perturbation of the titin/murf1 signaling complex is ...titin is the largest known single protein...

13
RESEARCH ARTICLE Perturbation of the titin/MURF1 signaling complex is associated with hypertrophic cardiomyopathy in a fish model and in human patients Yuta Higashikuse 1,2 , Nishant Mittal 1 , Takuro Arimura 3, *, Sung Han Yoon 4 , Mayumi Oda 1 , Hirokazu Enomoto 1 , Ruri Kaneda 1 , Fumiyuki Hattori 1 , Takeshi Suzuki 2 , Atsushi Kawakami 5 , Alexander Gasch 6 , Tetsushi Furukawa 7 , Siegfried Labeit 6 , Keiichi Fukuda 1 , Akinori Kimura 3 and Shinji Makino 1,8, ABSTRACT Hypertrophic cardiomyopathy (HCM) is a hereditary disease characterized by cardiac hypertrophy with diastolic dysfunction. Gene mutations causing HCM have been found in about half of HCM patients, while the genetic etiology and pathogenesis remain unknown for many cases of HCM. To identify novel mechanisms underlying HCM pathogenesis, we generated a cardiovascular- mutant medaka fish, non-spring heart (nsh), which showed diastolic dysfunction and hypertrophic myocardium. The nsh homozygotes had fewer myofibrils, disrupted sarcomeres and expressed pathologically stiffer titin isoforms. In addition, the nsh heterozygotes showed M-line disassembly that is similar to the pathological changes found in HCM. Positional cloning revealed a missense mutation in an immunoglobulin (Ig) domain located in the M- lineA-band transition zone of titin. Screening of mutations in 96 unrelated patients with familial HCM, who had no previously implicated mutations in known sarcomeric gene candidates, identified two mutations in Ig domains close to the M-line region of titin. In vitro studies revealed that the mutations found both in medaka fish and in familial HCM increased binding of titin to muscle-specific ring finger protein 1 (MURF1) and enhanced titin degradation by ubiquitination. These findings implicate an impaired interaction between titin and MURF1 as a novel mechanism underlying the pathogenesis of HCM. KEY WORDS: Genetics, Familial hypertrophic cardiomyopathy, Titin, Ubiquitin, Myocardial stiffness INTRODUCTION Zebrafish is well established as a model vertebrate in gene function studies (Seeley et al., 2007; Xu et al., 2002). The medaka fish, Oryzias latipes, recently emerged as another important such model system because of its smaller genome size and larger genome diversity (Kasahara et al., 2007). In particular, medaka is an excellent model for studying the cardiovascular system because the embryos are transparent and able to tolerate an absence of blood flow under sufficient oxygen delivery by diffusion (Burggren and Pinder, 1991). It is therefore possible to attribute cardiac phenotype directly to particular genes by ruling out the secondary effect of hypoxia. Hypertrophic cardiomyopathy (HCM) is characterized by cardiac hypertrophy accompanied by myofibrillar disarray and diastolic dysfunction. The first HCM-causing gene identified was a missense mutation in the cardiac β-myosin heavy chain gene (Geisterfer- Lowrance et al., 1990). Such causative mutations were also identified in genes encoding contractile apparatus of sarcomere, such as α-tropomyosin, cardiac troponin T and cardiac myosin- binding protein-C (Ho and Seidman, 2006), whereby HCM was thought to result from compensation for dysregulated sarcomeric contractile function. An HCM-associated mutation (Gerull et al., 2002; Satoh et al., 1999; Itoh-Satoh et al., 2002) identified in the titin gene (TTN) was the first causative gene not encoding contractile apparatus, which implied the existence of a novel pathogenesis without the compensation of systolic dysfunction. Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated muscle. Titin connects the Z-disc to the M-line region, where titin filaments extending from the opposite sides of half-sarcomeres overlap (Ojima et al., 2005; LeWinter and Granzier, 2010). Titin provides a scaffold for the assembly of thick and thin filaments (Gregorio et al., 1999), and provides passive stiffness during diastole (Granzier and Irving, 1995). Recently, titin was proposed to function as a sarcomeric stretch sensor that regulates muscle protein expression and turnover (Lange et al., 2006, 2005; Miller et al., 2004). Titin carries a kinase domain at the M-lineA-band transition zone, and adjacent to the kinase domain is a binding site for muscle-specific ring finger protein 1 (MURF1) (Centner et al., 2001), where the mechanical load on the sarcomere is transmitted through titin and its interaction partners to the nucleus. Functional alteration due to the TTN mutations may increase passive tension upon stretching of the sarcomere (Satoh et al., 1999; Kimura, 2008). Titin is extensively modular in structure and can be thought of as a microscopic necklace comprising up to 166 copies of 90- to 95-amino-acid-residue repeats from the immunoglobulin (Ig) domains. Generally, the Ig domains located in the highly repetitive A-band region of titin share high structural similarity (Müller et al., 2007). Received 1 July 2019; Accepted 25 September 2019 1 Department of Cardiology, Keio University School of Medicine, 35-Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan. 2 Division of Basic Biological Sciences, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan. 3 Laboratory of Genome Diversity, Graduate School of Biomedical Science, Tokyo Medical and Dental University, Tokyo 113-8510, Japan. 4 Department of Interventional Cardiology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, AHSP A9229, Los Angeles, CA 90048, USA. 5 Department of Biological Information, Tokyo Institute of Technology, Yokohama 226-8501, Japan. 6 Department of Integrative Pathophysiology, Medical Faculty Mannheim, Mannheim 68167, Germany. 7 Department of Bio-informational Pharmacology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo 113-8510, Japan. 8 Keio University Health Centre, 35-Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan. *Present address: Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima 890-0065, Japan. Author for correspondence ([email protected]) S.M., 0000-0003-3365-3369 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 1 © 2019. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103 Disease Models & Mechanisms

Upload: others

Post on 22-Sep-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

RESEARCH ARTICLE

Perturbation of the titin/MURF1 signaling complex is associatedwith hypertrophic cardiomyopathy in a fish model and inhuman patientsYuta Higashikuse1,2, Nishant Mittal1, Takuro Arimura3,*, Sung Han Yoon4, Mayumi Oda1, Hirokazu Enomoto1,Ruri Kaneda1, Fumiyuki Hattori1, Takeshi Suzuki2, Atsushi Kawakami5, Alexander Gasch6,Tetsushi Furukawa7, Siegfried Labeit6, Keiichi Fukuda1, Akinori Kimura3 and Shinji Makino1,8,‡

ABSTRACTHypertrophic cardiomyopathy (HCM) is a hereditary diseasecharacterized by cardiac hypertrophy with diastolic dysfunction.Gene mutations causing HCM have been found in about half ofHCM patients, while the genetic etiology and pathogenesis remainunknown for many cases of HCM. To identify novel mechanismsunderlying HCM pathogenesis, we generated a cardiovascular-mutant medaka fish, non-spring heart (nsh), which showeddiastolic dysfunction and hypertrophic myocardium. The nshhomozygotes had fewer myofibrils, disrupted sarcomeres andexpressed pathologically stiffer titin isoforms. In addition, the nshheterozygotes showed M-line disassembly that is similar to thepathological changes found in HCM. Positional cloning revealed amissense mutation in an immunoglobulin (Ig) domain located in the M-line–A-band transition zone of titin. Screening of mutations in 96unrelated patients with familial HCM, who had no previously implicatedmutations in known sarcomeric gene candidates, identified twomutations in Ig domains close to the M-line region of titin. In vitrostudies revealed that the mutations found both in medaka fish and infamilial HCM increased binding of titin to muscle-specific ring fingerprotein 1 (MURF1) and enhanced titin degradation by ubiquitination.These findings implicate an impaired interaction between titin andMURF1 as a novel mechanism underlying the pathogenesis of HCM.

KEYWORDS:Genetics, Familial hypertrophic cardiomyopathy, Titin,Ubiquitin, Myocardial stiffness

INTRODUCTIONZebrafish is well established as a model vertebrate in gene functionstudies (Seeley et al., 2007; Xu et al., 2002). The medaka fish,Oryzias latipes, recently emerged as another important such modelsystem because of its smaller genome size and larger genomediversity (Kasahara et al., 2007). In particular, medaka is an excellentmodel for studying the cardiovascular system because the embryosare transparent and able to tolerate an absence of blood flow undersufficient oxygen delivery by diffusion (Burggren and Pinder, 1991).It is therefore possible to attribute cardiac phenotype directly toparticular genes by ruling out the secondary effect of hypoxia.

Hypertrophic cardiomyopathy (HCM) is characterized by cardiachypertrophy accompanied by myofibrillar disarray and diastolicdysfunction. The first HCM-causing gene identified was a missensemutation in the cardiac β-myosin heavy chain gene (Geisterfer-Lowrance et al., 1990). Such causative mutations were alsoidentified in genes encoding contractile apparatus of sarcomere,such as α-tropomyosin, cardiac troponin T and cardiac myosin-binding protein-C (Ho and Seidman, 2006), whereby HCM wasthought to result from compensation for dysregulated sarcomericcontractile function. An HCM-associated mutation (Gerull et al.,2002; Satoh et al., 1999; Itoh-Satoh et al., 2002) identified in thetitin gene (TTN) was the first causative gene not encoding contractileapparatus, which implied the existence of a novel pathogenesiswithout the compensation of systolic dysfunction.

Titin is the largest known single protein and, after myosin andactin, the third most abundant protein of vertebrate striated muscle.Titin connects the Z-disc to the M-line region, where titin filamentsextending from the opposite sides of half-sarcomeres overlap(Ojima et al., 2005; LeWinter and Granzier, 2010). Titin provides ascaffold for the assembly of thick and thin filaments (Gregorio et al.,1999), and provides passive stiffness during diastole (Granzier andIrving, 1995). Recently, titin was proposed to function as asarcomeric stretch sensor that regulates muscle protein expressionand turnover (Lange et al., 2006, 2005; Miller et al., 2004). Titincarries a kinase domain at the M-line–A-band transition zone, andadjacent to the kinase domain is a binding site for muscle-specificring finger protein 1 (MURF1) (Centner et al., 2001), where themechanical load on the sarcomere is transmitted through titin and itsinteraction partners to the nucleus. Functional alteration due to theTTN mutations may increase passive tension upon stretching of thesarcomere (Satoh et al., 1999; Kimura, 2008). Titin is extensivelymodular in structure and can be thought of as a microscopic necklacecomprising up to 166 copies of 90- to 95-amino-acid-residue repeatsfrom the immunoglobulin (Ig) domains.

Generally, the Ig domains located in the highly repetitive A-bandregion of titin share high structural similarity (Müller et al., 2007).Received 1 July 2019; Accepted 25 September 2019

1Department of Cardiology, Keio University School of Medicine, 35-ShinanomachiShinjuku-ku, Tokyo 160-8582, Japan. 2Division of Basic Biological Sciences,Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan. 3Laboratory ofGenome Diversity, Graduate School of Biomedical Science, Tokyo Medical andDental University, Tokyo 113-8510, Japan. 4Department of InterventionalCardiology, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, AHSP A9229,Los Angeles, CA 90048, USA. 5Department of Biological Information, TokyoInstitute of Technology, Yokohama 226-8501, Japan. 6Department of IntegrativePathophysiology, Medical Faculty Mannheim, Mannheim 68167, Germany.7Department of Bio-informational Pharmacology, Medical Research Institute, TokyoMedical and Dental University, Tokyo 113-8510, Japan. 8Keio University HealthCentre, 35-Shinanomachi Shinjuku-ku, Tokyo 160-8582, Japan.*Present address: Joint Faculty of Veterinary Medicine, Kagoshima University,Kagoshima 890-0065, Japan.

‡Author for correspondence ([email protected])

S.M., 0000-0003-3365-3369

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (https://creativecommons.org/licenses/by/4.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

1

© 2019. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 2: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

However, a subset of Ig domains close to the titin kinase has uniquestructural features. For instance, Ig-A169 provides a binding site forMURF1 (Centner et al., 2001; Müller et al., 2007), a sarcomere-associated E3 ubiquitin ligase that conjugates ubiquitin to proteinsdestined for proteolysis (Bodine et al., 2001; Kedar et al., 2004).MURF1 belongs to the MURF family of proteins, which aretranscribed from three genes, form heterodimers (Centner et al.,2001) and interact with titin kinase. MURFs have also beenproposed to regulate protein degradation and gene expression inmuscle tissues (Lange et al., 2005).Here, we identified the non-spring heart (nsh) medaka fish, a

cardiovascular mutant with a missense mutation in an Ig domainlocated at the M-line–A-band transition zone of titin. The nsh fishshow diastolic dysfunction and hypertrophic heart. To decipher theroles of titin in these pathological effects, we focused on theM-line–A-band transition zone and identified two mutations in other Igdomains of titin in familial HCM patients. These titin mutationsboth increased the binding of titin to MURF1 and enhanced thedegradation of titin by ubiquitination. This is the first reportsuggesting that the impaired binding of MURF1 to titin is a newlyidentified pathogenesis causing HCM.

RESULTSMedaka nsh mutants showed hypertrophic heart anddisrupted sarcomeric structureIn a previous screening for N-ethyl-N-nitrosourea (ENU)-inducedmutations of medaka fish, we identified several mutants withabnormal heart development phenotypes (Taneda et al., 2010).Among these, one mutant showed rigid hearts with hypertrophy.Because the mutant heart had lost elasticity and beat in a rigidmanner, we designated it as non-spring heart (nsh). The nshembryos were indistinguishable from their wild-type (WT) siblingsup to the heart tube stage (48 h), and there was no difference in theheart rate. However, after 72 h, the nsh hearts showed increasedthickening of the myocardial wall, especially in the atrium and evenwithout blood flow (Fig. 1A,B). Despite these abnormalities, themutant embryos survived until 8 days post-fertilization (dpf ), astage around hatching, in a relatively healthy condition withoutforming pericardial edemas.Blood flow was very slow or clogged around 3 dpf in the mutants

due to defects in diastole (see Movies 1,2). However, hemoglobinactivity was normal in both WT embryos and nsh mutants at 3 dpfbased on an o-dianisidine staining analysis (data not shown). Aftercrossing with friend leukemia integration 1 transcription factor( fli1)-green fluorescent protein (GFP) transgenic medaka (Fig. S1),alkaline phosphatase staining analysis revealed normal vascularendothelial cells at 3 dpf and normal blood vessel formation in thenshmutants. To estimate the number of cardiomyocytes, we crossedthe nshmutants with a transgenic medaka [Tg(cmlc2:DsRed2-nuc)]expressing DsRed in the cardiomyocyte nuclei (Fig. 1C,D). Wecounted the nuclei of cardiomyocytes using confocal images andfound no difference in the numbers between WT and nshmutants at3 dpf (Fig. 1C-E).In situ hybridization with cardiac-differentiation markers – atrial

and ventricular myosin heavy chains and cardiac myosin lightchain – demonstrated normal expression of sarcomeric proteins inthe nsh mutant (Fig. 1F-I). A cardiac stretch marker, brainnatriuretic peptide (bnp), is normally expressed at 3 dpf in theWT ventricle, but its expression was increased in the atrium of nshmutants (Fig. 1J,K).We then examined the sarcomeric structures of embryonic

cardiomyocytes by transmission electron microscopy. Sarcomeric

units of the WT cardiomyocytes at 5 dpf showed highly organized,precisely aligned thick and thin myofilaments, flanked by Z-discsand M-lines (Fig. 1L). In the nsh homozygotes, cardiomyocytesappeared abnormal with loosely arranged contractile filaments,where thick and thin filaments were disassembled and Z-discs werenot parallel (Fig. 1M). We next analyzed the sarcomeric structure ofadult cardiomyocytes in the WT and nsh heterozygotes (Fig. 1N,O).Expression of sarcomeric α-actinin was maintained in the nshheterozygotes, although with fewer striations and patchy staining(Fig. 1O). Transmission electron microscopy demonstrated rupturedsarcomeric units with irregular and widened Z-discs and M-lines inthe cardiomyocytes of nsh heterozygotes (Fig. 1P,Q). We haveanalyzed body:heart weight ratio in 3- to 4-month-old adult medakafish. The hearts of nsh heterozygotes were heavier thanWT hearts inrelation to body weight (P=0.01, n=24) (Fig. S2). The nsh mutantsdid not show skeletal paralysis; however, the nsh homozygousembryos had a slightly smaller body, and skeletal myocytesdemonstrated severely disturbed sarcomeric integrity with rupturedmyofibrils (Fig. S3). Unlike inWT, staining forα-actininwas patchy inthe nsh heterozygotes (Fig. S3). By transmission electron microscopy,the skeletal myofibrils of nsh heterozygotes demonstrated occasionallydisrupted M-lines, although they displayed well-formed Z-discsand apparently mature myofibrils (Fig. S3). Body size, movement,lifespan and fertility of the nsh heterozygotes were not affected (datanot shown), although the abnormal sarcomeric structure was inheritedas an autosomal dominant trait.

TTN is the nsh geneTo identify the gene responsible for the nsh phenotype, we carriedout positional cloning. We scored 2016 fishes from nsh×nsh/+crosses and found that the nsh phenotype was linked to the medakalinkage group 21 (LG 21). Single-strand conformationpolymorphism markers from introns 193 to 194 and the 3′UTR ofTTN confined the nsh locus to a 19.0-kb critical region (Fig. 2A).TTN was the only transcript identified in the nsh critical region.Analysis of cDNAs from the nsh mutant and several WT strainsrevealed an adenine-to-thymine transversion causing thesubstitution of D (aspartic acid)-23186 to V (valine) in the nshtitin (Fig. 2B). Medaka titin is encoded by a single gene composedof 219 exons, and the nsh missense mutation was localized to TTNexon 204 near the MURF1-binding region that encodes an Igdomain (Fig. 2C). Alignment of titin from several species revealedthat the nsh mutation occurred at an evolutionary highly conservedamino acid residue (Fig. 2D).

Protein turnover of themutated-Ig-domain titin at theM-line–A-band transition zone was faster than that of WT-Ig-domaintitinTo investigate the molecular mechanisms of TTN mutation as thensh gene, we performed reverse-transcriptase PCR (RT-PCR)analysis of the TTN N-terminal, A-I junction, exon 204 and the titinkinase domain in WT and nsh mutant embryos at 3 dpf. Thisanalysis revealed unchanged expression levels of TTNmRNA in thensh mutant compared to WT (Fig. 3A).

Titin isoform shifts have been reported in pathological conditions(Lahmers et al., 2004; Neagoe et al., 2002; Opitz et al., 2004;Warren et al., 2004; Makarenko et al., 2004). In cardiac muscle, theelastic region expresses two types of titin isoform, termed N2B andN2BA, which arose from alternative splicing and result in titinmolecules that contain either a unique N2B element and shortPEVK region (N2B isoform) or unique N2B and N2A elements anda long PEVK region (N2BA isoform) (Miller et al., 2004).

2

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 3: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

Predominant expression of the N2B isoform results in stiffer cardiactissue, while the expression of the N2BA isoform results in morecompliant cardiac myofibrils (Wu et al., 2002). Changes in therelative proportions of N2B and N2BAwas paralleled by changes indiastolic stiffness (Ouzounian et al., 2008). The N2BA:N2B ratio in

myocardium under conditions of diastolic heart failure is lower thanthat observed during systolic heart failure (van Heerebeek et al.,2006). We detected different titin N2B isoform shifts between theWT and nshmutant titin in the nsh heart, which would be associatedwith less elasticity and increased passive stiffness (Fig. 3B).

Fig. 1. Hypertrophic myocardium and disrupted sarcomeric structure in the medaka nsh mutant heart. (A,B) Morphology of the wild-type (WT) andnsh mutant heart at 3 days post fertilization (dpf ). The mutant showed hypertrophic heart, especially in the atrium (double-headed arrow). The nsh heartlost elasticity and showed increased passive stiffness. Due to diastolic inability (see Movie 2), the blood flow was very slow or clogged around 3 dpf.(C,D) Confocal z-stacked images of the WT and nsh mutant hearts at 3 dpf, in which cardiomyocytes were visualized by the Tg(cmlc2: DsRed2-nuc) signal.These reconstructed confocal images show approximately half of the total cardiomyocytes. (E) Quantification of cardiomyocytes using the transgenic line.Cell counting was performed on the optical slices to attain an accurate count. There was no difference in the number of cardiomyocytes between the WT and nshmutant hearts. NS, statistically not significant (n=5 for WT and nsh). (F-K) In situ RNA hybridization analysis showing cardiac myosin light chain 2 (cmlc2) (F,G),and atrial and ventricular myosin heavy chains (amhc and vmhc, respectively) (H,I) in the WT and nsh mutant at 3 dpf. Normal sarcomeric proteinexpression was observed in the nsh mutant. (J,K) Strong expression of brain natriuretic peptide (bnp; arrowheads) was detected at 3 dpf in the WT ventricle (J),whereas bnp expression was strong in the atrium of nsh mutants (K). (L,M) Transmission electron microscopic findings of medaka embryonic cardiomyocytesat 5 dpf. Myofibrillar arrays were evident in theWT heart (L), but myofibrils were reduced with loosely arranged contractile filaments in the nshmutant (nsh −/−) (M).Thick-filament structures and integrity of the M-line region were disrupted, and Z-discs were not parallel. (N,O) Medaka adult cardiac myocytes in the WTand nsh heterozygotes (nsh +/−) were stained for α-actinin. Expression of sarcomeric α-actinin was maintained in the nsh heterozygous mutant, but fewerstriations and patchy staining was observed (O). (P,Q) Transmission electron microscopy showed the rupture of sarcomeric units, as well as irregular andwidened Z-discs and M-lines in cardiomyocytes of the nsh heterozygotes. The integrity of the thick filaments is disrupted (Q) Z-discs (arrowheads) and M-lines(arrows) are indicated. a, atrium; v, ventricle; Myo, myocardium. Scale bars: 50 µm (A-D),100 µm (F-K), 1 µm (L,M), 5 µm (N,O), 0.5 µm (P,Q).

3

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 4: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

Mutations in the TTN gene and protein have also been linked todisorders of skeletal muscles (Ojima et al., 2005; Gregorio et al.,1999). To analyze the expression of titin proteins in the nshmutant,we conducted indirect immunofluorescence on the skeletal muscleof 3 dpf embryos using an antibody to the A-I junction portion oftitin. In WT, titin appeared in a regularly striated pattern between thesarcomeres of adjacent myofibrils (Fig. 3C). However, the nshmutants showed severely decreased titin immunofluorescence at theA-I junction (Fig. 3D). Analysis of the titin C-terminal region and α-actinin staining demonstrated an altered muscle organization, butthere was no apparent decrease in expression of the titin C-terminalportion (Fig. 3E,F; Fig. S2). These findings indicated that certain titinisoforms at the A-I junction were selectively perturbed.To further investigate the dominant-negative effect of mutant

TTN in vivo, we overexpressed the Ig domain of TTN (GFP-taggedWT-TTN-A156 or E29783V-TTN-A156 construct, which was theequivalent of the D23186V mutation in medaka) (Fig. 3G-J) byinjecting mRNA corresponding to exon 204, in which themissense mutation was found, fused with the GFP fragment intoone-cell-stage medaka embryos. Overexpression of neither theWT nor nsh Ig domain induced a specific cardiac phenotype(n=200 in each case). However, in the nsh-Ig-domain-injectedmedaka embryos, the GFP signal disappeared faster than in theWT-Ig-domain-injected embryos (Fig. 3G-J), demonstrating thatthe nsh Ig domain was susceptible to degradation and/or wasunstable in vivo. The nsh-Ig-domain fragments could not beobserved 2 days after the injection, before the heart began beating,and this could explain why we could not replicate the nshphenotype in the injected embryos (Fig. 3G-J).

Next, we examined the faster turnover of the nsh-Ig titindomain, focusing on the ubiquitin proteasome (UPS) systembecause of the proximity of the nsh mutation to the MURF1-binding site. We found that the nsh-Ig-domain protein wasdegraded faster via the UPS-dependent pathway in vitro than theWT Ig domain (Fig. 3K), and that this effect was rescued by theUPS inhibitor epoxomicin (Fig. 3K), indicating that the turnoverof this specific titin Ig domain is controlled by the UPS. Thisregulation mechanism could involve MURF1 because it is thoughtto act in muscle as a rate-limiting E3 ligase to initiate UPS-mediated degradation.

Medaka nsh mutants showed rigid hearts with normalheart rhythmWe quantified cardiac systolic and diastolic motion of WT and nshmutants (see Movies 1,2) using high-speed video imaging andanalysis with the motion vector prediction (MVP) method asdescribed in the Materials and Methods section. This method couldextract contraction and relaxation motions of cardiomyocytes non-invasively, and evaluate characteristics such as beating rate,orientation of contraction, beating homogeneity and wavepropagation of contraction (da Silva Lopes et al., 2011). Asshown in Fig. 4B,C, maximum systolic and diastolic velocities ofatriumwere decreased from∼183 μm/s inWTembryos to∼64 μm/sin nshmutants, and from ∼140 μm/s inWTembryos to∼35 μm/s innsh mutants, respectively. Similarly, the maximum systolic anddiastolic velocities of ventricles were decreased from ∼189 μm/s inWT embryos to ∼29 μm/s in nsh mutants, and from ∼136 μm/s inWT embryos to ∼22 μm/s in nsh mutant embryos, respectively.

Fig. 2. TTN is the nsh gene. (A) Genetic mapping of the nsh locus on the medaka linkage group 21 (LG 21). To define the nsh gene, we scored 2016 fishfrom nsh/+×nsh/+ mapping crosses. Single-strand conformation polymorphism markers from intron 193-194 and the 3′UTR of TTN confined nsh to a19.0-kb critical region. (B) DNA sequence analysis of cDNAs from the nsh and WT strains revealed an adenine-to-thymine mutation in nsh, causing D(aspartic acid)-23186 to V (valine) mutation (red arrow), located in exon 204. (C) Modular structure of the M-line–A-band transition zone of titin contains abinding site for muscle-specific ring finger protein 1 (MURF1). The missense mutation identified in nsh was located in an Ig domain near this site. (D) Clustal Walignment of human, mouse, zebrafish andmedaka TTN sequences. Medaka titin and human titin proteins share 62% similarity. GenBank or Ensembl accessionnumbers used for the analysis are as follows: human TTN (NM_133378), mouse Ttn (NP_035782), zebrafish ttna (DQ649453) and medaka TTN(ENSORLT00000022736). Alignment of titin protein from several species along with the nsh mutation showed that the mutation affected the evolutionaryconserved amino acid residue (red asterisk). Identical and similar amino acids are indicated by dark blue and light blue boxes, respectively.

4

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 5: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

However, the heart rate between WT and nsh mutants was notchanged significantly (Fig. 4B,C). These data demonstrated that thensh mutated gene was not required for the establishment of cardiacrhythm, but rather for control of cardiac elasticity (Fig. 4B). As wehad previously shown that the nsh-Ig-domain protein was degradedfaster than the WT Ig domain (Fig. 3K), we hypothesized thatcardiac rigidity might be due to excessive degradation of titin. Toconfirm our hypothesis, we performed a rescue experiment with

morpholino knockdown of MURF1. After the injection ofmorpholino, we analyzed cardiac movement by motion vectoranalysis. The maximum systolic and diastolic velocities of both theatrium and ventricle was increased when nsh mutant embryos weremicroinjected with MURF1 morpholino, suggesting that cardiacelasticity was partially recovered (Fig. 4B,C). However, we stillobserved cardiac hypertrophy in MURF1-morpholino-injected nshembryos. This may be because the effects of morpholinos generally

Fig. 3. The mutant TTN Ig domain binds morestrongly to MURF1 and is degraded fasterthan wild type, both in vitro and in vivo.(A) RT-PCR analysis of TTN for the N-terminalportion, A-I junction region, exon 204 and titinkinase domain in the WT and nsh mutantembryos at 3 dpf. Despite the presence of amissense mutation, the level of TTN mRNAs forthese portions in the nsh mutant was similar tothat in the WT. The ef1α transcript was used as acontrol. (B) RT-PCR analysis of titin N2Bisoforms in the WT and nsh mutants. N2Bisoforms were increased. (C-F) Embryonicskeletal muscle of the WT and nsh mutant at 3dpf were stained with antibodies to A-I junctiontitin (C,D) and C-terminal titin (E,F). Despite thesimilar expression level of TTN mRNA,expression of titin containing the A-I junctionisoform was reduced (D). Sarcomeric structureswere disrupted in the nsh mutant (F). (G-J) WTand mutant (E29783V which was the equivalentof D23186V mutation in medaka) Ig fragmentsmRNA injection into medaka embryos. Despitethe same amount of mRNA injection, mutant(E29783V) Ig fragments degraded faster thanWT TTN Ig fragments. (K) Western blots (WBs)of HEK293FT cells transfected with HA-taggedWT and mutant (E29783V) Ig fragments with orwithout a proteosome inhibitor, epoxomicin.Myc-tagged ubiquitin was co-transfected tovisualize ubiquitination. WBs of anti-HA IPs andcontrol normal IgG IPs are shown. Note that themyc-labeled large molecules in mutant TTNtransfectants were present only in the presenceof epoxomicin. (L) GFP-tagged TTN-A156domain co-precipitated with myc-tagged MURF1are shown (top panel). Expression levels of GFP-tagged TTN-A156 (middle panel) and myc-tagged MURF1 (lower panel) was confirmed byWB of whole-cell lysates. Binding pairs were WTor E29783V mutant TTN-A156 in combinationwith MURF1. Dashes indicate no myc-taggedproteins (transfected only with pCMV-Tag3).(M) Densitometric data obtained from the co-IPassays. Bar graphs indicate the amount of IPsnormalized to the input amount of GFP-TTN.Value for WT TTN-A156 with MURF1 wasarbitrarily defined as 1.00 arbitrary unit (AU).Data are represented as means±s.e.m. (n=10).**P<0.001 versus WT. Scale bars: 50 µm.

5

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 6: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

only persist up to 3-4 dpf and the cardiac hypertrophy phenotype isdeveloped at a later stage in nsh mutant embryos.Next, we investigated the binding between MURF1 and the Ig

domainwith or without themutations using the coimmunoprecipitations(co-IPs) assays, and found that co-IPs from cells transfected with themyc-taggedMURF1 construct in combination with the GFP-taggedWT-TTN-A156 or E29783V-TTN-A156 construct revealed that theA156 domain bound MURF1 and that the E29783V mutationsignificantly increased the binding (9.23±2.11 AU; P<0.001)(Fig. 3L,M). Notably, the expression level of mutant-TTN-A156was lower than that of WT-TTN-A156 (Fig. 3L).Taken together, our data indicate that the Ig domain of the

M-line–A-band transition zone of titin plays an important role inmaintaining and organizing the myofibril structure. The severelyreduced titin protein levels in combination with the genetic linkagedata implicated the TTN mutation as being causative of the nshphenotype. The alternations in N2B/N2BA titin isoforms in the nsh

phenotype in turn underscored the probable key role of titin inregulating and maintaining cardiac muscle elasticity. The nextlogical step would have been to rescue the nsh phenotype byinjecting full-length TTN mRNA. However, since the medaka TTNgene is very large, we were unable to clone the full-length cDNA(∼15 kb) to perform the rescue experiments, and as a result thereremains a chance that the wrong variant has been identified.

Mutational analysis of the human TTN gene around theMURF1-binding domain in patients with familial HCMThe proximity of the nsh mutation to the titin-MURF1 bindingdomain and the cardiac phenotype in mutant medaka implicated thetitin M-line–A-band transition zone as a candidate region formutations associated with HCM in humans. We therefore analyzeda total of 96 genetically unrelated Japanese patients with familialHCM who carried apparent family history of HCM consistent withan autosomal dominant trait. They were diagnosed with HCM

Fig. 4. Medaka nshmutants showed rigid hearts with normal heart rhythm. The beating motions of medaka atrium and ventricle were captured with a high-speed camera and were analyzed with the motion vector prediction (MVP) method. (A) Phase-contrast images of medaka embryos of WT (left), nsh mutant(middle) and nsh mutant+MURF1 morpholino (right). The yellow squares in the images indicate the regions of interest (ROI) employed for the motion vectorcalculation of atrium wall. (B) Typical beating profile of atrium observed for WT (left), nsh mutant (middle) and nsh mutant+MURF1 morpholino (right).Motion velocity (the vertical scale) represents averaged motion vector length in the ROI. In both figures, each large peak followed by a small peak represents asingle beat of atrium. Since we obtained the vector lengths simply from the square root of x2+y2 (x and y represent the vector components), the systolic anddiastolic peaks both had positive values. (C) Maximum velocities observed for systolic and diastolic motions in WT, nsh mutant and nsh mutant+MURF1morpholino are shown for atrium and ventricle. Systolic and diastolic velocities were decreased in nshmutants and were partially rescued byMURF1morpholino,while the heart rate was not changed. Data are represented as means±s.e.m. (n=3).

6

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 7: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

according to the diagnostic criteria as described previously (Arimuraet al., 2009). They had been analyzed for mutations in the knowndisease genes for HCM, i.e. genes for contractile elements (MYH7,TNNT2, MYBPC3, TNNI3, MYL2, MYL3, ACTC1, TNNC1),Z-disc elements and I-band elements, and no disease-associatedmutation was found (Arimura et al., 2009). Control subjects were400 healthy Japanese individuals selected at random from non-HCMpatients at Tokyo Medical and Dental Hospital.Exon 204 of medaka TTN, containing the nsh mutation,

corresponds to exon 297 of human TTN; thus, we searched formutations in the patients by direct sequencing of exons 296-307 ofTTN (GenBank accession no. NM_133378), which encoded for theM-line–A-band transition region of titin, including the titin kinasedomain. We identified two disease-associated missense mutations(Fig. 5A): TCT→GCT, leading to the replacement of serine withalanine at position 30186 (S30186A) in exon 301 (Fig. 5B), andGAT→AAT, with the exchange of aspartic acid for asparagine atposition 30994 (D30994N) in exon 306 (Fig. 5C). The probandpatients and their affected relatives were heterozygous for themutations, and both mutations affected highly conserved aminoacids. Quite interestingly, the latter mutation in the M-line–A-bandtransition zone of titin was mapped within the binding site for

MURF1. It should be noted here that all three mutations found in themedaka nshmutant and human HCM patients were located in the Igdomains (Fig. 5A).

Altered binding of titin to MURF1 caused by the TTNmutationsBased on the D30994N mutation being located in the MURF1-binding Ig domain of titin, A168-170, we investigated a possiblefunctional change in binding to MURF1 due to the mutation.GFP-tagged WT- or D30994N-TTN-A168-170 construct wasco-transfected with a myc-tagged MURF1 construct into COS-7cells. Western blot (WB) analysis of co-IPs of the transfected cellsusing anti-myc antibody demonstrated that the D30994N mutationsignificantly increased binding toMURF1 (1.58±0.19 AU; P<0.01)(Fig. 6A,B). The A168-170 domain contains two Ig domains, A168and A169, of which the A169 domain is the predominant binderof MURF1 (Müller et al., 2007), despite the D30994N mutationbeing located in the A168 domain. To further characterize theincreased binding of titin to MURF1 in the presence ofthe D30994N mutation, we performed additional WB analysesof co-IPs from the cells transfected with the myc-tagged MURF1construct in combination with GFP-tagged WT-TTN-A168,

Fig. 5. Mutations in the Ig domains of titin are associated with HCM. (A) Schematic structure of titin at the C-terminal A-band and M-line region. C-terminalends of titin meet in theM-line. Arrows indicate the position of twomutations found in familial HCM. The nshmissensemutation corresponds to exon 297 of humantitin. Fn-3 indicates fibronectin type 3. (B,C) Pedigrees of Japanese families with HCM; affected and unaffected members are indicated by black and whitesymbols, respectively. Upon screening for mutations in exons 296-307 of TTN, which encode for the A-band region of titin, including the titin-kinase domain, twodisease-associated mutations, S30186A (B) and D30994N (C), were identified. The latter mutation is located at the edge of the M-line region of titin and within abinding site for MURF1. Both mutations were found in the Ig domains of titin as indicated by red boxes in A. The proband patient of each family is indicated by thearrow. Presence (+) of the mutations is noted.

7

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 8: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

Fig. 6. Altered binding of titin and MURF1 caused by the TTN mutations. (A) GFP-tagged TTN-A168-170 domain co-immunoprecipitated with myc-taggedMURF1 is shown (top panel). Expression levels of GFP-tagged TTN-A168-170 (middle panel) and myc-tagged MURF1 (lower panel) were evaluated by WB ofwhole-cell lysates. Binding pairs were WT or D30994N mutant TTN-A168-170 in combination with MURF1. Dashes indicate no GFP-tagged or myc-taggedproteins (transfected only with pEGFP-C1 or pCMV-Tag3, respectively). (B) Densitometric data obtained from the co-IP assays. Bar graph indicates the amount ofprecipitate normalized to the input amount of GFP-TTN. Value for WT TTN-A168-170 with MURF1 was arbitrarily defined as 1.00 AU. *P<0.01 versus WT.(C) GFP-tagged TTN-A168 or TTN-A169 domains co-immunoprecipitated with myc-tagged MURF1 are shown (top panel). Expression levels of GFP-taggedTTN-A168 or TTN-A169 (middle panel) and myc-tagged MURF1 (lower panel) were evaluated by WB of whole-cell lysates. Binding pairs were WT or D30994Nmutant TTN-A168 in combination with MURF1, andWTTTN-A169 in combination with MURF1. (D) Densitometric data obtained from the co-IP assays. Bar graphindicates the amount of precipitate normalized to the input amount of GFP-TTN. Value for WT TTN-A168 with MURF1 was arbitrarily defined as 1.00 AU.Data are represented as means±s.e.m. (n=10). *P<0.01 versus WT. (E) GFP-tagged TTN-A160-161 domains co-precipitated with myc-tagged MURF1 areshown (top panel). Expression levels of GFP-tagged TTN-A160-161 (middle panel) and myc-tagged MURF1 (lower panel) was confirmed by WB of whole-celllysates. Binding pairs were WT or S30186A mutant TTN-A160-161 in combination with MURF1. Dashes indicate no myc-tagged proteins (transfected onlywith pCMV-Tag3). (F) Densitometric data obtained from the co-IP assays. Bar graph indicates the amount of IPs normalized to the input amount of GFP-TTN. Value forWT TTN-A160-161 with MURF1 was arbitrarily defined as 1.00 AU. Data are represented as means±s.e.m. (n=10). **P<0.001 versus WT. (G) Titin A168-170 wasubiquitinated by MURF1 and MURF2. The titin fragment A168-170 containing the MURF1-binding site was used as a substrate for in vitro ubiquitination assays.Addition of recombinant MURF1 or MURF2 catalyzes multi-ubiquitination of titin fragments. Lanes: Co, control (no E3 ligase added); M1, inclusion of MURF1;M2, inclusion of MURF2. After incubation, A168-170 fragments were detected by WB, using specific antibodies to titin A169 (left), and A168-170 (right).

8

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 9: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

D30994N-TTN-A168 or WT-TTN-A169 constructs. WT-TTN-A169 showed significantly higher binding to MURF1 thanWT-TTN-A168 (6.69±1.53 AU; P<0.01) (Fig. 6C,D). On theother hand, the D30994N mutation also significantly increasedthe binding of the A168 domain to MURF1 (3.06±0.68 AU;P<0.01) (Fig. 6C,D).Because the other mutation (S30186A) also located in the Ig

domain (A160), we also investigated the binding between MURF1and the A160 Ig domain with or without the mutation using the co-IP assays. The co-IPs from cells transfected with the myc-taggedMURF1 construct in combination with GFP-tagged WT- orS30186A-TTN-A160-161 construct revealed that the A160-161domains also bound MURF1 and that the S30186A mutationsignificantly increased the binding (3.11±0.71 AU; P<0.001)(Fig. 6E,F). Notably, the expression level of mutant-TTN-A160-161 was lower than that of WT-TTN-A160-161 (Fig. 6E). Thesedata suggested that mutations in the Ig domains of the M-line–A-band transition zone enhanced the interactions with MURF1. Thisraises an interesting possibility that the association of MURFs withthe M-line regions could link ubiquitin-transfer pathways to the titinfilament system, which may relate to the physiological turnover oftitin in myofibrils. To test this hypothesis, we performed an in vitroubiquitination assay of titin fragment. The titin fragment A168-170,containing the MURF1-binding site, was used as a substrate.Addition of recombinant MURF1 or MURF2 indeed catalyzedmulti-ubiquitination of titin fragment (Fig. 6G). Moreover, toconfirm this idea, we also performed immuno-electron microscopywith anti-ubiquitin antibodies in WT and MURF1-knockout mice(Bodine et al., 2001). Ubiquitin signals were detected at the Z-line,central I-band and M-line regions in WT sarcomere. It was hard toquantify the signals; however, ubiquitin seemed to be reduced inMURF1-knockout mice at the M-line region (data not shown).Endogenous MURF1 levels were tightly regulated (Centner et al.,2001), in combination with ubiquitination of the titin A168-170fragment by MURFs (Fig. 6G), and increased binding of titin andMURF1 (Fig. 3L; A,C,E) suggested that the interactions of MURF1with titin are critical for maintaining the stability of titin.

DISCUSSIONWe generated a cardiovascular-mutant nsh medaka fish by ENUmutagenesis that developed hypertrophy and diastolic dysfunction.We identified a causative mutation in a region of titin around theMURF1-binding site. The nsh homozygous fish showed looselyarranged contractile units, and died around 8 dpf. The nshheterozygotes showed normal lifespan and fertility, but theyexhibited M-line disassembly in myofibrils, indicating that thephenotype of M-line disassembly was inherited as a dominant trait.Due to the extremely large size and presence of multiple

isoforms, it has been difficult to study the structure and function oftitin. A knock-in mouse of eGFP to the titin gene showed thatsarcomeric titin is more dynamic than previously suggested (daSilva Lopes et al., 2011). The large cardiac titin N2BA isoform israpidly replaced by the smaller N2B isoform both after birth andwith re-expression of the fetal gene program in cardiac pathologicalconditions (Lahmers et al., 2004; Neagoe et al., 2002; Opitz et al.,2004; Warren et al., 2004; Makarenko et al., 2004). The isoformswitch plays an important role in adjusting diastolic function. It hasbeen used with knockout technology to gain insight into the roles oftitin in sarcomere assembly, stability and mechanotransductionduring muscle pathophysiology (Weinert et al., 2006; Radke et al.,2007; Granzier et al., 2009). Based on these studies, it was proposedthat titin provides a scaffold for the developing sarcomere (Gregorio

et al., 1999), and that passive forces maintain the correct localizationof M-lines and tether various signaling and structural molecules(LeWinter and Granzier, 2010). Multiple interacting proteinsintegrate titin into the sarcomere and modulate the titin filamentvia links to basic cellular functions, including trophic signaling andenergy balance.

Herman et al. (2012) recently showed by next-generationsequencing that TTN-truncating mutations are the most commonknown genetic cause of dilated cardiomyopathy (DCM). Theirfindings revealed that mutations associated with DCM wereoverrepresented in the titin A-band, but were absent around theM-band regions of titin. One may therefore speculate that sometruncated titin peptides are degraded via the RNA- and protein-surveillance pathways in DCM. The consequently decreased titinlevels could limit sarcomere formation, resulting in cardiacdysfunction. However, this is not the case for previously reportedTTN mutations that delete only part of the M-band portion of titin(Carmignac et al., 2007); immunohistochemical studies showed thatsome of these C-terminal-truncated titin proteins are integrated intothe sarcomere. If more-proximal TTN-truncating mutations causedDCM through haploinsufficiency, the distribution of suchmutationswould be rather uniform across the susceptible portion of titin.

The current study of our HCM families found point mutationsaround the M-line–A-band transition zone, which is implicated insensing and modulating sarcomeric force. These mutations alsoinduced high binding affinity between titin and MURF1. Using themedaka fish and our in vitro system, we showed that mutated titinfragments are degraded faster by ubiquitination. This studyindicates that titin proteins found in families with HCM, as in theC-terminal titin truncations studied, are integrated into sarcomeresand cause HCM via a dominant-negative mechanism. Our resultsindicated that titin protein mutations in HCM families could beincorporated into the sarcomere and impair MURF1 binding,resulting in HCM through disturbance of the normal turnover of titinand abnormal sarcomere stiffness.

Until now, altered binding of MURF1 to titin has not beenidentified to be closely associated with human HCM or diastolicdysfunction. In this study, we analyzed exons 296-307 (M-line–Aband transition zone) of TTN for mutations in patients with familialHCM and identified two disease-associated missense mutations inthe Ig domains. One of the TTNmutations in familial HCMwas in abinding site for MURF1. Although MURF1 binds to the Ig domainof the M-line–A-band transition region, A168-170, MURF1 wasnot considered to target titin for degradation (Centner et al., 2001).Data in this study and previous studies indicate that MURF1 mayregulate the turnover of critical sarcomeric proteins (Kedar et al.,2004) including some titin isoforms (Bodine et al., 2001;McElhinny et al., 2002). We also found that all the TTN mutationsassociatedwithHCMboth inmedaka and human in the Ig domains ofthe M-line–A-band transition zone increased the binding of MURF1to titin (Fig. 6A,C,E). In addition, the TTN constructs carrying theHCM-associated mutations expressed less protein when they wereinjected into medaka embryos and transfected into cells (Fig. 3G-J).Although the molecular mechanisms underlying this reducedexpression remain to be clarified, it seems to be caused byaccelerated degradation by the ubiquitin proteasome pathway(Figs 3K and 6G). Further, the knockdown of MURF1 in nshmutants partially recovered the cardiac systolic and diastolic velocity.These studies therefore suggests the inhibition ofMURF1 function asa therapeutic intervention for HCM patients.

In summary, we have identified TTNmutations that increased thebinding of titin to MURF1 in a medaka model and in patients with

9

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 10: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

HCM. There are several pathways underlying the pathogenesis ofHCM, including abnormal power generation, power transmission,calcium sensitivity, energy homeostasis and stretch response.MURF1 may be involved with a novel pathway responsible forboth the regulation of myofibril turnover and isoform shift as well asmuscle gene expression, because MURF1 exists in multiplelocations within the cell, including the cytosol, sarcomere andnucleus. Our observations suggest that pathogenic mechanismsunderlying HCM cases that are related to mutations in the titin M-line–A-band transition zone might be a consequence of abnormalprotein turnover and impaired adjustment of diastolic functionregulated by titin and MURF1.

MATERIALS AND METHODSFish strains and maintenanceMedaka fish (Oryzias latipes) were maintained at 28.5°C in a recirculatingsystem with a 14-h day and 10-h night cycle. The Qurt strain, derived fromthe southern Japanese population, was used for the genetic screen. Anotherstrain, Kaga, which came from the northern population in Japan, was usedfor the mapping cross. The medaka heart mutant non-spring heart (nsh) wasisolated by mutational screening (Sakamoto et al., 2004). Fertilized eggswere collected and then incubated at 28.5°C in medaka Ringer’s solution(0.65% NaCl, 0.04% KCl, 0.011% CaCl2, 0.01% MgSO4, 0.01% NaHCO3

and 0.0001% Methylene Blue). Developmental stages were determined bytheir morphology according to the medaka stage table described byIwamatsu (2004). The fli1-GFP transgenic line was kindly gifted byDr A. Kudo (Moriyama et al., 2010). All experimental procedures andprotocols were approved by the Animal Care and Use Committee of KeioUniversity and conformed to the current NIH Guidelines for the Care andUse of Laboratory Animals.

Counting cardiomyocytes in vivoThe DsRed2-nuc gene under control of the zebrafish cardiac myosin lightchain 2 (cmlc2) promoter transgenic strain (Taneda et al., 2010) was used forscoring cardiomyocyte numbers. The WT and nsh homozygous heartscarrying Tg(cmlc: DsRed2-nuc) were dissected, fixed with 4%paraformaldehyde (PFA) in phosphate-buffered saline (PBS) overnight at4°C, and then mounted in 1% agarose gel. We routinely prepared 50-100optical sections from one heart sample, and counted cells on the mergedimages from one in every 25 sections. Data were presented as mean±s.e.m.Student’s t-test was used to test statistical significance.

Whole-mount RNA in situ hybridization (ISH) analysisWhole-mount ISH analysis was performed as described previously (Thisseand Thisse, 2008). After staining, tissues were fixed with 4% PFA in PBSfor color preservation, equilibrated with 80% glycerol and then mounted onslide glass for microscopic observation. The following primers were usedfor cloning the probes by PCR: for cmlc2, F 5′-AATGTCTTTTCCATGT-TTGAGC-3′ and R 5′-CTCCTCTTTCTCATCCCCATG-3′; for atriummyosin heavy chain (amhc), F 5′-TGCACTGATGGCTGAATTTG-3′and R 5′-ACTTGATCTACACCTTGGCC-3′; for ventricular myosinheavy chain (vmhc), F 5′-GCTGAGATGTCCGTGTATGGTGC-3′ and R5′-GCTCCTCACGAGGCCTCTGCTTG-3′; and for brain natriureticpeptide (bnp), F 5′-GATCCATCCATCCATCATCC-3′ and R 5′-TGATA-CTTTAAAGACACAATGTCCAA-3′. The PCR products were cloned intostandard vectors and used to prepare the digoxigenin (dig)-labeled RNA probes.

Histology and electron microscopyWe fixed medaka embryos with 4% PFA in PBS overnight at 4°C, beforedehydrating them in 70% ethanol, mounting in paraffin and sectioning at5 µm thickness. Sections were stained with hematoxylin and eosin (H&E)and viewed using a BIOREVO optical microscope (BZ-9000; Keyence,Japan). For transmission electron microscopy, samples were prepared asdescribed previously (Rottbauer et al., 2001), sectioned using an RMCMT6000 ultramicrotome and visualized using the JEM-1230 (JEOL)electron microscope.

ImmunohistochemistryMedaka embryos were fixed in 4% PFA/PBS overnight at 4°C. Followingfixation and removal of the egg chorion with forceps, samples werecryoprotected in sucrose at 4°C overnight, embedded in OCT compound(Sakura Finetec) and snap-frozen in hexane at −80°C. The sections wereincubated in primary antibodies (see below) overnight at 4°C, washed threetimes for 5 min each in PBS and then incubated in the appropriate secondaryantibodies (Invitrogen) for 1 h at room temperature. Stained sampleswere observed by confocal laser-scanning microscopy (LSM510; CarlZeiss, Jena, Germany). The following primary antibodies were used atthe indicated dilutions: anti-α-actinin (Sigma-Aldrich; A7811) at 1:300,anti-A-I junction Titin [T11 (Sigma-Aldrich; T9030)] at 1:100, and anti-C-terminal Titin [Titin H-300 (Santa Cruz Biotechnology; sc-28536)] at1:100. Secondary antibodies were goat anti-rabbit IgG-FITC (sc-2012,1:100) and goat anti-mouse IgG H&L–Alexa-Fluor-488 (ab150113, 1:100).

High-speed video microscopyMedaka embryos were placed in a bottom of the well (24-multiwell plate)and incubated with 500 μl of medaka Ringer’s solution. Embryoswere arranged to direct their heart towards the bottom of the plateso that the cross-sectional heart images for each embryo can beobserved from a similar direction. A high-speed digital CMOS camera(KP-FM400WCL, Hitachi Kokusai Electric, Tokyo, Japan) was mountedon an inverted microscope (Eclipse TE2000, Nikon, Tokyo, Japan),which was equipped with an xy scanning stage (Bios-T, Sigma Koki,Tokyo, Japan). The microscope was also equipped with a stage-top mini-incubator (WSKM, Tokai Hit, Shizuoka, Japan), which maintained thetemperature of the culture plate at 28.5°C. Videos of medaka embryoswere recorded as sequential phase-contrast images with a 10× or 20×objective at a frame rate of 150 fps, a resolution of 2048×2048 pixels anda depth of 8 bits.

Evaluation of beating heart with MVP algorithmTo evaluate cardiac systolic and diastolic motion of the medaka heart fromthe high-speed video imaging of the embryos, we have applied the MVPmethod as previously reported (Hayakawa et al., 2012; Ghanbari, 1990).Briefly, each frame was divided into square blocks of N×N pixels (Fig. S4).Then, for a maximum motion displacement of w pixels per frame, thecurrent block of pixels was matched to the corresponding block at the samecoordinates in the previous frame within a square window of width N+2w.(Optimal values of N and w for the cardiac motion detection may vary withthe observation magnification and resolution of the employed camera. Here,we set N=16 and w=4. These values were determined empirically based onthe throughput speed of calculation and accuracy of the block-matchingdetection.) The best match on the basis of a matching criterion yielded thedisplacement. The mean absolute error (MAE) was used as the matchingcriterion. The matching function is given by

Mði; jÞ ¼ 1

N2

XN

m¼1

XN

n¼1

j ftðm;nÞ � ft�1ðmþ i;nþ jÞj � w � i; j � w;

where ft(m,n) represents the intensity at coordinates (m,n) in the currentblock of N×N pixels and ft-1(m+i,n+j ) represents the intensity at newcoordinates (m+i,n+j ) in the corresponding block in the previous frame.We performed this calculation for every 4×4 pixels in the region of interest(ROI).

Genetic mapping and positional cloningGenetic mapping was performed as described previously (Kimura et al.,2005). Briefly, an nsh mutant on the Qurt genetic background was crossedwith the Kaga strain to obtain F1 offspring, which were then crossed toobtain the nsh mutant embryos for mapping. A pooled bulk segregationanalysis was performed to localize the nsh mutation on the medaka linkagegroup (Kimura et al., 2005). Further mapping was also done using custom-designed markers based on the sequence polymorphisms that differedbetween the Qurt and Kaga strains. The following primers and restrictionenzymes were used for the mapping: for TTN exon 90, F 5′-AACTTCAC-AGGCTTCAGGTC-3′ and R 5′-CAGCATCTGTGACGTAGCTC-3′

10

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 11: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

(HaeIII); for TTN intron 193-194, F 5′-CCGTGTAACCAACCTCACTG-3′and R 5′-GGCTTGGTCCATTCAAGTGT-3′ (HaeIII); for TTN 3′UTR,F 5′-CAGGAATTGGTCCTGAAAGG-3′ and R 5′-TCGACTTGGGTG-TTTTCAGA-3′; for LG 21_30057841, F 5′-AATAGGTTGAGGCTGC-ATGG-3′ and R 5′-TTCACTGGTTCTTGGCGTTC-3′ (HaeIII). Thereference coding sequence of TTN cDNA was in the Ensembl DNAsequence database under the accession number ENSORLT00000022736.

RT-PCR analysisHearts were harvested fromWT and nshmutant embryos (n=50 for each) at3 dpf. Total RNA was extracted using Trizol reagent (Invitrogen). For RT-PCR analysis, we carried out oligo-dT primed reverse transcription onaliquots (1 µg) of total RNA and used 1/10 of the single-strand cDNAproducts for each PCR amplification. Gene-specific primers were designedbased on published sequences. ef1α was used as an internal control. Thefollowing primers were used for RT-PCR analysis: for N-terminal TTN,F 5′-GCTCTGCACAAAGAGCTGTC-3′ and R 5′-GCAAGATGTGCTG-ATGGTTC-3′; for A-I junction TTN, F 5′-GCAAGCCAAGAACAAGTT-TGA-3′ and R 5′-AGAAAAGGTCGGGACAGAGG-3′; for TTN exon 204,F 5′-TTGTTCATGTCCATCGTGGT-3′ and R 5′-GCTCCTCTGGAGTTG-ATTGC-3′; for titin kinase domain, F 5′-AATGTTGCAAGGCACAAAAA-3′and R 5′-AGCCTCAAGGCTAATGTCCT-3′; for ef1α, F 5′-ATCGTTGCT-GCTGGTGTTG-3′ and R 5′-AGGCGATGTGAGCTGTGTG-3′; for N2Bisoform, F 5′-TATGCCACAACAGCAGGAGA-3′ and R 5′-AAGCCTT-GCAGGTGTATTCG-3′; for N2BA isoform, F 5′-TATGCCACAACAG-CAGGAGA-3′ and R 5′-TTTGGTCTTTGCAGCCTCTT-3′.

Overexpression of TTN Ig domainAWT TTN cDNA fragment covering A156 (from bp89426 to bp89711 ofNM_133378 corresponding to aa29735-aa29829) or TTN mutants carryingthe A-to-T change (E29783V mutation equivalent of the medaka mutationD23186V) was cloned into the pCS2+ expression vector (Turner andWeintraub, 1994). A capped RNA linked to the cDNA for GFP wassynthesized by an MMESSAGE mMACHINE Kit (Ambion) and injectedinto the cytoplasm of one-cell-stage embryos at a final concentration of500 ng/µl.

Cloning of titin fragmentWe prepared fragments of human TTN by PCR from a TTNA156 Ig domainconstruct as templates. AWT TTN cDNA fragment covering the A156 (frombp89426 to bp89711 of NM_133378 corresponding to aa29735-aa29829)domain was obtained by PCR from cDNAs prepared from total heart RNA,and the A-to-T substitution mutant (E29783V mutation equivalent of themedaka mutation D23186V) was created by the primer-mediatedmutagenesis method.

Constructs for epitope-tagged proteinsThe cDNA fragments of WT and mutant TTNwere cloned into 3HA-taggedpcDNA3-NTAP. Full-length MURF1 fragment in a Myc-tagged pCMV-Tag3B construct (McElhinny et al., 2002) was cloned into Flag-taggedpcDNA3-NTAP. The Myc-tagged ubiquitin construct was a kind gift fromDr Nakada (Osaka University, Osaka, Japan). These constructs weresequenced to ensure that no errors were introduced.

Cellular transfection, protein extraction andimmunoprecipitationA total of 8 µg of plasmid DNA was transfected into semi-confluentHEK293FT cells on 6-cm-diameter culture dishes using Lipofectamine2000 (Invitrogen). Epoxomicin or DMSO was added to the culture 4 hbefore lysate preparation. At 24 h after transfection, cells were lysed in400 µl RIPA buffer [25 mM Tris-HCl pH 7.6, 150 mM NaCl, 1% NP-40,1% sodium deoxycholate, 0.1% sodium dodecyl sulfate (SDS)] withprotease inhibitor, incubated on ice for 30 min and centrifuged at15,000 rpm (16,000 g) for 30 min. Supernatants were subjected to WBand immunoprecipitation (IP). For IP, lysates were diluted 1:10 using TGNbuffer (50 mMTris-HCl pH7.5, 150 mMNaCl, 1% Tween-20, 0.3% NP40)and precleaned by protein G PLUS-Agarose (Santa Cruz Biotechnology).The precleaned lysates were incubated with 1 µg anti-HA high-affinity

(3F10, Roche) or normal IgG (GE Healthcare) for 3 h and combined with20 µl protein G PLUS-Agarose (Santa Cruz Biotechnology). The lysateswith agarose-bead complexes were incubated overnight with rotation. Thebead complexes were washed in TGN four times and twice in PBS. Thebeads were suspended in 2×Laemmli sample buffer (Bio-Rad) containing15% 2-mercaptoethanol and boiled for 10 min.

Western blottingCellular lysates and IPs were separated by SDS-PAGE and transferred topolyvinylidene fluoride (PVDF) membranes. After a pre-incubation with3% skim milk in Tris-buffered saline (TBS), the membrane was incubatedwith primary antibody overnight. After TBS washing, the membrane wasincubated with an appropriate secondary antibody {anti-rabbit or anti-mouse IgG HRP-conjugated Ab (1:10,000 in 3% skim milk in TBS; GEHealthcare), mouse TrueBlot ULTRA: anti-mouse Ig HRP [1:1000 in 3%skim milk in TBS containing 0.1% Tween-20 (TBST)]} for 1 h. After fivemore washes in TBST, signals were visualized using the Chemi-Lumi OneSuper (Nacalai) and Luminescent Image Analyzer LAS-3000 mini(Fujifilm, Tokyo, Japan).

AntibodiesWe used the following primary antibodies: first antibodies: anti-Flag(1:1000 in 5%BSA/TBST,M2, F7425, Sigma-Aldrich), anti-HA (1:2000 in3% skim milk/TBST, clone HA-7, H9658 Sigma-Aldrich), myc (1:400 in5% BSA/TBST, clone 9E10, sc-40, Santa Cruz Biotechnology) andGAPDH (1:1000 in 5% BSA/TBST, 14C10, rabbit mAb; Cell SignalingTechnology); second antibodies: ECL anti-mouse IgG, peroxidase-linkedspecies-specific F(ab’)2 fragment (from sheep) (1:10,000 in 3% skim milk/TBST, NA9310; GE Healthcare), ECL anti-rabbit IgG, peroxidase-linkedspecies-specific F(ab’)2 fragment (from donkey) (1:10,000 in 3% skimmilk/TBST, NA9340; GE Healthcare), and mouse TrueBlot ULTRA: anti-mouse Ig HRP (1:1000 in 3% skim milk/TBST; eBioscience). IPs used theanti-HA high-affinity (3F10; Roche) normal mouse IgG (GE Healthcare).

Clinical subjectsWe studied 96 genetically unrelated Japanese patients with HCM, whocarried apparent family history of HCM consistent with an autosomaldominant trait. They were diagnosed with HCM according to the diagnosticcriteria as described previously (Arimura et al., 2009). They had beenanalyzed for mutations in the known disease genes for HCM, i.e. genes forcontractile elements, Z-disc elements and I band elements, and no disease-associated mutation was found (Arimura et al., 2009). Control subjects were400 healthy Japanese individuals selected at random from non-HCMpatients at Tokyo Medical and Dental Hospital. Blood samples wereobtained from the subjects after written informed consent and the researchprotocol was approved by the Ethics Review Committee of MedicalResearch Institute, Tokyo Medical and Dental University, Japan.

Mutational analysisGenomic DNA extracted from blood samples of each individual wassubjected to PCR in an exon-by-exon manner by using primer pairs specificto exons 296-307 of TTN (GenBank accession no. NM_133378), whichencode for titin at the M-line–A-band transition zone including the titin-kinase domain. Sequences of primers and PCR conditions used in this studyare listed in Table S1. PCR products were analyzed by direct sequencingusing Big Dye Terminator chemistry (Applied Biosystems) and an ABI3100DNA analyzer (Applied Biosystems).

Co-immunoprecipitation assayWe used cDNA fragments of human TTN and MURF1 by PCR from TTNA156, A160-161 and A168-170 Ig-like domain constructs and a MURF1full-length construct, respectively, as templates for PCR. A WT TTNcDNA fragment covering the A156 (from bp89426 to bp89711 ofNM_133378 corresponding to aa29735-aa29829), A160-161 (from bp90626to bp91202 corresponding to aa30134-aa30326), A168 (from bp93006 tobp93287 corresponding to aa30928-aa31021), A169 (from bp93288 tobp93581 corresponding to aa31022-aa31119) or A168-A170 (from bp93006to bp93908 corresponding to aa30928-aa31228) domainswas obtained by PCR,

11

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 12: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

and TTNmutants carryingA-to-T (E29783Vmutation equivalent of themedakamutation D23186V), T-to-G (an HCM-associated S30186A mutation) or T-to-A (another HCM-associated D30994N mutation) substitutions were created bythe primer-mediated mutagenesis method. A full-length MURF1 cDNAfragment spanning from bp137 to bp1198 of GenBank Accession No.NM_032588 (corresponding to aa1-aa354) was obtained by PCR. The TTNcDNA fragment was cloned into pEGFP-C1 (Clontech, CA, USA), while theMURF1 cDNA fragmentwas cloned intomyc-tagged pCMV-Tag3 (Stratagene,CA, USA). These constructs were sequenced to ensure that no errors wereintroduced.

Cellular transfection and protein extraction were performed as describedpreviously (Arimura et al., 2009) and co-IP assays were done using theCatch and Release v2.0 Reversible Immunoprecipitation System accordingto the manufacturer’s instructions. IPs were separated by SDS-PAGE andtransferred to nitrocellulose membrane. After a pre-incubation with 3% skimmilk in PBS, the membrane was incubated with primary rabbit anti-mycpolyclonal or mouse anti-GFP monoclonal Ab (1:100, Santa CruzBiotechnology), and with secondary goat anti-rabbit (for polyclonal Ab)or rabbit anti-mouse (for monoclonal Ab) IgGHRP-conjugated Ab (1:2000,Dako A/S, Glostrup, Denmark). Signals were visualized by ImmobilonWestern Chemiluminescent HRP Substrate (Millipore, MA, USA) andLuminescent Image Analyzer LAS-3000mini (Fujifilm, Tokyo, Japan), andtheir densities were quantified using Multi Gauge ver3.0 (Fujifilm, Tokyo,Japan). Numerical datawere expressed asmeans±s.e.m. Statistical differenceswere analyzed using Student’s t-test for paired values. A P-value of less than0.05 was considered to be statistically significant.

In vitro ubiquitination assayIn vitro ubiquitination reactions contained as E1 ligase 100 nM UBE1(Boston Biochem), and 1 µM UbcH5c as E2 ligase (Boston Biochem),150 µM ubiquitin, 4 mM ATP and 2.5 µM recombinant titin A168-170fragment. Reactions were started by addition of 2 µM MURF1 orMURF2, respectively. After incubation for 4 h at room temperature,samples were boiled in SDS loading buffer, and products were separatedby SDS-PAGE and blotted onto nitrocellulose membranes. Titin-specificproducts were detected by using antibodies directed to titin A169 or titinA168-170.

AcknowledgementsWe thank the National Bio Resource Project for supplying medaka strains and othersupport. We thank Dr T. Hayakawa and T. Kunihiro from Sony Corporation for theirtechnical help in the high-speed video recording of medaka embryos and in theimage analysis using the MVP method. We are also grateful to Ms Ono formaintenance of medaka.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsConceptualization: N.M., T.A., S.H.Y., M.O., H.E., R.K., F.H., T.S., A. Kawakami,A.G., T.F., S.L., S.M.; Methodology: Y.H., M.O., S.L., S.M.; Validation: S.M.; Formalanalysis: Y.H., S.H.Y.; Investigation: Y.H., N.M., T.A., S.H.Y., M.O.; Resources:A. Kawakami; Data curation: F.H., S.M.; Writing - original draft: Y.H., N.M., R.K.,A.G., S.L., S.M.; Writing - review & editing: N.M., H.E., F.H., T.S., A. Kimura, S.M.;Visualization: R.K., A.G., S.M.; Supervision: M.O., R.K., T.S., A. Kawakami, T.F.,K.F., A. Kimura, S.M.; Project administration: T.S., A.G., K.F., A. Kimura, S.M.;Funding acquisition: T.S., T.F., K.F., A. Kimura, S.M.

FundingThis study was supported in part by the program for Grant-in-Aid for ScientificResearch (21390248, 24390248, 16H05305, 25670397, 25293096 and19590832) from the Ministry of Education, Culture, Sports, Science andTechnology of Japan, a research grant for Idiopathic Cardiomyopathy from theMinistry of Health, Labour and Welfare, Japan, and a grant for Basic ScientificCooperation Program between Japan and Korea from the Japan Society for thePromotion of Science. It was also supported by the Joint Usage/ResearchProgram of Medical Research Institute, a follow-up grant from the TokyoMedical and Dental University and Association Française contre les Myopathies(Grant No. 15261). This investigation was also supported in parts byThe Mochida Memorial Foundation for Medical and Pharmaceutical Research,

The NOVARTIS Foundation (Japan) for the Promotion of Science, Japan HeartFoundation, the Institute of Life Science, KEIO Gijuku Academic DevelopmentFunds and Keio Kanrinmaru project, Japan.

Supplementary informationSupplementary information available online athttp://dmm.biologists.org/lookup/doi/10.1242/dmm.041103.supplemental

ReferencesArimura, T., Bos, J. M., Sato, A., Kubo, T., Okamoto, H., Nishi, H., Harada, H.,

Koga, Y., Moulik, M., Doi, Y. L. et al. (2009). Cardiac ankyrin repeat protein gene(ANKRD1) mutations in hypertrophic cardiomyopathy. J. Am. Coll. Cardiol. 54,334-342. doi:10.1016/j.jacc.2008.12.082

Bodine, S. C., Latres, E., Baumhueter, S., Lai, V. K., Nunez, L., Clarke, B. A.,Poueymirou, W. T., Panaro, F. J., Na, E., Dharmarajan, K. et al. (2001).Identification of ubiquitin ligases required for skeletal muscle atrophy. Science294, 1704-1708. doi:10.1126/science.1065874

Burggren, W. W. and Pinder, A. W. (1991). Ontogeny of cardiovascular andrespiratory physiology in lower vertebrates. Annu. Rev. Physiol. 53, 107-135.doi:10.1146/annurev.ph.53.030191.000543

Carmignac, V., Salih, M. A., Quijano-Roy, S., Marchand, S., Al Rayess, M. M.,Mukhtar, M. M., Urtizberea, J. A., Labeit, S., Guicheney, P., Leturcq, F. et al.(2007). C-terminal titin deletions cause a novel early-onset myopathy with fatalcardiomyopathy. Ann. Neurol. 61, 340-351. doi:10.1002/ana.21089

Centner, T., Yano, J., Kimura, E., McElhinny, A. S., Pelin, K., Witt, C. C., Bang,M.-L., Trombitas, K., Granzier, H., Gregorio, C. C. et al. (2001). Identification ofmuscle specific ring finger proteins as potential regulators of the titin kinasedomain. J. Mol. Biol. 306, 717-726. doi:10.1006/jmbi.2001.4448

Da Silva Lopes, K., Pietas, A., Radke, M. H. and Gotthardt, M. (2011). Titinvisualization in real time reveals an unexpected level of mobility within andbetween sarcomeres. J. Cell Biol. 193, 785-798. doi:10.1083/jcb.201010099

Geisterfer-Lowrance, A. A., Kass, S., Tanigawa, G., Vosberg, H.-P., McKenna,W., Seidman, C. E. and Seidman, J. G. (1990). A molecular basis for familialhypertrophic cardiomyopathy: a beta cardiac myosin heavy chain gene missensemutation. Cell 62, 999-1006. doi:10.1016/0092-8674(90)90274-I

Gerull, B., Gramlich, M., Atherton, J., McNabb, M., Trombita¡S, K., Sasse-Klaassen, S., Seidman, J. G., Seidman, C., Granzier, H., Labeit, S. et al.(2002). Mutations of TTN, encoding the giant muscle filament titin, cause familialdilated cardiomyopathy. Nat. Genet. 30, 201-204. doi:10.1038/ng815

Ghanbari, M. (1990). The cross-search algorithm formotion estimation. IEEE Trans.Commun. 38, 950. doi:10.1109/26.57512

Granzier, H. L. and Irving, T. C. (1995). Passive tension in cardiac muscle:contribution of collagen, titin, microtubules, and intermediate filaments. Biophys.J. 68, 1027-1044. doi:10.1016/S0006-3495(95)80278-X

Granzier, H. L., Radke, M. H., Peng, J., Westermann, D., Nelson, O. L., Rost, K.,King, N. M. P., Yu, Q., Tschope, C., Mcnabb, M. et al. (2009). Truncation of titin’selastic PEVK region leads to cardiomyopathy with diastolic dysfunction.Circ. Res.105, 557-564. doi:10.1161/CIRCRESAHA.109.200964

Gregorio, C. C., Granzier, H., Sorimachi, H. and Labeit, S. (1999). Muscleassembly: a titanic achievement? Curr. Opin. Cell Biol. 11, 18-25. doi:10.1016/S0955-0674(99)80003-9

Hayakawa, T., Kunihiro, T., Dowaki, S., Uno, H., Matsui, E., Uchida, M.,Kobayashi, S., Yasuda, A., Shimizu, T. and Okano, T. (2012). Noninvasiveevaluation of contractile behavior of cardiomyocyte monolayers based on motionvector analysis. Tissue Eng. Part CMethods 18, 21-32. doi:10.1089/ten.tec.2011.0273

Herman, D. S., Lam, L., Taylor, M. R., Wang, L., Teekakirikul, P., Christodoulou,D., Conner, L., Depalma, S. R., Mcdonough, B., Sparks, E. et al. (2012).Truncations of titin causing dilated cardiomyopathy. N. Engl. J. Med. 366,619-628. doi:10.1056/NEJMoa1110186

Ho, C. Y. and Seidman, C. E. (2006). A contemporary approach to hypertrophiccardiomyopathy. Circulation 113, e858-e862. doi:10.1161/CIRCULATIONAHA.105.591982

Itoh-Satoh, M., Hayashi, T., Nishi, H., Koga, Y., Arimura, T., Koyanagi, T.,Takahashi, M., Hohda, S., Ueda, K., Nouchi, T. et al. (2002). Titin mutations asthe molecular basis for dilated cardiomyopathy. Biochem. Biophys. Res.Commun. 291, 385-393. doi:10.1006/bbrc.2002.6448

Iwamatsu, T. (2004). Stages of normal development in the medaka Oryzias latipes.Mech. Dev. 121, 605-618. doi:10.1016/j.mod.2004.03.012

Kasahara, M., Naruse, K., Sasaki, S., Nakatani, Y., Qu, W., Ahsan, B., Yamada,T., Nagayasu, Y., Doi, K., Kasai, Y. et al. (2007). The medaka draft genome andinsights into vertebrate genome evolution. Nature 447, 714-719. doi:10.1038/nature05846

Kedar, V., McDonough, H., Arya, R., Li, H.-H., Rockman, H. A. and Patterson, C.(2004). Muscle-specific RING finger 1 is a bona fide ubiquitin ligase that degradescardiac troponin I. Proc. Natl. Acad. Sci. USA 101, 18135-18140. doi:10.1073/pnas.0404341102

Kimura, A. (2008). Molecular etiology and pathogenesis of hereditarycardiomyopathy. Circ. J. 72 Suppl. A, A38-A48. doi:10.1253/circj.CJ-08-0050

12

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms

Page 13: Perturbation of the titin/MURF1 signaling complex is ...Titin is the largest known single protein and, after myosin and actin, the third most abundant protein of vertebrate striated

Kimura, T., Yoshida, K., Shimada, A., Jindo, T., Sakaizumi, M., Mitani, H.,Naruse, K., Takeda, H., Inoko, H., Tamiya, G. et al. (2005). Genetic linkage mapof medaka with polymerase chain reaction length polymorphisms. Gene 363,24-31. doi:10.1016/j.gene.2005.07.043

Lahmers, S., Wu, Y., Call, D. R., Labeit, S. and Granzier, H. (2004).Developmental control of titin isoform expression and passive stiffness in fetaland neonatal myocardium. Circ. Res. 94, 505-513. doi:10.1161/01.RES.0000115522.52554.86

Lange, S., Ehler, E. and Gautel, M. (2006). From A to Z and back?Multicompartment proteins in the sarcomere. Trends Cell Biol. 16, 11-18.doi:10.1016/j.tcb.2005.11.007

Lange, S., Xiang, F. and Yakovenko, A. (2005). The kinase domain of titin controlsmuscle gene expression and protein turnover. Science 308, 1599-1603. doi:10.1126/science.1110463

Lewinter, M. M. and Granzier, H. (2010). Cardiac titin: a multifunctional giant.Circulation 121, 2137-2145. doi:10.1161/CIRCULATIONAHA.109.860171

Makarenko, I., Opitz, C. A., Leake, M. C., Neagoe, C., Kulke, M., Gwathmey, J. K.,del Monte, F., Hajjar, R. J. and Linke, W. A. (2004). Passive stiffness changescaused by upregulation of compliant titin isoforms in human dilated cardiomyopathyhearts. Circ. Res. 95, 708-716. doi:10.1161/01.RES.0000143901.37063.2f

McElhinny, A. S., Kakinuma, K., Sorimachi, H., Labeit, S. and Gregorio, C. C.(2002). Muscle-specific RING finger-1 interacts with titin to regulate sarcomeric M-line and thick filament structure and may have nuclear functions via its interactionwith glucocorticoid modulatory element binding protein-1. J. Cell Biol. 157,125-136. doi:10.1083/jcb.200108089

Miller, M. K., Granzier, H. and Ehler, E. (2004). The sensitive giant: the role of titin-based stretch sensing complexes in the heart. Trends Cell Biol. 14, 119-126.doi:10.1016/j.tcb.2004.01.003

Moriyama, A., Inohaya, K., Maruyama, K. and Kudo, A. (2010). Bef medakamutant reveals the essential role of c-myb in both primitive and definitivehematopoiesis. Dev. Biol. 345, 133-143. doi:10.1016/j.ydbio.2010.06.031

Muller, S., Lange, S., Gautel, M. and Wilmanns, M. (2007). Rigid conformation ofan immunoglobulin domain tandem repeat in the A-band of the elastic muscleprotein titin. J. Mol. Biol. 371, 469-480. doi:10.1016/j.jmb.2007.05.055

Neagoe, C., Kulke, M., Del Monte, F., Gwathmey, J. K., de Tombe, P. P., Hajjar,R. J. and Linke, W. A. (2002). Titin isoform switch in ischemic human heartdisease. Circulation 106, 1333-1341. doi:10.1161/01.CIR.0000029803.93022.93

Ojima, K., Ono, Y., Hata, S., Koyama, S., Doi, N. and Sorimachi, H. (2005).Possible functions of p94 in connectin-mediated signaling pathways in skeletalmuscle cells. J. Muscle Res. Cell Motil. 26, 409-417. doi:10.1007/s10974-005-9023-8

Opitz, C. A., Leake, M. C., Makarenko, I., Benes, V. and Linke, W. A. (2004).Developmentally regulated switching of titin size alters myofibrillar stiffness in theperinatal heart.Circ. Res. 94, 967-975. doi:10.1161/01.RES.0000124301.48193.E1

Ouzounian,M., Lee, D. S. and Liu, P. P. (2008). Diastolic heart failure: mechanismsand controversies. Nat. Clin. Pract. Cardiovasc. Med. 5, 375-386. doi:10.1038/ncpcardio1245

Radke, M. H., Peng, J., Wu, Y., Mcnabb, M., Nelson, O. L., Granzier, H. andGotthardt, M. (2007). Targeted deletion of titin N2B region leads to diastolicdysfunction and cardiac atrophy. Proc. Natl. Acad. Sci. USA 104, 3444-3449.doi:10.1073/pnas.0608543104

Rottbauer, W., Baker, K., Wo, Z. G., Mohideen, M.-A. P. K., Cantiello, H. F. andFishman, M. C. (2001). Growth and function of the embryonic heart depend uponthe cardiac-specific L-type calcium channel alpha1 subunit. Dev. Cell 1, 265-275.doi:10.1016/S1534-5807(01)00023-5

Sakamoto, D., Kudo, H., Inohaya, K., Yokoi, H., Narita, T., Naruse, K., Mitani, H.,Araki, K., Shima, A., Ishikawa, Y. et al. (2004). A mutation in the gene for delta-aminolevulinic acid dehydratase (ALAD) causes hypochromic anemia in themedaka, Oryzias latipes.Mech. Dev. 121, 747-752. doi:10.1016/j.mod.2004.03.030

Satoh, M., Takahashi, M., Sakamoto, T., Hiroe, M., Marumo, F. and Kimura, A.(1999). Structural analysis of the titin gene in hypertrophic cardiomyopathy:identification of a novel disease gene. Biochem. Biophys. Res. Commun. 262,411-417. doi:10.1006/bbrc.1999.1221

Seeley, M., Huang, W., Chen, Z., Wolff, W. O., Lin, X. and Xu, X. (2007). Depletionof zebrafish titin reduces cardiac contractility by disrupting the assembly of Z-discsand A-bands.Circ. Res. 100, 238-245. doi:10.1161/01.RES.0000255758.69821.b5

Taneda, Y., Konno, S., Makino, S., Morioka, M., Fukuda, K., Imai, Y., Kudo, A.and Kawakami, A. (2010). Epigenetic control of cardiomyocyte production inresponse to a stress during themedaka heart development.Dev. Biol. 340, 30-40.doi:10.1016/j.ydbio.2010.01.014

Thisse, C. and Thisse, B. (2008). High-resolution in situ hybridization to whole-mount zebrafish embryos. Nat. Protoc. 3, 59-69. doi:10.1038/nprot.2007.514

Turner, D. L. and Weintraub, H. (1994). Expression of achaete-scute homolog 3 inXenopus embryos converts ectodermal cells to a neural fate. Genes Dev. 8,1434-1447. doi:10.1101/gad.8.12.1434

Van Heerebeek, L., Borbely, A., Niessen, H. W., Bronzwaer, J. G. F., Van DerVelden, J., Stienen, G. J. M., Linke, W. A., Laarman, G. J. and Paulus, W. J.(2006). Myocardial structure and function differ in systolic and diastolic heartfailure.Circulation 113, 1966-1973. doi:10.1161/CIRCULATIONAHA.105.587519

Warren, C. M., Krzesinski, P. R., Campbell, K. S., Moss, R. L. and Greaser, M. L.(2004). Titin isoform changes in rat myocardium during development.Mech. Dev.121, 1301-1312. doi:10.1016/j.mod.2004.07.003

Weinert, S., Bergmann, N., Luo, X., Erdmann, B. and Gotthardt, M. (2006). Mline-deficient titin causes cardiac lethality through impaired maturation of thesarcomere. J. Cell Biol. 173, 559-570. doi:10.1083/jcb.200601014

Wu, Y., Bell, S. P., Trombitas, K., Witt, C. C., Labeit, S., Lewinter, M. M. andGranzier, H. (2002). Changes in titin isoform expression in pacing-inducedcardiac failure give rise to increased passive muscle stiffness. Circulation 106,1384-1389. doi:10.1161/01.CIR.0000029804.61510.02

Xu, X., Meiler, S. E., Zhong, T. P., Mohideen,M., Crossley, D. A., Burggren,W.W.and Fishman, M. C. (2002). Cardiomyopathy in zebrafish due to mutation in analternatively spliced exon of titin. Nat. Genet. 30, 205-209. doi:10.1038/ng816

13

RESEARCH ARTICLE Disease Models & Mechanisms (2019) 12, dmm041103. doi:10.1242/dmm.041103

Disea

seModels&Mechan

isms