypoxia-mediated mitochondrial frag-mentation. Left: Representative images of mitochondrial morphologies of NRCMs loaded with MitoTracker Green. Right: Mitochondrial morphologies of NRCMs treated with cilnidipine (1 M) for 1 hour before 16 hours of normoxia (N) or hypoxia (H) (n = 3 independent experiments). Scale bars, 10 m. (C) Effects of cilnidipine on hypoxia/reoxygenation-induced cardiomyocyte senescence. NRCMs were treated with 1 M cilnidipine or amlodipine (AML) for 1 hour before 16 hours of hypoxia, followed by 12 hours of reoxygenation (n = 5 independent experiments). Scale bars, 20 m. (D and E) Interaction of FLAG-Drp1 with endogenous FLNa under hypoxia and effects of cilnidipine (D) or Mdivi-1 (E) treatment. HeLa cells expressing FLAG-Drp1 were treated with hypoxia with or without cilnidipine (3 M) or Mdivi-1 (10 M) [n = 5 (D) and n = 3 (E) independent experiments]. Data are shown as the means ± SEM. *P < 0.05, **P < 0.01 by one-way ANOVA.

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

9 of 14

CIL − −+ +MI − − + +

GTP

-Drp

1/in

put D

rp1

(fold

)

CIL − −+ +MI − − + +

012345

G

GTP-Drp1

Input-Drp1

0

2

4

6

8

HW

/BW

(mg/

g)

CIL − 30100 − 30100MISham

B

∗∗ ∗

∗∗∗∗

∗∗10

0

5

10

15

20

Sha

mM

I

Vehicle CIL

CIL − −+ +MI − − + +

SA

-β-g

al–p

ositi

ve a

rea

(%)

CIL − 30 100

0

300

CIL − 30100 − 30100MISham

∗∗∗ ∗∗C

200

100CS

A (µ

m2 )

D

0

5

10

15

20E

Cav2.2

+/+

HW

/BW

(mg/

g)

CILMI

−−

+−

−+

++

−−

+−

−+

++

Cav2.2

−/−

∗ ∗∗ ∗∗

0

10

20

30

40

50

ANP α-SKA

ACE Perio

stin

Colla

gen

1a1

TGF

-β1CTGF

##

Colla

gen

3a1

I

CIL − 30 100

H

0

3

6

9

12

CIL − 30100MISham

CV

F (%

)

Vehicle shamCIL shamVehicle MICIL MI

mR

NA

/GA

PD

H (f

old)

TGF

-β2TGF

-β3

60

# ##

#### ##

# ## ##

Sha

mM

IS

ham

MI

− 30 100

∗∗ ∗∗∗∗

††††

††

††††

††

††††

††

††

HO-1

Mn-SOD

CIL − −+ +MI − − + +

J

CIL − −+ +MI − − + +

0

5

10

15

20H

O-1

/Mn-

SO

D (f

old)

∗∗∗∗

∗∗ ∗∗

EF

(%)

0

20

40

60

80

100

CILVeh

AMLCIL

VehAML

MISham

∗∗∗ ∗

A

F∗

Sham-VehMI-VehSham-CILMI-CIL

Circ

ular

ity0

0.5

1.0

CILMI

−−

+−

−+

++

∗∗ ∗∗

Fig. 7. Improvement of cardiac function after MI through suppression of Drp1-mediated myocardial senescence by cilnidipine. (A) Effect of cilnidipine on ejection fraction (EF) in mice 4 weeks after MI. An osmotic pump filled with cilnidipine (20 mg/kg per day) or amlodipine (2.5 mg/kg per day) was implanted intraperitoneally 7 days after MI (n = 5 to 8 mice per treatment). (B) Effects of cilnidipine on the heart weight (HW)/body weight (BW) ratio in mice 4 weeks after MI. Cilnidipine (30 or 100 mg/kg per day) was infused at 1 day after MI (n = 4 to 10 mice per treatment). (C) Hematoxylin and eosin–stained images of hearts at 4 weeks after MI. Scale bars, 1.5 mm. Right: Cell surface area (CSA) of cardiomyocytes (n = 4 to 5 mice per treatment). (D) Effects of cilnidipine (30 mg/kg per day) on increased heart weight/body weight ratio in Cav2.2+/+ and Cav2.2−/− mice 4 weeks after MI (n = 3 to 6 mice per treatment). (E) Effects of cilnidipine (30 mg/kg per day) on mitochondrial morphology in peri-infarct zone myo-cardium 1 week after MI. Left: Representative images of interfibrillar mitochondria. Right: Quantification of mitochondrial circularity. n > 200 mitochondria from three mice per group. Scale bars, 1 m. (F) Effects of cilnidipine (30 mg/kg per day) on Drp1 activity in peri-infarct zone myocardium 1 week after MI (n = 3 mice per treatment). (G) Effect of cilnidipine (30 mg/kg per day) on the proportion of SA--gal–positive area in peri-infarct zone myocardium 4 weeks after MI (n = 3 to 4 mice per treatment). Scale bars, 100 m. (H) Picrosirius red–stained images of peri-infarct zone myocardium 4 week after MI. Scale bars, 100 m. Right: Collagen volume fractions (CVFs) (n = 4 mice per treatment). (I) mRNA amounts of hypertrophy-related (ANP and -SKA) and fibrosis-related (ACE, Periostin, Collagen 1a1, Collagen 3a1, TGF-1, TGF-2, TGF-3, and CTGF) genes in peri-infarct zone myocardium at 1 week after MI with or without cilnidipine (30 mg/kg per day) (n = 3 to 4 mice per treatment). (J) Effects of cilnidipine on protein abundances in peri-infarct zone myocardium at 1 week after MI (n = 3 mice per treatment). *P < 0.05, **P < 0.01 by one-way ANOVA (A to H and J); ††P < 0.01 compared to vehicle sham, #P < 0.05, ##P < 0.01 compared to vehicle MI by one-way ANOVA (I).

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

10 of 14

in Drp1-mediated mitochondrial fission and senescence and whether cilnidipine could be used to treat FLNc-related cardiomyopathy.

Chronic heart failure after MI is a major cause of mortality and morbidity worldwide. In this study, we demonstrated that Drp1- dependent mitochondrial fragmentation occurred in the peri-infarct zone in mouse heart after MI. Fragmented mitochondria in the myo-cardium are also observed in patients with dilated cardiomyopathy (61), and pharmacological inhibition of mitochondrial fission by Mdivi-1 attenuates heart failure in mice induced by pressure over-load (7, 45), ischemia/reperfusion (8, 46), and doxorubicin (9). These results suggest that abnormal mitochondrial fission in the myocar-dium leads to the progression of heart failure and that inhibition of mitochondrial fission is an attractive target for stress-induced car-diomyopathy. However, the mitochondrial fission/fusion cycle is physiologically critical to maintain mitochondrial quality control. Genetic ablation of Drp1 leads to mitochondrial enlargement and evokes cardiomyocyte necrosis and lethal dilated cardiomyopathy (5). Therefore, long-term inhibition of mitochondrial fission, which would block physiological fission for quality control, might not be an appropriate therapeutic strategy.

In the present study, cilnidipine treatment attenuated the progres-sion of heart failure after MI. Cilnidipine suppressed hypoxia- mediated mitochondrial fragmentation and subsequent senescence by inhibiting Drp1-FLNa complex formation. FLNa colocalized with Drp1 in peri-infarct zone myocardium of MI-treated heart, whereas the interaction of FLNa and Drp1 did not appear to occur under normoxia (Fig. 2). Therefore, cilnidipine would effectively prevent hypoxia-mediated mitochondrial fragmentation without disrupting the fission/fusion balance under normoxia.

An abnormal balance between mitochondrial fission and fusion has been linked to various diseases, including not only cardiac diseases but also neurologic diseases, cancer, and diabetes (62). Our results suggest that hypoxia induced the functional coupling of Drp1 and FLNa to enhance mitochondrial fission, and cilnidipine selectively inhibited the hypoxia-induced Drp1-FLNa interaction. Because FLNa is ubiquitously distributed and also correlated with cancer and Alzheimer’s disease (63, 64), elucidation of the universality of the Drp1-FLNa interaction for mitochondrial quality control may contribute to expanded approval of cilnidipine for other mitochondria- related diseases.

MATERIALS AND METHODSReagents and antibodiesAmlodipine was from Tokyo Chemical Industry Co. Ltd. Mdivi-1, sildenafil, atenolol, propranolol, metoprolol, thiocarbohydrazide solu-tion, durcupan resin, c-Myc peptide, and GTP-agarose were from Sigma-Aldrich. Dulbecco’s modified Eagle’s medium (DMEM), dilti-azem, nifedipine, nisoldipine, nicardipine, nitrendipine, verapamil, losartan, uranium acetate, anti-Myc, and anti–-actin were from Wako. MitoTracker Green FM, MitoTracker Red CM-H2XRos, and MitoSox Red were from Invitrogen. Collagenase II was from Worthington. Osmium tetroxide was from Electron Microscopy Science. Bovine serum albumin (BSA), sodium cacodylate, and phenol red–free DMEM were from Nacalai Tesque. Anti-Drp1 (H-300), anti-GAPDH (FL-335), anti-SOD2 (Mn-SOD: N-20), anti-Mfn1 (H-65), and anti-filamin 1 (FLNa: H-300 and E-3) were from Santa Cruz Biotechnology. Anti- p53 (7F5) and horseradish peroxidase (HRP)–conjugated anti-mouse and rabbit IgG were from Cell Signaling Technology. Anti-tafazzin,

anti–HIF-1, anti-sarcomeric -actinin (EA-53), and anti-Mfn2 were from Abcam. Anti-Opa1 was from BD Biosciences. Anti–HO-1 was from Enzo Life Sciences. EGTA and Hepes were from Dojindo.

AnimalsAll experiments using mice and rats were approved by an Ethics Committee of National Institutes of Natural Sciences and carried out in accordance with their guidelines. C57BL/6J mice and Sprague- Dawley (SD) rats were purchased from Japan SLC Inc. The Cav2.2−/− mice and their wild-type littermates (Cav2.2+/+) were generated as previously described (65). All mice were maintained in a specific pathogen–free area on a 12-hour light/dark cycle.

Induction of MISurgery to induce MI was performed on 6-week-old male C57BL/6J, Cav2.2+/+, and Cav2.2−/− mice as described (34). All surgical proce-dures were performed in mice anesthetized with a mixture of domitor (0.75 mg/kg; Nihon Zenyaku Kogyo), midazolam (4 mg/kg; Sandoz), and butorphanol (5 mg/kg; Meiji Seika Pharma). A mini-osmotic pump (ALZET) filled with vehicle (saline) or cilnidipine (20, 30, or 100 mg/kg per day) or amlodipine (2.5 or 20 mg/kg per day) was implanted intraperitoneally into 6-week-old male C57BL/6J mice at 1 or 7 days after MI.

Transthoracic echocardiography and cardiac catheterizationEchocardiography and cardiac catheterization were performed in mice anesthetized with a mixture of domitor, midazolam, and butorphanol. Echocardiography was performed by Nemio-XG echocardiography (Toshiba) with a 14-MHz transducer at 4 weeks after MI. Cardiac functions were measured using a micromanometer catheter (Millar 1.4F, SPR 671, Millar Instruments) at 4 weeks after MI.

Morphological analysisMouse hearts were removed, washed in phosphate-buffered saline (PBS), and fixed with 10% neutral-buffered formalin (Nacalai Tesque). For quantitative assessment of collagen I and III deposition, hearts were embedded in paraffin, sectioned at a thickness of 3 m, and stained with picrosirius red using 0.1% Direct red 80 (Polysciences). For assessment of cardiomyocyte cross-sectional areas, specimens were stained with hematoxylin and eosin (Sigma). Three regions were randomly selected for each specimen in the LV area, and the average values were calculated using BZ-II Analyzer software (Keyence).

ImmunohistochemistryMouse heart tissues were embedded in optimal cutting temperature (O.C.T.) compound (Sakura Finetek) and snap-frozen with liquid nitrogen. Samples were sectioned at a thickness of 20 m. Glass- mounted fresh frozen sections were fixed in 4% paraformaldehyde in PBS at room temperature for 10 min. After rinsing with PBS, sections were blocked in PBS with 1% BSA, 0.3% Triton X-100 at room temperature for 30 min and then incubated overnight with primary antibodies directed against FLNa and Drp1 at 4°C, followed by incubation with secondary fluorescent anti-mouse and anti-rabbit antibodies. Sections were incubated with the Vector TrueVIEW Autofluorescence Quenching Kit (Vector Laboratories Inc.) for 1 min, before being mounted with VECTASHIELD HardSet Antifade Mount-ing Medium (Vector Laboratories Inc.). Images were obtained using an A1 confocal microscope (Nikon).

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

11 of 14

Transmission electron microscopyMouse LV tissues were prefixed with 2% paraformaldehyde solution containing 0.15 M sodium cacodylate and 2 mM CaCl2 (pH 7.4) for 3 hours on ice and cut into 1- to 2-mm cubes. After washing with 0.15 M cacodylate solution, the tissue blocks were immersed in a solution containing 2% osmium tetroxide, 1.5% potassium ferro-cyanide, 0.15 M sodium cacodylate, and 2 mM CaCl2 (pH 7.4) for 1.5 hours at room temperature. After washing with distilled water, the tissue blocks were immersed in thiocarbohydrazide (0.01 mg/ml) solution for 40 min and post-fixed with 2% osmium for 1 hour. En bloc staining was performed by sequentially immersing the tissue blocks in a solution of 1% uranium acetate overnight at 4°C and an aqueous solution of lead aspartic acid for 60 min with oven-drying. After dehydration in a graded ethanol series and acetone, the speci-mens were embedded in durcupan resin. The surface (thickness of 70 nm) of the resin-embedded tissue was exposed using a diamond knife on an Ultracut UC7 (Leica Microsystems) and imaged with a Veleta CCD camera (Olympus) equipped on a JEOL1010 microscope (JEOL). We defined “perinuclear mitochondria” as mitochondria around nuclear and “interfibrillar mitochondria” as a single row of mitochondria between the myofibrils. Mitochondria for each animal were traced, and quantification of area of perinuclear mitochondria and circularity of interfibrillar mitochondria was performed by ImageJ software (National Institutes of Health).

Cell culture and transfectionNRCMs and fibroblasts were prepared from the ventricles of 1- to 2-day-old SD rats as described (34). H9c2 cardiac myoblasts were purchased from the American Type Culture Collection and cultured in DMEM supplemented with 10% fetal bovine serum (FBS). Plasmid DNA was transfected using Viafect (Promega) for HeLa cells or a Neon transfection system (Invitrogen) for cardiomyocytes, fibro-blasts, and H9c2 cells. For siRNA knockdown, cells were trans-fected with each siRNA (20 nM) using Lipofectamine RNAiMAX reagent (Invitrogen) for 72 hours. Stealth siRNAs for rat FLNa (#1, RSS308835; #2, RSS308836) and rat Drp1 (#1, RSS300120; #2, RSS300122) were from Invitrogen.

Western blottingMouse hearts were homogenized using a Physcotron (Microtec) in lysis buffer [20 mM Hepes (pH 7.4), 100 mM NaCl, 3 mM MgCl2, 1% NP- 40, 50 mM NaF, 1 mM Na3VO4, and 20 mM -glycerophosphate] containing a protease inhibitor cocktail at 4°C. Cleared lysates were mixed with sample buffer. Proteins separated by SDS-PAGE were electrotransferred onto polyvinylidene difluoride membranes (Millipore) at 2 mA/cm2 for 1 hour. After blocking with 1% BSA, the membrane-bound proteins were incubated with primary antibodies, followed by HRP-conjugated secondary antibodies. Antibody- bound proteins were detected using the Western Lightning Plus-ECL Kit (PerkinElmer).

GTP-agarose pulldown assayGTP-agarose pulldown assays were performed as described (24). Tissue samples were homogenized using the Physcotron in ice-cold GTP-binding buffer [50 mM Hepes (pH 7.4), 1% Triton X-100, 10% glycerol, 150 mM NaCl, 1.5 mM MgCl2, 1 mM EGTA, and 1% protease inhibitor cocktail]. NRCMs were washed with ice-cold PBS, collected in GTP-binding buffer, and homogenized by sonica-tion (Qsonica, Waken B Tech) for 3 s. The lysate was centrifuged

(16,000g for 15 min at 4°C), and an aliquot of the supernatant (100 g protein) was incubated with 20 l of GTP-agarose beads (equilibrated in GTP-binding buffer) for 45 min at room tempera-ture. The beads were centrifuged (1000g for 1 min at 4°C) and washed twice with GTP-binding buffer. The GTP-bound proteins were eluted with sample buffer containing dithiothreitol (DTT) and subjected to SDS-PAGE.

Measurement of mitochondrial functionsMitochondrial membrane potential was analyzed with 5′,6,6′- tetrachloro- 1,1′,3,3′-tetraethylbenzimidazolylcarbocyanine iodide (JC-1, Invitrogen). NRCMs were incubated with JC-1 (0.5 M) for 20 min. After changing the medium to phenol red–free DMEM, digital images were taken at 40× magnification using a fluorescence microscope (BZ-X700, Keyence), with five regions randomly selected for each sample. The mitochondrial membrane potential was repre-sented as the ratio of green-to-red fluorescence. Mitochondrial energy state was evaluated by lactate production. Lactate concen-tration in culture medium was measured by Determinar LA (Kyowa Medic).

Mitochondrial morphologyMitochondrial morphology was detected with MitoTracker Green FM and fluorescence microscopy using the BZ-X700 microscope with a sectioning function. Cells were serum-starved for 48 hours and incubated under normoxic (21% O2) or hypoxic (1% O2) con-ditions for 16 hours in a multigas incubator (Panasonic). For LatB treatment, cells were treated with 20 nM LatB 1 hour before hypoxia. The cells were then loaded with MitoTracker Green (50 nM) for 20 min. For colocalization of Drp1 and mitochondria, fibroblasts expressing GFP-Drp1 were incubated with MitoTracker Red CM-H2XRos (50 nM) for 20 min. After incubation, the cells were washed with PBS, changed to phenol red–free DMEM, and imaged. To analyze actin structure, cells were incubated with 20 nM LatB for 18 hours, and actin cytoskeleton was stained with Alexa 594–phalloidin. Ten images were randomly taken for each sample. The average mitochondrial fragment length per cell was quantified using ImageJ, and the mitochondrial morphology was classified into three groups (vesicle, intermediate, and tubule) according to the average mitochondrial fragment length.

Mitochondrial ROS productionMitochondrial ROS were examined using MitoSOX Red. Cells were incubated with MitoSOX (5 M) for 10 min at 37°C, washed with PBS, changed to phenol red–free DMEM, and imaged by confocal microscopy (FV10i, Olympus).

Myocardial senescenceMyocardial senescence was evaluated by senescence-associated -gal activity or cytosolic p53 accumulation. SA--gal activity was deter-mined using the Senescence -Galactosidase Staining Kit (Cell Sig-naling Technology) according to the manufacturer’s instructions. NRCMs were pretreated with drugs for 1 hour, subjected to 16 hours of hypoxia followed by 12 hours of reoxygenation, fixed with fixa-tive solution for 15 min at room temperature, and incubated over-night with -gal staining solution at 37°C. For mouse hearts, -gal staining solution was used at a final pH of 5.0. For immunofluores-cence to detect p53, cells were fixed with 4% paraformaldehyde for 10 min and incubated with blocking buffer (1% FBS in PBS containing

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

12 of 14

0.05% Triton X-100) for 1 hour. Cells were incubated with anti-p53 and anti–-actinin antibodies and then with fluorescence-conjugated secondary antibodies. Samples were imaged with the BZ-X700 microscope. The population of senescent cells accompanying p53 punctae in -actinin–positive cardiomyocytes was analyzed.

ImmunoprecipitationHeLa cells were lysed in GTP-binding buffer, and the cell lysates were clarified by centrifugation (16,000g for 15 min). FLAG-tagged proteins were immunoprecipitated from the supernatants contain-ing 20-l bed volume of anti-FLAG M2 agarose beads (Sigma). The immune complexes were washed three times and eluted with FLAG peptide (300 g/ml). To identify Drp1-binding proteins, HeLa cells expressing FLAG-Drp1 were incubated under normoxia or hypoxia for 16 hours. After washing with PBS, the cells were incubated with 100 M dithiobis (succinimidyl propionate) cross-linker for 30 min at room temperature. The cross-linking reaction was quenched by addition of 50 mM tris (pH 7.5) in PBS. After two washes with PBS, the cells were lysed and immunoprecipitated. Proteins were sepa-rated by SDS-PAGE and analyzed by Coomassie brilliant blue stain-ing. Bands were identified after in-gel trypsin digestion and mass spectrometry. Mass spectrometric analysis was performed once.

FractionationHeLa cells expressing Myc-FLNa and FLAG-Drp1 were collected from five 10-cm dishes, and mitochondrial and cytosolic fractions were separated using a Mitochondria/Cytosol Fractionation kit (BioVision) according to the manufacturer’s instructions. Mito-chondrial pellets were resuspended into the GTP-binding buffer, and mitochondrial lysates were clarified by centrifugation (16,000g for 15 min). FLAG-Drp1 was immunoprecipitated from mitochon-drial and cytosolic supernatants.

Steady-state GTP hydrolysis activity of Drp1Recombinant His-tagged Drp1 was purified from Escherichia coli using Ni–nitrilotriacetic acid agarose and stocked in buffer [20 mM Hepes (pH 7.4), 100 mM NaCl, 3 mM MgCl2, 1 mM DTT, 10% glycerol, and 10 M GDP]. FLNa was purified from HEK293 cells expressing Myc-FLNa. Myc-FLNa was immunoprecipitated using anti– Myc-tag monoclonal antibody agarose (Medical & Biological Laboratories Co. Ltd.) and collected in elution buffer [20 mM Hepes (pH 7.4), 50 mM KCl, 2 mM MgCl2, 1 mM DTT, and c-Myc peptide (500 g/ml)]. Drp1 (0.6 M) was mixed with or without FLNa (0.2 M) in hydro-lysis buffer [20 mM Hepes (pH 7.4), 50 mM KCl, 10 mM MgCl2, and 1 mM DTT]. To promote spontaneous GDP/GTP exchange of Drp1, 1 mM EDTA and 2 mM MgCl2 were used instead of 10 mM MgCl2. GTP hydrolysis was determined by incubating Drp1 with 100 M GTP for 60 min at 37°C. At each time point, the reaction was stopped by adding 25 mM EDTA, and released free phosphate was measured using a Malachite Green Phosphate Assay kit (BioAssay Systems).

Real-time reverse transcription polymerase chain reactionTotal RNA was isolated from frozen mouse hearts using an RNeasy Fibrous Tissue Mini Kit (Qiagen) according to the manufacturer’s instructions. Complementary DNA was synthesized with Prime Script RT (Takara Bio). Real-time polymerase chain reaction (PCR) was performed using Power SYBR Green PCR Master Mix (Applied Biosystems) or the QuantiTect Probe RT-PCR Kit (Qiagen) and TaqMan probes with an ABI PRISM 7500 Real-Time PCR System

(Applied Biosystems) according to the manufacturer’s instructions. The primers are listed in table S4.

Statistical analysisThe results are presented as means ± SEM from at least three inde-pendent experiments. Statistical comparisons were carried out by two-tailed Student’s t test or one-way ANOVA, followed by a Tukey or Newman-Keuls comparison procedure. Values of P < 0.05 were considered to be statistically significant.

SUPPLEMENTARY MATERIALSwww.sciencesignaling.org/cgi/content/full/11/556/eaat5185/DC1Fig. S1. Mitochondrial fission and premature myocardial senescence in peri-infarct zone myocardium after MI.Fig. S2. Changes in protein abundances and activities in peri-infarct zone myocardium after MI.Fig. S3. Mitochondrial morphology in NRCMs.Fig. S4. Reoxygenation-induced mitochondrial ROS production plays an indispensable role in myocardial senescence.Fig. S5. Effects of gene silencing and pharmacological inhibition of Drp1 on mitochondrial morphology and ROS production.Fig. S6. Identification of FLNa by mass spectrometric analysis.Fig. S7. No interaction between Drp1 and the Ig24 dimerization domain of FLNa.Fig. S8. Effect of FLNa knockdown on hypoxia-induced mitochondrial fission and function.Fig. S9. Mitochondrial morphology in cardiac fibroblasts.Fig. S10. Mortality in mice after MI.Table S1. Echocardiographic parameters of mice with cilnidipine administration.Table S2. Cardiac parameters of mice with cilnidipine administration.Table S3. Echocardiographic parameters of Cav2.2(+/+) or Cav2.2(−/−) mice with cilnidipine administration.Table S4. List of Taqman probe and primer sets for real-time PCR.

REFERENCES AND NOTES 1. M. Song, G. W. Dorn II, Mitoconfusion: Noncanonical functioning of dynamism factors in

static mitochondria of the heart. Cell Metab. 21, 195–205 (2015). 2. G. W. Dorn II, Mitochondrial fission/fusion and cardiomyopathy. Curr. Opin. Genet. Dev.

38, 38–44 (2016). 3. S.-B. Ong, S. B. Kalkhoran, H. A. Cabrera-Fuentes, D. J. Hausenloy, Mitochondrial fusion

and fission proteins as novel therapeutic targets for treating cardiovascular disease. Eur. J. Pharmacol. 763 (Pt. A), 104–114 (2015).

4. T. Wai, J. García-Prieto, M. J. Baker, C. Merkwirth, P. Benit, P. Rustin, F. J. Rupérez, C. Barbas, B. Ibañez, T. Langer, Imbalanced OPA1 processing and mitochondrial fragmentation cause heart failure in mice. Science 350, aad0116 (2015).

5. M. Song, K. Mihara, Y. Chen, L. Scorrano, G. W. Dorn II, Mitochondrial fission and fusion factors reciprocally orchestrate mitophagic culling in mouse hearts and cultured fibroblasts. Cell Metab. 21, 273–286 (2015).

6. H. Chen, S. Ren, C. Clish, M. Jain, V. Mootha, J. M. McCaffery, D. C. Chan, Titration of mitochondrial fusion rescues Mff-deficient cardiomyopathy. J. Cell Biol. 211, 795–805 (2015).

7. Y.-W. Chang, Y.-T. Chang, Q. Wang, J. J.-C. Lin, Y.-J. Chen, C.-C. Chen, Quantitative phosphoproteomic study of pressure-overloaded mouse heart reveals dynamin-related protein 1 as a modulator of cardiac hypertrophy. Mol. Cell. Proteomics 12, 3094–3107 (2013).

8. W. W. Sharp, Dynamin-related protein 1 as a therapeutic target in cardiac arrest. J. Mol. Med. 93, 243–252 (2015).

9. M. Gharanei, A. Hussain, O. Janneh, H. Maddock, P. Mukhopadhyay, Attenuation of doxorubicin-induced cardiotoxicity by mdivi-1: A mitochondrial division/mitophagy inhibitor. PLOS ONE 8, e77713 (2013).

10. A. A. Rosdah, J. K. Holien, L. M. D. Delbridge, G. J. Dusting, S. Y. Lim, Mitochondrial fission—A drug target for cytoprotection or cytodestruction? Pharmacol. Res. Perspect. 4, e00235 (2016).

11. J. M. van Deursen, The role of senescent cells in ageing. Nature 509, 439–446 (2014). 12. Y. Maejima, S. Adachi, H. Ito, K. Hirao, M. Isobe, Induction of premature senescence in

cardiomyocytes by doxorubicin as a novel mechanism of myocardial damage. Aging Cell 7, 125–136 (2008).

13. M. A. Sussman, P. Anversa, Myocardial aging and senescence: Where have the stem cells gone? Annu. Rev. Physiol. 66, 29–48 (2004).

14. A. Hoshino, Y. Mita, Y. Okawa, M. Ariyoshi, E. Iwai-Kanai, T. Ueyama, K. Ikeda, T. Ogata, S. Matoba, Cytosolic p53 inhibits Parkin-mediated mitophagy and promotes mitochondrial dysfunction in the mouse heart. Nat. Commun. 4, 2308 (2013).

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

13 of 14

15. R. Sun, B. Zhu, K. Xiong, Y. Sun, D. Shi, L. Chen, Y. Zhang, Z. Li, L. Xue, S. Gupta, Senescence as a novel mechanism involved in -adrenergic receptor mediated cardiac hypertrophy. PLOS ONE 12, e0182668 (2017).

16. R. A. Boon, K. Iekushi, S. Lechner, T. Seeger, A. Fischer, S. Heydt, D. Kaluza, K. Tréguer, G. Carmona, A. Bonauer, A. J. G. Horrevoets, N. Didier, Z. Girmatsion, P. Biliczki, J. R. Ehrlich, H. A. Katus, O. J. Müller, M. Potente, A. M. Zeiher, H. Hermeking, S. Dimmeler, MicroRNA-34a regulates cardiac ageing and function. Nature 495, 107–110 (2013).

17. H. Hermeking, MicroRNAs in the p53 network: Micromanagement of tumour suppression. Nat. Rev. Cancer 12, 613–626 (2012).

18. S. Hoppins, L. Lackner, J. Nunnari, The machines that divide and fuse mitochondria. Annu. Rev. Biochem. 76, 751–780 (2007).

19. O. C. Losón, Z. Song, H. Chen, D. C. Chan, Fis1, Mff, MiD49, and MiD51 mediate Drp1 recruitment in mitochondrial fission. Mol. Biol. Cell 24, 659–667 (2013).

20. J. E. Lee, L. M. Westrate, H. Wu, C. Page, G. K. Voeltz, Multiple dynamin family members collaborate to drive mitochondrial division. Nature 540, 139–143 (2016).

21. C. Figueroa-Romero, J. A. Iñiguez-Lluhí, J. Stadler, C.-R. Chang, D. Arnoult, P. J. Keller, Y. Hong, C. Blackstone, E. L. Feldman, SUMOylation of the mitochondrial fission protein Drp1 occurs at multiple nonconsensus sites within the B domain and is linked to its activity cycle. FASEB J. 23, 3917–3927 (2009).

22. N. Nakamura, Y. Kimura, M. Tokuda, S. Honda, S. Hirose, MARCH-V is a novel mitofusin 2- and Drp1-binding protein able to change mitochondrial morphology. EMBO Rep. 7, 1019–1022 (2006).

23. D.-H. Cho, T. Nakamura, J. Fang, P. Cieplak, A. Godzik, Z. Gu, S. A. Lipton, S-nitrosylation of Drp1 mediates -amyloid-related mitochondrial fission and neuronal injury. Science 324, 102–105 (2009).

24. T. Gawlowski, J. Suarez, B. Scott, M. Torres-Gonzalez, H. Wang, R. Schwappacher, X. Han, J. R. Yates, M. Hoshijima, W. Dillmann, Modulation of dynamin-related protein 1 (DRP1) function by increased O-linked--N-acetylglucosamine modification (O-GlcNAc) in cardiac myocytes. J. Biol. Chem. 287, 30024–30034 (2012).

25. T. Akaike, T. Ida, F.-Y. Wei, M. Nishida, Y. Kumagai, M. M. Alam, H. Ihara, T. Sawa, T. Matsunaga, S. Kasamatsu, A. Nishimura, M. Morita, K. Tomizawa, A. Nishimura, S. Watanabe, K. Inaba, H. Shima, N. Tanuma, M. Jung, S. Fujii, Y. Watanabe, M. Ohmuraya, P. Nagy, M. Feelisch, J. M. Fukuto, H. Motohashi, Cysteinyl-tRNA synthetase governs cysteine polysulfidation and mitochondrial bioenergetics. Nat. Commun. 8, 1177 (2017).

26. Y. Chen, Y. Liu, G. W. Dorn II, Mitochondrial fusion is essential for organelle function and cardiac homeostasis. Circ. Res. 109, 1327–1331 (2011).

27. K. N. Papanicolaou, R. Kikuchi, G. A. Ngoh, K. A. Coughlan, I. Dominguez, W. C. Stanley, K. Walsh, Mitofusins 1 and 2 are essential for postnatal metabolic remodeling in heart. Circ. Res. 111, 1012–1026 (2012).

28. J. R. Friedman, L. L. Lackner, M. West, J. R. DiBenedetto, J. Nunnari, G. K. Voeltz, ER tubules mark sites of mitochondrial division. Science 334, 358–362 (2011).

29. W. Wang, X. Wang, H. Fujioka, C. Hoppel, A. L. Whone, M. A. Caldwell, P. J. Cullen, J. Liu, X. Zhu, Parkinson’s disease–associated mutant VPS35 causes mitochondrial dysfunction by recycling DLP1 complexes. Nat. Med. 22, 54–63 (2016).

30. S. Li, S. Xu, B. A. Roelofs, L. Boyman, W. J. Lederer, H. Sesaki, M. Karbowski, Transient assembly of F-actin on the outer mitochondrial membrane contributes to mitochondrial fission. J. Cell Biol. 208, 109–123 (2015).

31. W.-K. Ji, A. L. Hatch, R. A. Merrill, S. Strack, H. N. Higgs, Actin filaments target the oligomeric maturation of the dynamin GTPase Drp1 to mitochondrial fission sites. eLife 4, e11553 (2015).

32. A. S. Moore, Y. C. Wong, C. L. Simpson, E. L. F. Holzbaur, Dynamic actin cycling through mitochondrial subpopulations locally regulates the fission–fusion balance within mitochondrial networks. Nat. Commun. 7, 12886 (2016).

33. F. Korobova, T. J. Gauvin, H. N. Higgs, A role for myosin II in mammalian mitochondrial fission. Curr. Biol. 24, 409–414 (2014).

34. M. Nishida, T. Sawa, N. Kitajima, K. Ono, H. Inoue, H. Ihara, H. Motohashi, M. Yamamoto, M. Suematsu, H. Kurose, A. van der Vliet, B. A. Freeman, T. Shibata, K. Uchida, Y. Kumagai, T. Akaike, Hydrogen sulfide anion regulates redox signaling via electrophile sulfhydration. Nat. Chem. Biol. 8, 714–724 (2012).

35. H. Kim, M. C. Scimia, D. Wilkinson, R. D. Trelles, M. R. Wood, D. Bowtell, A. Dillin, M. Mercola, Z. A. Ronai, Fine-tuning of Drp1/Fis1 availability by AKAP121/Siah2 regulates mitochondrial adaptation to hypoxia. Mol. Cell 44, 532–544 (2011).

36. L. Chen, Q. Gong, J. P. Stice, A. A. Knowlton, Mitochondrial OPA1, apoptosis, and heart failure. Cardiovasc. Res. 84, 91–99 (2009).

37. P. Davalli, T. Mitic, A. Caporali, A. Lauriola, D. D’Arca, ROS, cell senescence, and novel molecular mechanisms in aging and age-related diseases. Oxid. Med. Cell. Longev. 2016, 3565127 (2016).

38. A. Cassidy-Stone, J. E. Chipuk, E. Ingerman, C. Song, C. Yoo, T. Kuwana, M. J. Kurth, J. T. Shaw, J. E. Hinshaw, D. R. Green, J. Nunnari, Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization. Dev. Cell 14, 193–204 (2008).

39. F. Nakamura, T. P. Stossel, J. H. Hartwig, The filamins: Organizers of cell structure and function. Cell Adh. Migr. 5, 160–169 (2011).

40. D. Y. Kim, S. Y. Jung, Y. J. Kim, S. Kang, J. H. Park, S. T. Ji, W. B. Jang, S. Lamichane, B. D. Lamichane, Y. C. Chae, D. Lee, J. S. Chung, S.-M. Kwon, Hypoxia-dependent mitochondrial fission regulates endothelial progenitor cell migration, invasion, and tube formation. Korean J. Physiol. Pharmacol. 22, 203–213 (2018).

41. M. G. J. Ford, S. Jenni, J. Nunnari, The crystal structure of dynamin. Nature 477, 561–566 (2011).

42. I. Segura, C. Lange, E. Knevels, A. Moskalyuk, R. Pulizzi, G. Eelen, T. Chaze, C. Tudor, C. Boulegue, M. Holt, D. Daelemans, M. Matondo, B. Ghesquière, M. Giugliano, C. Ruiz de Almodovar, M. Dewerchin, P. Carmeliet, The oxygen sensor PHD2 controls dendritic spines and synapses via modification of filamin A. Cell Rep. 14, 2653–2667 (2016).

43. H. Damke, T. Baba, D. E. Warnock, S. L. Schmid, Induction of mutant dynamin specifically blocks endocytic coated vesicle formation. J. Cell Biol. 127, 915–934 (1994).

44. R. Valdés-Mas, A. Gutiérrez-Fernández, J. Gómez, E. Coto, A. Astudillo, D. A. Puente, J. R. Reguero, V. Álvarez, C. Morís, D. León, M. Martín, X. S. Puente, C. López-Otín, Mutations in filamin C cause a new form of familial hypertrophic cardiomyopathy. Nat. Commun. 5, 5326 (2014).

45. S. Givvimani, C. Munjal, N. Tyagi, U. Sen, N. Metreveli, S. C. Tyagi, M. Tjwa, Mitochondrial division/mitophagy inhibitor (Mdivi) ameliorates pressure overload induced heart failure. PLOS ONE 7, e32388 (2012).

46. S.-B. Ong, S. Subrayan, S. Y. Lim, D. M. Yellon, S. M. Davidson, D. J. Hausenloy, Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation 121, 2012–2022 (2010).

47. M. Ding, Q. Dong, Z. Liu, Z. Liu, Y. Qu, X. Li, C. Huo, X. Jia, F. Fu, X. Wang, Inhibition of dynamin-related protein 1 protects against myocardial ischemia–reperfusion injury in diabetic mice. Cardiovasc. Diabetol. 16, 19 (2017).

48. E. C. So, C.-H. Hsing, C.-H. Liang, S.-N. Wu, The actions of mdivi-1, an inhibitor of mitochondrial fission, on rapidly activating delayed-rectifier K+ current and membrane potential in HL-1 murine atrial cardiomyocytes. Eur. J. Pharmacol. 683, 1–9 (2012).

49. E. A. Bordt, P. Clerc, B. A. Roelofs, A. J. Saladino, L. Tretter, V. Adam-Vizi, E. Cherok, A. Khalil, N. Yadava, S. X. Ge, T. C. Francis, N. W. Kennedy, L. K. Picton, T. Kumar, S. Uppuluri, A. M. Miller, K. Itoh, M. Karbowski, H. Sesaki, R. B. Hill, B. M. Polster, The putative Drp1 inhibitor mdivi-1 is a reversible mitochondrial complex I inhibitor that modulates reactive oxygen species. Dev. Cell 40, 583–594.e6 (2017).

50. M. Packer, C. M. O’Connor, J. K. Ghali, M. L. Pressler, P. E. Carson, R. N. Belkin, A. B. Miller, G. W. Neuberg, D. Frid, J. H. Wertheimer, A. B. Cropp, D. L. DeMets, Effect of amlodipine on morbidiaty and mortality in severe chronic heart failure. Prospective Randomized Amlodipine Survival Evaluation Study Group. N. Engl. J. Med. 335, 1107–1114 (1996).

51. A. Ogino, G. Takemura, H. Kanamori, H. Okada, R. Maruyama, S. Miyata, M. Esaki, M. Nakagawa, T. Aoyama, H. Ushikoshi, M. Kawasaki, S. Minatoguchi, T. Fujiwara, H. Fujiwara, Amlodipine inhibits granulation tissue cell apoptosis through reducing calcineurin activity to attenuate postinfarction cardiac remodeling. Am. J. Physiol. Heart Circ. Physiol. 293, H2271–H2280 (2007).

52. S. Sandmann, R. Claas, J. P. M. Cleutjens, M. J. A. P. Daemen, T. Unger, Calcium channel blockade limits cardiac remodeling and improves cardiac function in myocardial infarction-induced heart failure in rats. J. Cardiovasc. Pharmacol. 37, 64–77 (2001).

53. T. Shimada, M. Yoshiyama, K. Takeuchi, T. Omura, Y. Takemoto, S. Kim, H. Iwao, J. Yoshikawa, Long acting calcium antagonist amlodipine prevents left ventricular remodeling after myocardial infarction in rats. Cardiovasc. Res. 37, 618–626 (1998).

54. S. Sandmann, J. Spormann, F. Prenzel, L. Shaw, T. Unger, Calcium channel blockade limits transcriptional, translational and functional up-regulation of the cardiac calpain system after myocardial infarction. Eur. J. Pharmacol. 453, 99–109 (2002).

55. B. L. Tang, MIRO GTPases in mitochondrial transport, homeostasis and pathology. Cells 5, E1 (2015).

56. F. Korobova, V. Ramabhadran, H. N. Higgs, An actin-dependent step in mitochondrial fission mediated by the ER-associated formin INF2. Science 339, 464–467 (2013).

57. W. Wu, C. Lin, K. Wu, L. Jiang, X. Wang, W. Li, H. Zhuang, X. Zhang, H. Chen, S. Li, Y. Yang, Y. Lu, J. Wang, R. Zhu, L. Zhang, S. Sui, N. Tan, B. Zhao, J. Zhang, L. Li, D. Feng, FUNDC1 regulates mitochondrial dynamics at the ER–mitochondrial contact site under hypoxic conditions. EMBO J. 35, 1368–1384 (2016).

58. R. A. Norris, R. Moreno-Rodriguez, A. Wessels, J. Merot, P. Bruneval, A. H. Chester, M. H. Yacoub, A. Hagège, S. A. Slaugenhaupt, E. Aikawa, J. J. Schott, A. Lardeux, B. S. Harris, L. K. Williams, A. Richards, R. A. Levine, R. R. Markwald, Expression of the familial cardiac valvular dystrophy gene, filamin-A, during heart morphogenesis. Dev. Dyn. 239, 2118–2127 (2010).

59. V. E. Koteliansky, M. A. Glukhova, G. N. Gneushev, J.-L. Samuel, L. Rappaport, Isolation and localization of filamin in heart muscle. Eur. J. Biochem. 156, 619–623 (1986).

60. Y. Feng, M. H. Chen, I. P. Moskowitz, A. M. Mendonza, L. Vidali, F. Nakamura, D. J. Kwiatkowski, C. A. Walsh, Filamin A (FLNA) is required for cell–cell contact in vascular

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

Nishimura et al., Sci. Signal. 11, eaat5185 (2018) 13 November 2018

S C I E N C E S I G N A L I N G | R E S E A R C H A R T I C L E

14 of 14

development and cardiac morphogenesis. Proc. Natl. Acad. Sci. U.S.A. 103, 19836–19841 (2006).

61. J. Schaper, R. Froede, S. Hein, A. Buck, H. Hashizume, B. Speiser, A. Friedl, N. Bleese, Impairment of the myocardial ultrastructure and changes of the cytoskeleton in dilated cardiomyopathy. Circulation 83, 504–514 (1991).

62. S. L. Archer, Mitochondrial dynamics—Mitochondrial fission and fusion in human diseases. N. Engl. J. Med. 369, 2236–2251 (2013).

63. Q.-Q. Shao, T.-P. Zhang, W.-J. Zhao, Z.-W. Liu, L. You, L. Zhou, J.-C. Guo, Y.-P. Zhao, Filamin A: Insights into its exact role in cancers. Pathol. Oncol. Res. 22, 245–252 (2016).

64. Q. Lu, K. Ding, M. P. Frosch, S. Jones, M. Wolfe, W. Xia, G. W. Lanford, Alzheimer’s disease-linked presenilin mutation (PS1M146L) induces filamin expression and -secretase independent redistribution. J. Alzheimers Dis. 22, 235–245 (2010).

65. M. Ino, T. Yoshinaga, M. Wakamori, N. Miyamoto, E. Takahashi, J. Sonoda, T. Kagaya, T. Oki, T. Nagasu, Y. Nishizawa, I. Tanaka, K. Imoto, S. Aizawa, S. Koch, A. Schwartz, T. Niidome, K. Sawada, Y. Mori, Functional disorders of the sympathetic nervous system in mice lacking the 1B subunit (Cav 2.2) of N-type calcium channels. Proc. Natl. Acad. Sci. U.S.A. 98, 5323–5328 (2001).

Acknowledgments: We thank H. Ishihara, K. Murata, and N. Miyazaki (electron microscopy facilities in NIPS, Okazaki, Japan) for technical guidance in the transmission electron microscopy imaging and the Functional Genomics Facility, NIBB Core Research Facilities for technical support in the Mass Spectrometry, Spectrography and Bioimaging Facility. We also thank A. Sherwin from Edanz Group (www.edanzediting.com/ac) for editing a draft of this manuscript. Funding: This work was supported by grants from JST PRESTO Program (13417243 to M.N.), JSPS KAKENHI [15K14959 (to M.N.) and 17K15464 (to A.N.)], and Innovative

Areas [Research in a Proposed Research Area ‘Oxygen Biology ’ (26111011)] from the Ministry of Education, Culture, Sports, Science and Technology. This work was also supported by the Platform Project for Supporting Drug Discovery and Life Science Research [Basis for Supporting Innovative Drug Discovery and Life Science Research (BINDS)] from AMED (JP18am0101091) and the Japan Research Foundation for Clinical Pharmacology (to M.N.). Author contributions: M.N., A.N., T.S., and T. Ishikawa designed the research. A.N., T.S., T. Tanaka, K.S., N.K., N.S., and T. Toyama performed experiments. T.N.-T., S.Y., Y.S., K.K., T. Ide, Y.K., T.A., A.O., and Y.M. contributed new reagents/analytic tools. A.N., T.S., T. Tanaka, T. Ishikawa, T. Ide, Y.K., T.A., A.O., Y.M., and M.N. analyzed and interpreted data. A.N., T.S., and M.N. wrote the paper. Competing interests: A patent (JPWO2016080516A1) has been filed in the International Patent System for part of this work. M.N. and T.I. are named as inventors on this patent. The other authors have declared that they have no competing interests. Data and materials availability: The mass spectrometry data have been deposited in jPOSTrepo (http://repository.jpostdb.org) with the identifier JPST000503. All other data needed to evaluate the conclusions in the paper are present in the paper or the Supplementary Materials.

Submitted 7 March 2018Accepted 12 October 2018Published 13 November 201810.1126/scisignal.aat5185

Citation: A. Nishimura, T. Shimauchi, T. Tanaka, K. Shimoda, T. Toyama, N. Kitajima, T. Ishikawa, N. Shindo, T. Numaga-Tomita, S. Yasuda, Y. Sato, K. Kuwahara, Y. Kumagai, T. Akaike, T. Ide, A. Ojida, Y. Mori, M. Nishida, Hypoxia-induced interaction of filamin with Drp1 causes mitochondrial hyperfission–associated myocardial senescence. Sci. Signal. 11, eaat5185 (2018).

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from

associated myocardial senescence−Hypoxia-induced interaction of filamin with Drp1 causes mitochondrial hyperfission

Akaike, Tomomi Ide, Akio Ojida, Yasuo Mori and Motohiro NishidaIshikawa, Naoya Shindo, Takuro Numaga-Tomita, Satoshi Yasuda, Yoji Sato, Koichiro Kuwahara, Yoshito Kumagai, Takaaki Akiyuki Nishimura, Tsukasa Shimauchi, Tomohiro Tanaka, Kakeru Shimoda, Takashi Toyama, Naoyuki Kitajima, Tatsuya

DOI: 10.1126/scisignal.aat5185 (556), eaat5185.11Sci. Signal. 

all-too-common event.mechanism that increases Drp1 activity after myocardial infarction and a drug that could be repurposed to treat this

channels. These results identify a2+senescence, and myocardial dysfunction independently of the drug's effects on CaAdministering cilnidipine to mice after the induction of myocardial infarction reduced mitochondrial fission, cardiomyocyte

channel blocker, as a small-molecule inhibitor of the interaction between filamin and Drp1.2+characterized as a Caincreased in infarcted heart tissue from mice. Screening identified the antihypertensive drug cilnidipine, initiallyan effect that led to cardiomyocyte senescence. The association of filamin with Drp1, which was actin dependent, was developed nations. They found that the cytoskeletal regulator filamin increased the mitochondrial fission activity of Drp1,why Drp1 activity is increased after myocardial infarction, an unfortunately common cause of mortality and morbidity in

. (see also the Focus by Boyer and Eguchi) sought to understandet aldevelopment of cardiac dysfunction. Nishimura Blocking the excessive mitochondrial fission mediated by Drp1 that occurs after myocardial infarction prevents the

Breaking mitochondria and hearts

ARTICLE TOOLS http://stke.sciencemag.org/content/11/556/eaat5185

MATERIALSSUPPLEMENTARY http://stke.sciencemag.org/content/suppl/2018/11/09/11.556.eaat5185.DC1

CONTENTRELATED

http://stke.sciencemag.org/content/sigtrans/12/587/eaaw1920.fullhttp://stke.sciencemag.org/content/sigtrans/11/560/eaau0144.fullhttp://stm.sciencemag.org/content/scitransmed/10/434/eaan4935.fullhttp://science.sciencemag.org/content/sci/360/6386/336.fullhttp://stke.sciencemag.org/content/sigtrans/10/490/eaal4161.fullhttp://stke.sciencemag.org/content/sigtrans/11/536/eaam5855.fullhttp://stke.sciencemag.org/content/sigtrans/11/556/eaav3267.full

REFERENCES

http://stke.sciencemag.org/content/11/556/eaat5185#BIBLThis article cites 65 articles, 18 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

is a registered trademark of AAAS.Science SignalingYork Avenue NW, Washington, DC 20005. The title (ISSN 1937-9145) is published by the American Association for the Advancement of Science, 1200 NewScience Signaling

Science. No claim to original U.S. Government WorksCopyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of

on July 15, 2020http://stke.sciencem

ag.org/D

ownloaded from