Titin-truncating variants (TTNtvs) are the major cause of dilated cardiomyopathy (DCM); however, allelic heterogeneity (TTNtvs in different exons) results in variable phenotypes, and remains a major hurdle for disease diagnosis and therapy. Here, we generated a panel of ttn mutants in zebrafish. Four single deletion mutants in ttn.2 or ttn.1 resulted in four phenotypes and three double ttn.2/ttn.1 mutants exhibited more severe phenotypes in somites. Protein analysis identified ttnxu071 as a near-null mutant and the other six mutants as hypomorphic alleles. Studies of ttnxu071 uncovered a function of titin in guiding the assembly of nascent myofibrils from premyofibrils. By contrast, sarcomeres were assembled in the hypomorphic ttn mutants but either became susceptible to biomechanical stresses such as contraction or degenerated during development. Further genetic studies indicated that the exon usage hypothesis, but not the toxic peptide or the Cronos hypothesis, could account for these exon-dependent effects. In conclusion, we modeled TTNtv allelic heterogeneity during development and paved the way for future studies to decipher allelic heterogeneity in adult DCM.

The gene that encodes the largest protein, titin (TTN) (Labeit et al., 1992), was recently established as a causative gene for cardiomyopathies and muscular dystrophies (LeWinter and Granzier, 2013; Chauveau et al., 2014). Approximately 25% of patients with end-stage dilated cardiomyopathy (DCM) have truncating mutations in TTN (Herman et al., 2012). However, the clinical translation of this genetic knowledge has been complicated (Norton et al., 2013). While TTN-truncating variants (TTNtvs) in exons encoding A-band domains cause DCM, many TTNtvs in exons encoding Z-disk domains are benign (Herman et al., 2012). Moreover, mutations found in patients with limb-girdle muscular dystrophy type 2J and tibial muscular dystrophy are more common in C-terminal exons that encode M-line domains of TTN (Hackman et al., 2002; Udd et al., 2005). A dominant toxic peptide hypothesis has been proposed to explain the allelic heterogeneity phenotypes, whereby A-band, but not Z-disk, truncated TTN is incorporated into normal sarcomeres to exert toxic effects on heart function (Herman et al., 2012). On the other hand, exon-dependent phenotypes could be explained by the differential use of the 363 exons of the human TTN gene (reviewed by Guo et al., 2010). In line with this loss-of-function hypothesis, a study of patients with DCM identified a correlation between exon usage and severity of the DCM phenotype (Roberts et al., 2015). More recently, the identification of a short TTN isoform, termed Cronos, prompted a new hypothesis. Driven by an internal promoter located in the intron before exon 240 of TTN, the Cronos isoform only encodes A-band and M-line domains and misses all Z-disk and most I-band domains (Zou et al., 2015). As such, the authors posited that mutations in A-band exons are deleterious because of the disrupted Cronos isoform, whereas mutations in Z-disk exons are benign because of the normal Cronos isoform.

One TTN molecule spans half of a sarcomere, and sequences of TTN domains from N-terminus to C-terminus match the different sarcomeric substructures – including the Z-disk, I-band, A-band and M-line (Labeit and Kolmerer, 1995). Therefore, full-length TTN has been proposed to act as a template during de novo sarcomere assembly (Trinick, 1996). Moreover, the A-band region of TTN has been proposed to function as a ‘molecular ruler’ to govern the assembly of thick filaments and define the length of the A-band at 1.6 µm (Whiting et al., 1989). Despite multiple genetic studies of TTN in the past 20 years, however, the template and ruler hypotheses remain inconclusive. Several Ttn mutants have been generated in mice, including a TTNtv affecting the A-band (Gramlich et al., 2009) and internal deletion mutants that remove the N2B exon (Radke et al., 2007) or an M-line exon (Weinert et al., 2006). However, all mutants were not null, which contributes to the inconclusive findings regarding function of TTN in de novo sarcomere assembly.

Zebrafish is a new vertebrate model for studying sarcomere assembly (Schoenebeck and Yelon, 2007; Huang et al., 2009; Sanger et al., 2009; Yang et al., 2014b). From a large-scale screen using N-ethyl-N-nitrosourea as a mutagen, a group of ttn mutants, named pickwick (pik), have been identified (Xu et al., 2002). Because of the gigantic gene size, the precise location of the mutation was only mapped for the pikm171 allele. Other zebrafish ttn mutant alleles, such as runzel (Steffen et al., 2007) and herzschlag (Myhre et al., 2014), were reported later, but the precise locations of these mutations were not identified. Zebrafish have two ttn homologs, ttna and ttnb (Seeley et al., 2007), which are referred to here as ttn.2 and ttn.1, respectively, according to a new nomenclature (Table S1). We previously used splice-blocking morpholinos to target the N2B and N2A exons of ttn.2 and ttn.1 (Seeley et al., 2007). However, concerns about the specificity of the morpholino technology (Kok et al., 2015; Rossi et al., 2015) and the advent of gene-editing technologies such as transcription activator-like effector nuclease (TALEN)-based methods (Campbell et al., 2013) prompted the generation of stable ttn knockout zebrafish mutants. Whereas a panel of ttn.2 mutants have previously been generated using CRISPR-Cas9 technology (Zou et al., 2015), here, we used TALEN (transcription activator-like effector nuclease) technology to introduce a series of deletion mutations. We detected five phenotypes of different severity from seven different ttn mutants, recapitulating exon-dependent phenotypes. We characterized a near-null ttn mutant and provided the first genetic evidence uncovering functions of titin in de novo sarcomere assembly.

Deletional mutations in four different exons of zebrafish ttn.2 or ttn.1 result in four distinct phenotypes

We designed TALEN pairs targeting sequences in exons 5 and 201 of ttn.2, which encode domains in the Z-disk and the A-band, respectively (xu064 and xu065; Fig. 1A), and in exons 5 and 165 of ttn.1, which encode the corresponding domains in the Z-disk and the A-band (xu066 and xu067; Fig. 1A). For each TALEN, at least two deletion mutant alleles with two different frameshifts were obtained (data not shown). Because the two alleles exhibited the same phenotypes, suggesting no off-target effects, we focused on one allele for each mutant locus.

Fig. 1.

Four ttn deletion mutants are hypomorphic ttn alleles. (A) Schematic representation of the ttn.2 and ttn.1 mutants. Sequences targeted by TALEN pairs are underlined. Presumed stop codons are shown in red text. Restriction enzyme recognition sites for genotyping purposes are boxed. Amino acids after the shift in reading frame are underlined. (B) Protein extracts from 2 dpf larvae of ttn.2xu064 and ttn.2xu065 mutants analyzed by protein gel electrophoresis. A 1 kb DNA ladder is used for molecular size reference. The arrow indicates the anticipated region for the Cronos isoform. WT, wild type. (C) Protein extracts from 2 dpf ttn.1xu066 and ttn.1xu067 mutants analyzed by protein gel electrophoresis. The arrow indicates the smaller band with estimated size. (D,E) ttn.2 and ttn.1 mRNA expression in ttn.2xu064 and ttn.2xu065 larvae (D) and ttn.1xu066 and ttn.1xu067 larvae (E) relative to WT, as revealed by qPCR using primers to exons in the M-line. Data shown are the average of four experiments. gapdh was used as a reference gene. Means±s.d., N=4. *P<0.05, **P<0.01.

Fig. 1.

Four ttn deletion mutants are hypomorphic ttn alleles. (A) Schematic representation of the ttn.2 and ttn.1 mutants. Sequences targeted by TALEN pairs are underlined. Presumed stop codons are shown in red text. Restriction enzyme recognition sites for genotyping purposes are boxed. Amino acids after the shift in reading frame are underlined. (B) Protein extracts from 2 dpf larvae of ttn.2xu064 and ttn.2xu065 mutants analyzed by protein gel electrophoresis. A 1 kb DNA ladder is used for molecular size reference. The arrow indicates the anticipated region for the Cronos isoform. WT, wild type. (C) Protein extracts from 2 dpf ttn.1xu066 and ttn.1xu067 mutants analyzed by protein gel electrophoresis. The arrow indicates the smaller band with estimated size. (D,E) ttn.2 and ttn.1 mRNA expression in ttn.2xu064 and ttn.2xu065 larvae (D) and ttn.1xu066 and ttn.1xu067 larvae (E) relative to WT, as revealed by qPCR using primers to exons in the M-line. Data shown are the average of four experiments. gapdh was used as a reference gene. Means±s.d., N=4. *P<0.05, **P<0.01.

To determine the protein level of titin in these mutants, we used vertical agarose gels followed by silver staining (Warren et al., 2003) and detected two Ttn bands in larvae at 2 days post-fertilization (dpf). Surprisingly, all four mutants showed the same two bands as in wild-type (WT) larvae (Fig. 1B,C). No truncated Ttn bands of smaller size were detected in the ttn.2 mutants (Fig. 1B), which might be partially ascribed to nonsense-mediated mRNA decay since ttn.2 mRNA expression was decreased by more than half in both ttn.2 mutants (Fig. 1D). We detected a shorter Ttn band at ≈2.11 MDa in ttn.1xu067 (Fig. 1C). The ttn.1 transcript was decreased in ttn.1xu066 but remained unchanged in the ttn.1xu067 mutant (Fig. 1E). At the mRNA level, ttn.2 and ttn.1 seemed to regulate each other, as indicated by the decrease in ttn.1 mRNA expression in ttn.2xu064 (Fig. 1D) and the 2- to 3-fold increase in ttn.2 mRNA expression in both ttn.1xu066 and ttn.1xu067 mutants (Fig. 1E).

Both ttn.2 homozygous mutants showed pericardial edema with reduced ventricle size (Fig. S1A), slightly decreased heart rates (Fig. S1B), and nearly depleted shortening fraction at 2 dpf (Fig. S1C). Their hearts appeared as a string at 5 dpf, the bodies exhibited severe abdominal edema at 7 dpf, and most embryos died at ∼10 dpf (Fig. S1D,E). Compared with ttn.2xu064 mutants, which still swam at 3 dpf, the ttn.2xu065 mutants were completely paralyzed (Fig. S1D). Compared with well-assembled myofibrils in somites of WT fish at 2 dpf, myofibrils in ttn.2xu064 appeared slightly misaligned, with reduced sarcomere length (mean±s.d. length, 1.89±0.23 µm in ttn.2xu064 versus 2.21±0.16 µm in WT; Fig. 2A); myofibrils in ttn.2xu065 were severely disrupted, with only residue-striated myofibrils consisting of much shorter sarcomeres (1.32±0.20 µm; Fig. 2A).

Fig. 2.

Exon-dependent functions of titin in sarcomere assembly in somites. (A) ttn.2xu065, but not ttn.2xu064, exhibits disrupted Z-disk formation in somites at 2 dpf. Shown are whole-mount immunostaining for α-Actinin. The inset is a magnified image showing residual striated structure in ttn.2xu065. (B) ttn.1xu066, but not ttn.1xu067, exhibits sarcomere disarray in somites at 9 dpf. (C) Schematic representation of the ttn.2 cis double mutant ttn.2xu068. Arrows indicate locations of mutations. (D) Immunostaining for α-Actinin in somites at 2 dpf indicates that the Z-disk structure in ttn.2xu065 and ttn.2xu068 mutants is disrupted to the same extent. Scale bars: 20 μm and 8 μm (inset in A).

Fig. 2.

Exon-dependent functions of titin in sarcomere assembly in somites. (A) ttn.2xu065, but not ttn.2xu064, exhibits disrupted Z-disk formation in somites at 2 dpf. Shown are whole-mount immunostaining for α-Actinin. The inset is a magnified image showing residual striated structure in ttn.2xu065. (B) ttn.1xu066, but not ttn.1xu067, exhibits sarcomere disarray in somites at 9 dpf. (C) Schematic representation of the ttn.2 cis double mutant ttn.2xu068. Arrows indicate locations of mutations. (D) Immunostaining for α-Actinin in somites at 2 dpf indicates that the Z-disk structure in ttn.2xu065 and ttn.2xu068 mutants is disrupted to the same extent. Scale bars: 20 μm and 8 μm (inset in A).

Unlike ttn.2, the ttn.1 mutants appeared normal until 5 dpf, when an inflated swim bladder failed to develop (Fig. S1D), and both mutants exhibited decreased mobility upon touch stimulation with a needle. ttn.1xu066 mutants died at ∼12 dpf, whereas ttn.1xu067 mutants survived up to 17 dpf (Fig. S1E). Consistently, disarranged myofibrils were noted in ttn.1xu066 somites at 9 dpf when myofibrils in ttn.1xu067 were normal (Fig. 2B). Together, our data revealed four distinct phenotypes resulting from deletion mutations in four different exons of ttn.2 or ttn.1, recapitulating the allelic heterogeneity of TTNtvs in DCM (Herman et al., 2012).

Exon-dependent phenotypes of ttn.2 and ttn.1 may be explained by the exon usage hypothesis

The observation of more severe sarcomere phenotypes in ttn.2xu065 than ttn.2xu064 seems consistent with findings in patients with DCM – that is, a TTNtv in an A-band exon, but not a Z-disk exon, results in DCM (Herman et al., 2012). To directly test the toxic peptide hypothesis, we generated a ttn.2xu068 cis-compound mutant by introducing a Z-disk nonsense mutation into ttn.2xu065/+ (Fig. 2C). The ttn.2xu068 cis-compound homozygous mutant exhibited comparable defects to those in the ttn.2xu065 mutant, which indicates that introducing a Z-disk nonsense mutation fails to rescue the phenotype (Fig. 2D). Together with the lack of truncated proteins in our vertical agarose gels (Fig. 1B), our data argue against the toxic peptide hypothesis.

To assess the Cronos hypothesis (Zou et al., 2015), we tested for the presence of Cronos by protein gel analysis but detected no bands in the region corresponding to the predicted size for the Cronos protein in either WT embryos or ttn mutants (Fig. 1B,C).

To test the exon usage hypothesis (Roberts et al., 2015), we used absolute quantitative reverse transcriptase polymerase chain reaction (qPCR) to compare the expression level of these four exons, with the N2B and N2A exons as controls. In 2 dpf WT larvae, the expression level of exon 5 in ttn.2 was half that of exon 201 (Fig. 3A) and expression of exon 5 in ttn.1 was 6-fold higher than that of exon 165 (Fig. 3B). The different expression levels of the two exons in ttn.2 remain at 5 dpf (Fig. S3B,C) and 9 dpf, respectively (Fig. S3D,E), whereas expression levels of the two exons in ttn.1 become similar. We posited that the different exon usage might result from alternative splicing events. Using qPCR with primers targeting exon 2 of ttn.2 and exon 7 of ttn.1, we detected alternative splicing events around exon 5 of ttn.2 but not around exon 5 of ttn.1 (Fig. 3C). Three alternative splicing isoforms of ttn.2 were found, all of which skipped exon 5 (Fig. 3D). In contrast, we found no alternative splicing events around exon 199 to exon 203 of ttn.2 (Fig. S2A,B) but did note exon-skipping events on exon 165 and exon 166 of ttn.1 (Fig. 3E,F). These findings are consistent with the exon usage data and correlate with the severity of phenotypes in these four ttn.2 or ttn.1 mutants.

Fig. 3.

Alternative splicing and usage of different exons in WT ttn.2 and ttn.1. (A) Exon usage was quantified using absolute qPCR with primers targeting exons of ttn.2. (B) Exon usage in ttn.1. Means±s.d., N=5. **P<0.01. (C) An alternative splicing event can be detected for exon 5 of ttn.2 but not ttn.1. Smaller PCR products (*) can be detected by PCR using primers targeting exon (e)2 and e7 of ttn.2. (D) Schematic representation of three splicing isoforms around e5 of ttn.2. (E) Schematic representation of an exon-skipping event around exon 165 of ttn.1. The red and white blocks indicate Ig and Fn-III domains, respectively. Premature stop codons introduced by the TALEN mutants are labeled. (F) RT-PCR using different combinations of primers targeting ∼e163-e167 of ttn.1 revealed an exon-skipping event on exon 165 and 166.

Fig. 3.

Alternative splicing and usage of different exons in WT ttn.2 and ttn.1. (A) Exon usage was quantified using absolute qPCR with primers targeting exons of ttn.2. (B) Exon usage in ttn.1. Means±s.d., N=5. **P<0.01. (C) An alternative splicing event can be detected for exon 5 of ttn.2 but not ttn.1. Smaller PCR products (*) can be detected by PCR using primers targeting exon (e)2 and e7 of ttn.2. (D) Schematic representation of three splicing isoforms around e5 of ttn.2. (E) Schematic representation of an exon-skipping event around exon 165 of ttn.1. The red and white blocks indicate Ig and Fn-III domains, respectively. Premature stop codons introduced by the TALEN mutants are labeled. (F) RT-PCR using different combinations of primers targeting ∼e163-e167 of ttn.1 revealed an exon-skipping event on exon 165 and 166.

Double ttn.2 and ttn.1 mutants lack compensation and result in more severe phenotypes

The presence of two full-size Ttn bands in all four ttn mutants containing a single deletion mutation suggested their identity as hypomorphic mutants. To test this hypothesis, we generated the ttnxu069 (double-Z) mutant that contains nonsense mutations in Z-disk exons of both ttn homologs and the ttnxu070 (double-A) mutant that contains deletion mutations in A-band exons in both ttn homologs (Fig. 4A). The higher Ttn band disappeared in both double-Z and double-A mutants, but the lower band remained (Fig. 4B,C). The compensation of the Ttn protein level was at least partially removed at the transcriptional level, as indicated by the suppression of ttn.2 mRNA upregulation induced by ttn.1 mutations in these double mutants (Fig. 4D). Compared with ttn.2xu064 and ttn.1xu067 mutants that could swim, further reduction of Ttn.1 in ttn.2xu064 or reduction of Ttn.2 in ttn.1xu067 paralyzed the fish (data not shown). Myofibrils were severely disrupted in both double-Z and double-A mutants (Fig. 4E).

Fig. 4.

Mutants ttnxu69 and ttnxu070 overcome protein compensation and exhibit more severe phenotypes. (A) Schematic representation of two double mutants, ttnxu069 and ttnxu070. Arrows indicate locations of mutations. (B) Protein extracts from 2 dpf larvae of ttnxu69 mutants analyzed by 2% SDS-agarose gel and silver staining. (C) Protein analysis of ttnxu070. (D) ttn.2 and ttn.1 mRNA expression levels in ttnxu070 and ttnxu69 mutants, as revealed by qPCR using primers targeting ttn.2 and ttn.1, respectively. Means±s.d., N=5. **P<0.01. (E) Immunostaining of 2 dpf embryos for α-Actinin. Scale bar: 20 μm.

Fig. 4.

Mutants ttnxu69 and ttnxu070 overcome protein compensation and exhibit more severe phenotypes. (A) Schematic representation of two double mutants, ttnxu069 and ttnxu070. Arrows indicate locations of mutations. (B) Protein extracts from 2 dpf larvae of ttnxu69 mutants analyzed by 2% SDS-agarose gel and silver staining. (C) Protein analysis of ttnxu070. (D) ttn.2 and ttn.1 mRNA expression levels in ttnxu070 and ttnxu69 mutants, as revealed by qPCR using primers targeting ttn.2 and ttn.1, respectively. Means±s.d., N=5. **P<0.01. (E) Immunostaining of 2 dpf embryos for α-Actinin. Scale bar: 20 μm.

Because the lower Ttn band still existed in these cis double mutants, we targeted two highly used exons of ttn.2 and ttn.1, and generated ttnxu071 (Fig. 5A). Both Ttn bands were undetectable in ttnxu071 (Fig. 5B), and both ttn.2 and ttn.1 transcripts were reduced (Fig. 5C). Sarcomeres were disrupted to a similar degree as in the double-A and double-Z mutants (Fig. 5D). Only residual thick-filament-like structures can be detected in the electron micrographs (Fig. 5E).

Fig. 5.

Generation of a ttn.2/1 near-null mutant by targeting two highly used exons. (A) Schematic representation of the ttnxu071 mutant. Arrows indicate locations of mutations. (B) Analysis of protein extracts from 2 dpf WT larvae and ttn.2xu065, ttn.1xu066 and ttnxu071 mutants. (C) qPCR with primers targeting ttn.2 and ttn.1, respectively. Means±s.d., N=5. *P<0.05, ** P<0.01. (D) The Z-disk structure marked by α-Actinin was disrupted in the ttnxu071 mutant. (E) Electron micrographs of somites of 2 dpf WT and ttnxu071 mutant larvae. (F) Z-disk structure was normal in ttn.2xu065/+ and ttn.1xu066 at 2 dpf, but sarcomere disarray was noted in ttnxu071/+ and ttn.1xu066. Scale bars: 20 μm (D,F) and 1 µm (E).

Fig. 5.

Generation of a ttn.2/1 near-null mutant by targeting two highly used exons. (A) Schematic representation of the ttnxu071 mutant. Arrows indicate locations of mutations. (B) Analysis of protein extracts from 2 dpf WT larvae and ttn.2xu065, ttn.1xu066 and ttnxu071 mutants. (C) qPCR with primers targeting ttn.2 and ttn.1, respectively. Means±s.d., N=5. *P<0.05, ** P<0.01. (D) The Z-disk structure marked by α-Actinin was disrupted in the ttnxu071 mutant. (E) Electron micrographs of somites of 2 dpf WT and ttnxu071 mutant larvae. (F) Z-disk structure was normal in ttn.2xu065/+ and ttn.1xu066 at 2 dpf, but sarcomere disarray was noted in ttnxu071/+ and ttn.1xu066. Scale bars: 20 μm (D,F) and 1 µm (E).

The functional redundancy between ttn.2 and ttn.1 was further underscored by our data on ttn.2xu071/+;ttn.1xu066, a mutant generated by crossing ttn.1xu066 into ttnxu071/+. In contrast to the ttn.2xu065/+ and ttn.1xu066 mutants with normal striated Z-disks at 2 dpf, ttn.2xu071/+;ttn.1xu066 larvae showed myofibril disarray (Fig. 5F).

Characterization of the near-null ttnxu071 mutant uncovers the functions of Ttn in sarcomere and thick filament assembly

In a zebrafish embryo at the 18-somite stage, different stages of sarcomere assembly in the slow muscle can be observed simultaneously (Sanger et al., 2009). Z-bodies formed dotted structures with a mean regular distance of 0.89±0.19 µm (mean±s.d.) at the 15th somite, became better striated with an expanded sarcomere length of 1.35±0.27 µm at the 12th somite, and further expanded to 2.14±0.2 µm at the 6th somite (Fig. 6A,C). At the 6th somite, the Z-bodies completed the initial lateral growth process and formed the wider Z-disk with a width of 0.86±0.18 µm (Fig. 6A,B). Ttn started to be expressed as random dots at the 18th somite but became striated at the 12th to 13th somites (Fig. S4).

Fig. 6.

Phenotypes of de novo sarcomere assembly in the ttn.2/1-null mutant. (A) WT or ttnxu071 mutant embryos at the 18-somite stage were stained for α-Actinin. The images were taken at the 6th, 12th and 15th somites (ST). Scale bar: 5 μm. Z-bodies were scattered as irregular dots (open arrowheads) at the 15th somites and became periodic and aligned longitudinally (solid arrowheads) at the 12th somite. In the 6th somite, Z-bodies underwent lateral growth to form striated Z-disks (arrows) in WT embryos but not in the ttnxu071 mutant (solid arrowheads). (B) The width of the Z-disc of WT and ttnxu071 mutants at the 6th somite was quantified. Means±s.d. N=90 for each group from six embryos. **P<0.01. (C) The length of sarcomeres, marked by the distance between two adjacent Z-bodies or Z-disks, in 6th, 12th and 15th somites of WT and ttnxu071 mutant embryos. The length of sarcomeres did not increase at the 6th somite in the ttnxu071 mutant compared with WT, where sarcomere length increased to 2.2 µm. Means±s.d. ***P<0.00001. N=80 for each group in different somites from six embryos. n.s., not significant (P>0.05).

Fig. 6.

Phenotypes of de novo sarcomere assembly in the ttn.2/1-null mutant. (A) WT or ttnxu071 mutant embryos at the 18-somite stage were stained for α-Actinin. The images were taken at the 6th, 12th and 15th somites (ST). Scale bar: 5 μm. Z-bodies were scattered as irregular dots (open arrowheads) at the 15th somites and became periodic and aligned longitudinally (solid arrowheads) at the 12th somite. In the 6th somite, Z-bodies underwent lateral growth to form striated Z-disks (arrows) in WT embryos but not in the ttnxu071 mutant (solid arrowheads). (B) The width of the Z-disc of WT and ttnxu071 mutants at the 6th somite was quantified. Means±s.d. N=90 for each group from six embryos. **P<0.01. (C) The length of sarcomeres, marked by the distance between two adjacent Z-bodies or Z-disks, in 6th, 12th and 15th somites of WT and ttnxu071 mutant embryos. The length of sarcomeres did not increase at the 6th somite in the ttnxu071 mutant compared with WT, where sarcomere length increased to 2.2 µm. Means±s.d. ***P<0.00001. N=80 for each group in different somites from six embryos. n.s., not significant (P>0.05).

Because ttnxu071 is the first near-null ttn mutant allele in a vertebrate (Fig. S4), we interrogated the functions of ttn in de novo sarcomere assembly. The earlier steps of Z-body assembly appeared normal in ttnxu071, as indicated by a regular dotted pattern of Z-bodies at an average distance of 0.80±0.18 µm at the 15th somite and the expansion of the striated Z-bodies to 1.18±0.21 µm at the 12th somite (Fig. 6A,C). However, these Z-bodies failed to grow further in either width (0.45±0.09 µm; Fig. 6B) or length (1.29±0.16 µm) at the 6th somite (Fig. 6A,C).

Similarly, the assembly process of the thick filament in the slow muscle can also be revealed in a single embryo at the 18-somite stage using an F59 antibody. We found that myosin filaments in the WT zebrafish were partially bundled along the long axis of the somite, with an average thick filament length of 0.8±0.15 µm at the 15th somite, became better organized and striated with expanded length (1.46±0.2 µm) at the 12th somite, and finally formed mature A-bands with a length of 1.64±0.13 µm at the 6th somite (Fig. 7A,C). Similar results were obtained using an A4.1025 antibody, which also detects myosin heavy chain isoforms (data not shown). Resulting from lateral growth, the width of myosin filaments also expanded to 0.92±0.14 µm at the 6th somite (Fig. 7B). In contrast, only random myosin dots with a length of 0.83±0.13 µm could be detected in ttnxu071 at the 15th somite, which remained unorganized and were 0.87±0.18 µm long at the 12th somite (Fig. 7A,C). Myosin filaments with a width of 0.6±0.11 µm and length of 1.1±0.21 µm did emerge at the 6th somite (Fig. 7A-C). Different from the WT embryos, these thick filaments did not form mature A-bands, which indicates an instructive role for titin in thick filament assembly.

Fig. 7.

Phenotypes of thick filament assembly in the ttn.2/1-null mutant. (A) Immunostaining was performed in WT or ttnxu071 mutant embryos at the 18-somite stage with an F59 antibody against the myosin heavy chain. Scale bar: 5 μm. Myosin filaments were aligned longitudinally (solid arrowheads) at the 15th somite in WT embryos but remain scattered in ttnxu071 (open arrowheads). Expansion of thick filaments is seen in WT embryos at the 12th and 6th somites (brackets). However, the thick filaments in the ttnxu071 mutant remain scattered at the 12th somite (open arrowheads) and become aligned at the 6th somite (solid arrowheads). (B) The width of the thick (myosin) filaments of WT and ttnxu071 mutants at the 6th somite was quantified. Means±s.d., N=84 for each group from six embryos. **P<0.01. (C) The length of myosin filaments in the 6th, 12th and 15th somites of WT and ttnxu071 mutant embryos. The length of thick filaments does not increase in the ttnxu071 mutant, whereas thick filament length increases to 1.6 µm in WT. Means±s.d., ***P<0.00001. N=80 for each group in different somites from six embryos. n.s., not significant (P>0.05).

Fig. 7.

Phenotypes of thick filament assembly in the ttn.2/1-null mutant. (A) Immunostaining was performed in WT or ttnxu071 mutant embryos at the 18-somite stage with an F59 antibody against the myosin heavy chain. Scale bar: 5 μm. Myosin filaments were aligned longitudinally (solid arrowheads) at the 15th somite in WT embryos but remain scattered in ttnxu071 (open arrowheads). Expansion of thick filaments is seen in WT embryos at the 12th and 6th somites (brackets). However, the thick filaments in the ttnxu071 mutant remain scattered at the 12th somite (open arrowheads) and become aligned at the 6th somite (solid arrowheads). (B) The width of the thick (myosin) filaments of WT and ttnxu071 mutants at the 6th somite was quantified. Means±s.d., N=84 for each group from six embryos. **P<0.01. (C) The length of myosin filaments in the 6th, 12th and 15th somites of WT and ttnxu071 mutant embryos. The length of thick filaments does not increase in the ttnxu071 mutant, whereas thick filament length increases to 1.6 µm in WT. Means±s.d., ***P<0.00001. N=80 for each group in different somites from six embryos. n.s., not significant (P>0.05).

A mutation on exon 5 of ttn.2 affects sarcomere structure in slow but not fast muscles in a contraction-dependent fashion

In ttn.2xu064, sarcomere structures and Z-disks remained relatively normal at 2 dpf (Fig. 2A). Interestingly, thick and thin filaments were undetectable in the middle part of each myofibril, and the blanked middle section was enclosed by two rod-like structures that connected to the remaining myofibrils in each somite segment (Fig. 8A). The rod-like structures could be stained by both Phalloidin and an anti-myosin antibody. This unusual phenotype only occurred in the slow muscles located superficially under the skin and not in the fast muscles located deeper and oblique to the anterior-posterior axis (Devoto et al., 1996), as indicated by both Phalloidin staining and immunostaining using antibodies against fast-muscle tropomyosin and myomesin (Fig. 8A,B).

Fig. 8.

Contraction-dependent myofibril damage to the slow muscle of ttn.2xu064. (A) Slow muscle defects in ttn.2xu064 embryos. Immunostaining for Myosin and Phalloidin staining to label Actin filaments in 2 dpf embryos. Arrows indicate the two rod-like structures enclosing the damaged myofibril region. (B) Fast muscle is intact in ttn.2xu064. Immunostaining with antibodies against Tropomyosin and Myomesin or staining with Phalloidin in 2 dpf embryos. (C) ttn.2xu064 mutants and siblings treated with 1% tricaine for 24 h or 0.07% bleach at 1 dpf for 5 min were fixed and immunostained for Myosin at 2 dpf. Scale bars: 5 μm.

Fig. 8.

Contraction-dependent myofibril damage to the slow muscle of ttn.2xu064. (A) Slow muscle defects in ttn.2xu064 embryos. Immunostaining for Myosin and Phalloidin staining to label Actin filaments in 2 dpf embryos. Arrows indicate the two rod-like structures enclosing the damaged myofibril region. (B) Fast muscle is intact in ttn.2xu064. Immunostaining with antibodies against Tropomyosin and Myomesin or staining with Phalloidin in 2 dpf embryos. (C) ttn.2xu064 mutants and siblings treated with 1% tricaine for 24 h or 0.07% bleach at 1 dpf for 5 min were fixed and immunostained for Myosin at 2 dpf. Scale bars: 5 μm.

Given that a developing zebrafish larva starts to twitch at 27 hpf, when slow muscle is mainly used (McKeown et al., 2009), we posited that slow-muscle-specific phenotypes in ttn.2xu064 may be due to the mechanical stress induced by muscle usage in the setting of reduced sarcomere stability. To test this hypothesis, we inhibited muscle contraction by anesthetizing ttn.2xu064 mutant larvae with 1% tricaine at 24 hpf, right before the first twitch, for 24 h. Indeed, the disrupted myofibrils were significantly rescued (Fig. 8C and Fig. S5). Similar rescuing effects were noted in embryos treated with 10 µM blebbistatin (data not shown), a myosin inhibitor that inhibits muscle contraction via a different mechanism (Straight et al., 2003). By contrast, the disrupted myofibrils were further exaggerated by increasing muscle contraction via either bleaching the chorion (Zhang et al., 2009) or treatment with galanthamine (Otten and Abdelilah-Seyfried, 2013), an acetylcholine esterase inhibitor (Fig. 8C, Fig. S5, and data not shown). Together, our data suggested that partially decreased expression of Ttn affects the stability of sarcomeres in the slow muscle, resulting in higher susceptibility to damage from mechanical stress. Alternatively, this result might reflect different titin isoform usage in slow and fast muscles.

Generation of hypomorphic and near-null mutants of ttn in zebrafish using gene-editing technology

Using TALEN-based genome-editing technology, we generated four zebrafish ttn mutants with mutations affecting single exons in either ttn.2 or ttn.1 and three double mutants. All mutations were small deletions that purposely shift the reading frame and presumably result in stop codons. However, both Ttn bands could be identified by protein gel analysis of the four single mutants, indicating that they are hypomorphic mutants. No truncated Ttn band was detected in any of the mutants except ttn.1xu067. Strong compensational effects must exist, which might include a scavenger system that removes the truncated proteins.

This prediction was validated by our follow-up studies with double mutants. Whereas double-A and double-Z mutants lack the upper band, the ttnxu071 double mutant lacks both bands, resulting in a near-null mutant. We did not term the mutant as a null because of possible residual Ttn protein that is beyond the detection limit of silver staining. Our data challenge the concept that the upper band represents the product of ttn.2 and that the lower band denotes the product of ttn.1 (Steffen et al., 2007). Instead, each Ttn band likely consists of a mixture of ttn.2 and ttn.1 products because neither upper nor lower band can be completely depleted in single mutants. Based on results from mammalian Ttn gels (Granzier and Labeit, 2004), we predict that the two bands might represent N2B- and N2BA-based Ttn isoforms. Given that ttn.2 consists of 234 exons with a 176,965 bp coding sequence, which is longer than ttn.1 (201 exons with 137,988 bp coding sequence) (Seeley et al., 2007), our observation further suggests that there is a length-control mechanism for maintaining the size of Ttn proteins.

Our qPCR studies suggest that the length-control mechanisms and the scavenger mechanism might involve regulation at the transcriptional level. We detected significant downregulation of ttn.2 or ttn.1 transcripts in all mutants except ttn.1xu067, which indicates that nonsense-mediated decay is involved. We detected increased ttn.2 activation in two ttn.1 mutants (Fig. 1E), which suggests genetic compensation induced by the loss of ttn.1. Of note, the changes at the protein level do not always match the changes at the RNA level, as indicated by the decreased protein bands in ttn.1xu066 (Fig. 1C), suggesting additional regulation at the translational level. Further detailed studies are warranted using these zebrafish mutants to uncover compensational mechanisms for regulating the length and isoforms of Ttn.

Hypomorphic ttn mutants uncover titin functions in maintaining sarcomere integrity

By depleting Ttn proteins to different degrees, we noticed five sarcomeric phenotypes of different severity among the seven ttn mutants (Fig. S6). First, de novo sarcomere assembly is completely arrested in the double-Z, double-A and near-null mutants (Fig. 4E and Fig. 5D). The phenotypes are indistinguishable among this group using immunostaining assays, despite the fact that Ttn proteins are disrupted to different degrees, ranging from loss of the upper band to loss of both bands. Second, de novo sarcomere assembly was also arrested in ttn.2xu065, but residue-striated myofibrils could be transiently seen. The ttn.2xu068 double mutant also belongs to this group. Third, sarcomeres can be successfully assembled in ttn.2xu064, despite the disarray of myofibrils shown at 2 dpf. Fourth, sarcomere structure appears normal in ttn.1xu066 at 2 dpf, but myofibril disarray appears at 9 dpf, and the larvae die at 12 dpf. Fifth, sarcomere assembly appears normal in ttn.1xu067 at 9 dpf, but the larvae die at 17 dpf.

This panel of different sarcomeric phenotypes clearly demonstrates a crucial function of Ttn in maintaining the integrity of the sarcomere (Fig. S6). A slight decrease in total Ttn protein expression is not sufficient to affect de novo sarcomere assembly; however, myofibrils become unstable and exhibit phenotypes under biomechanical stress. It remains to be determined whether the detachment of thin and thick filaments in the center of a myofibril is a common initial subcellular form of damage occurring in patients with a TTN mutation.

The generation and characterization of seven ttn mutants with defined mutation locations help to interpret other reported zebrafish ttn mutants. On the basis of their cardiac phenotypes at 2 dpf, it is reasonable to predict that all pik mutants affect ttn.2 (Xu et al., 2002). By contrast, the late-onset phenotypes in both ttn.1 mutants are similar to those described in runzel (Steffen et al., 2007), which suggests that the runzel mutant affects ttn.1 but not ttn.2.

Exon-dependent phenotypes of ttn and their underlying mechanisms

As to the mechanism underlying exon-dependent functions of Ttn.2 or Ttn.1 in sarcomere assembly, our data do not favor the toxic peptide hypothesis for three reasons. First, we did not detect truncated Ttn protein in most ttn mutants. Second, although we did detect a truncated Ttn band in the ttn.1xu067 mutant (but not the ttn.1xu066 mutant), the phenotype was more severe in the Z-disk ttn.1xu066 mutant than the A-band ttn.1xu067 mutant (Fig. 2B). Third, the phenotype of the ttn.2xu068 cis double mutant was similar to that of ttn.2xu065 but not ttn.2xu064 (Fig. 2D).

It remains to be determined whether our conclusions based on fish embryos can be extrapolated to TTN-based adult human diseases. Truncated TTN protein was not detected in samples of skeletal muscle (Gerull et al., 2006) or left ventricles (Roberts et al., 2015) from patients with cardiomyopathy. A recent study using engineered induced pluripotent stem cell-derived cardiomyocytes mimicking different A-band truncation mutations from human patients reported a smaller TTN fragment in only one of the three cell lines (Hinson et al., 2015). In contrast, truncated TTN was found in skeletal muscle biopsy specimens from patients with DCM (Gerull et al., 2002) and in a mouse model (Gramlich et al., 2009). Obviously, a much larger sample size is needed to clarify whether truncated Ttn results from TTNtvs and whether these truncated proteins contribute to pathogenesis.

Our data also do not support the Cronos hypothesis for several reasons: (1) our protein gel analysis failed to detect the Cronos protein, (2) ttn.1xu066 exhibits more severe phenotypes than ttn.1xu067 (Fig. 2B) and (3) the double-Z mutant exhibits phenotypes similar to the double-A mutant (Fig. 4E).

Among the three current hypotheses for explaining exon-dependent phenotypes of ttn.2/1 mutants, our data agree only with the exon usage hypothesis. We demonstrated a correlation among exon usage, alternative splicing patterns (Fig. 3) and severity of phenotypes in the somites (Fig. 2A,B). Consistent with this finding, a recent study showed a strong correlation between ‘percentage spliced in’ of nonsense mutation-containing exons and DCM severity (Roberts et al., 2015), and another study using engineered induced pluripotent stem cells suggested that nonsense mutations in exons of TTN with higher levels of percentage spliced in can cause significant contractile deficits via haploinsufficiency of full-length TTN (Hinson et al., 2015). Our generation of a near-null ttn mutant allele by targeting highly used exons in ttn.2 and ttn.1 gives more supporting evidence that exon usage is the underlying mechanism (Fig. 5).

A near-null ttn mutant validates template functions of titin in assembly of sarcomeres and thick filaments

Generation of the first near-null ttn mutant in a vertebrate enables us to genetically test the function of Ttn, which was proposed 20 years ago (Whiting et al., 1989; Trinick, 1996). We found that depletion of Ttn did not affect the alignment of Z-bodies to form a striated pattern at the 12th to 15th somites in an 18-somite embryo (Fig. 6A,C), which indicates that Ttn is not required for assembly of the premyofibrils. Ttn is needed, however, for the formation of nascent myofibrils from premyofibrils, which is characterized as length expansion of the sarcomere from 0.8 to 2.2 µm and widening of the Z-bodies to form Z-disks.

The ttnxu071 mutant also uncovered two functions of Ttn in thick filament assembly. First, Ttn facilitates the incorporation of the random myosin dots into thin filament networks. When Ttn is absent, myosin filaments remain scattered in the 12th and 15th somites in an 18-somite embryo. Incorporation of the thick filaments into the thin filament networks is delayed until a later developmental stage, as represented by the 6th somite in an 18-somite embryo. Second, Ttn is required for the expansion of the rod-like thick filaments, with a length of ∼0.8 µm, to a uniform A-band with a length of 1.6 µm. This assembly step of the thick filaments is part of the sarcomere assembly step when premyofibrils transform into nascent myofibrils.

Together, our genetic evidence supports the following hypothesis regarding functions of Ttn (Fig. 9). At the premyofibril stage, the Ttn N-terminus binds to the Z-bodies in the existing myofibril network, and its flexible C-terminus binds the thick filaments that are assembled independently and still scattered in the cell (Hill et al., 1986; Wang et al., 1988; Komiyama et al., 1990). Ttn facilitates the incorporation of the thick filaments into the thin filament networks and then guides the lateral alignment of thick filaments and Z-bodies to form mature nascent myofibrils (Fig. 9). Of note, our studies were carried out at the 18 somite stage to eliminate interference from the fast muscle, as its myofibrillogenesis occurs several hours later. Whether our conclusion based on studies in slow muscle is applicable to fast muscle requires future experimental validation. Moreover, our genetic data cannot be used to support the idea that titin functions as a ‘molecular ruler’ to define the length of the A-band – a hypothesis that has also been disproved by a study of a mouse mutant harboring an internal deletion of titin at its I-band/A-band junction (Granzier et al., 2014).

Fig. 9.

Schematic indicating functions of titin in de novo sarcomere assembly. ST, somite; 18S, 18 somite stage.

Fig. 9.

Schematic indicating functions of titin in de novo sarcomere assembly. ST, somite; 18S, 18 somite stage.

Our data seem to contradict a previous publication stating that the A-band rod domain of Ttn.2 is not essential for thick filament assembly (Myhre et al., 2014). The conclusion was based on herzschlag, a ttn.2 mutant harboring an unidentified mutation mapped to the I-band region of ttn.2. It was assumed that the unidentified mutation produced a truncated Ttn.2 protein that loses its thick-filament binding domains, which might not be correct. It is possible that herzschlag is a hypomorphic allele but not a null allele.

Implications for TTN-based human diseases

In summary, our studies of a panel of ttn mutants uncover functions of Ttn in both de novo sarcomere assembly and sarcomere integrity after assembly. Importantly, our data show that a TTNtv cannot be assumed to be a truncating mutation without supporting evidence from a protein gel. Thus, the nomenclature for these nonsense variants as TTNtvs can be misleading. A slight decrease in the Ttn pool is likely to be sufficient to result in a wide spectrum of phenotypes. The weakened myofibrils have much higher susceptibility to damage incurred by stresses such as muscle contraction.

Caution must be taken when extrapolating these mechanisms uncovered in sarcomere assembly during somitogenesis to cardiomyopathy and muscular dystrophy, which mainly occur in adults. An obvious future direction that is enabled by our study is to raise this panel of ttn mutants to adulthood and then assess phenotypes in both heart and muscle. With the convenience of introducing mutations precisely into any exon of ttn, it is anticipated that future studies using the zebrafish model will considerably facilitate our understanding of titinopathy and its related therapies.

Fish management

Zebrafish were handled following the guidelines of the Mayo Clinic Institutional Animal Care and Use Committee (IACUC protocol A17814). The WIK line was used.

Generation of ttn mutants via TALENs

TALEN pairs in ttn.2 and ttn.1 (Seeley et al., 2007) were designed using Zifit (http://zifit.partners.org/ZiFiT/ChoiceMenu.aspx) and assembled using a Golden Gate kit (Addgene) (Cermak et al., 2011). Capped mRNAs were synthesized using mMESSAGE mMachine T3 (Ambion) and injected into 1-cell-stage embryos. Successful mutagenesis was confirmed by PCR followed by digestion with restriction enzymes (Table S2). Two mutant alleles that result in two different shifts of reading frame for each mutant locus were selected. Because ttn.2 and ttn.1 are located on the same chromosome, double ttn mutants were generated by injecting a second TALEN pair into an existing ttn mutant.

Vertical agarose gel electrophoresis

Protein samples from zebrafish larvae were extracted by homogenization using a mortar and pestle (Thermo Fisher Scientific) in sample buffer [8 M urea, 3% v/v SDS, 2 M thiourea, 0.05 M Tris-HCl (pH 8.0), 0.03% w/v Bromophenol Blue, 75 mM DTT and 0.01% protease inhibitors (Roche)]. After the samples were incubated in 65°C prewarmed water for 10 min and centrifuged at 4°C for 5 min, the samples were separated on a 2% SDS/agarose gel (30% v/v glycerol, 0.05 M Tris base, 0.384 M glycine, 0.1% w/v SDS and 2% w/v SeaKem Gold agarose) using a vertical agarose gel electrophoresis system (Warren et al., 2003); 1 kb Plus DNA Ladder (Invitrogen) was used for size markers. The gel was fixed in prefixing solution (50% v/v methanol, 12% v/v glacial acetic acid and 5% w/v glycerol), dried in an oven at 50°C overnight and visualized by silver staining (Bio-Rad).

Relative and absolute qPCR studies

Total RNA from 2 dpf zebrafish larvae was extracted using TRIzol (Sigma). Purified RNA (500 ng) was used to generate cDNA using the Superscript III First-Strand Synthesis System (Invitrogen). Relative qPCR was carried out in 96-well QPCR plates (Roche) using a Roche LightCycler 480 QPCR apparatus with the primers listed in Table S2. Gene expression was normalized to the expression level of glyceraldehyde-3-phosphate dehydrogenase (gapdh) using –ΔΔCt (cycle threshold) values. The efficiency of the primers and absolute qPCR was performed using an external standard curve with calibrated 10-fold serial dilutions of the RT-PCR product. Each measurement contained at least three biological replicates and data were analyzed with JMP statistical software.

Whole-mount immunostaining

Dechorionated embryos at the desired stages were fixed in 4% paraformaldehyde in PBS for 20 min at room temperature, permeabilized with 0.1% Triton X-100 in PBS (PBST), blocked in 10% normal sheep serum (Jackson ImmunoResearch) in PBS for 1 h, and stained with primary antibody for 1 h at room temperature. After the embryos were washed in PBST three times for 10 min each, they were incubated for 1 h with secondary antibodies conjugated with Alexa Fluor 488 or 568 (1:1000; Invitrogen). After the embryos were washed in PBST for 10 min three times, they were incubated with the mounting medium with DAPI (Vector Laboratories). Antibodies against the following targets were used: α-Actinin (1:1000; Sigma, A7811), Myosin heavy chain (F59 and A4.1025, both at 1:10; DSHB), Myomesin B4 (mMAC, 1:10; DSHB), Tropomyosin (CH1, 1:200; Sigma, T9283) and Ttn (MIR, 1:200; a gift from the Siegfried Labeit laboratory). Actin filaments were visualized using Alexa Fluor 568 Phalloidin (1:40, Thermo Fisher Scientific). After image acquisition, the genotypes of all embryos used were determined by PCR and restriction enzyme digestion. For each experiment, more than ten mutant embryos were analyzed to confirm the phenotypes.

Microscope image acquisition

After immunostaining, embryos were imaged using a Zeiss Axioplan 2 microscope with Apotome (Zeiss) and an Axiocam Camera (Carl Zeiss) using 20× (Plan-Apochromat) and 63× (Plan-Neofluar) objective lenses combined with a 10× ocular lens. The images were taken and analyzed using AxioVision SE64 4.8.1 software (Zeiss).

Transmission electron microscopy

Embryos at 2 dpf were fixed in Trump's solution (4% paraformaldehyde and 1% glutaraldehyde in 0.1 M phosphate buffer, pH 7.2) at room temperature for 1 h and then overnight at 4°C. Fixed tissues were then embedded in 2% agarose and subsequently processed and imaged at the Mayo Clinic Electron Microscopy Core Facility using a Philips CM10 transmission electron microscope.

Tricaine and blebbistatin treatment

To stop the movement of embryonic somites, embryos at 24 hpf were incubated in E3 water containing either 1% tricaine (Argent Labs) or 10 µM blebbistatin (Sigma) for 24 h (Yang and Xu, 2012; Yang et al., 2014a). To activate muscle contraction, embryos at 24 pdf were incubated in 0.07% bleach to harden the chorion (Zhang et al., 2009) or treated with 0.5 mM galanthamine (Otten and Abdelilah-Seyfried, 2013).

Statistical analysis

Data were analyzed by JMP software using one-way ANOVA to compare groups. P<0.05 was considered significant. *P<0.05, ***P<0.01 and ***P<0.00001. Data are presented as the mean±s.d.

We thank Beninio Gore and Kashia Stragey for zebrafish maintenance.

Author contributions

Y.-H.S., A.V.D., P.Z., X.M., M.K. and Y.D.: Acquisition, analysis and interpretation of data; Y.-H.S. and X.X.: Conception and design of the study and drafting or revising the article. All authors approved the manuscript.

Funding

This work was supported by funds from the National Heart, Lung, and Blood Institute (HL81753 and HL107304 to X.X.) and the Mayo Foundation for Medical Education and Research to X.X. Deposited in PMC for release after 12 months.

Campbell
,
J. M.
,
Hartjes
,
K. A.
,
Nelson
,
T. J.
,
Xu
,
X.
and
Ekker
,
S. C.
(
2013
).
New and TALENted genome engineering toolbox
.
Circ. Res.
113
,
571
-
587
.
Cermak
,
T.
,
Doyle
,
E. L.
,
Christian
,
M.
,
Wang
,
L.
,
Zhang
,
Y.
,
Schmidt
,
C.
,
Baller
,
J. A.
,
Somia
,
N. V.
,
Bogdanove
,
A. J.
and
Voytas
,
D. F.
(
2011
).
Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting
.
Nucleic Acids Res.
39
,
e82
.
Chauveau
,
C.
,
Rowell
,
J.
and
Ferreiro
,
A.
(
2014
).
A rising titan: TTN review and mutation update
.
Hum. Mutat.
35
,
1046
-
1059
.
Devoto
,
S. H.
,
Melançon
,
E.
,
Eisen
,
J. S.
and
Westerfield
,
M.
(
1996
).
Identification of separate slow and fast muscle precursor cells in vivo, prior to somite formation
.
Development
122
,
3371
-
3380
.
Gerull
,
B.
,
Gramlich
,
M.
,
Atherton
,
J.
,
McNabb
,
M.
,
Trombitás
,
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 familial dilated cardiomyopathy
.
Nat. Genet.
30
,
201
-
204
.
Gerull
,
B.
,
Atherton
,
J.
,
Geupel
,
A.
,
Sasse-Klaassen
,
S.
,
Heuser
,
A.
,
Frenneaux
,
M.
,
McNabb
,
M.
,
Granzier
,
H.
,
Labeit
,
S.
and
Thierfelder
,
L.
(
2006
).
Identification of a novel frameshift mutation in the giant muscle filament titin in a large Australian family with dilated cardiomyopathy
.
J. Mol. Med.
84
,
478
-
483
.
Gramlich
,
M.
,
Michely
,
B.
,
Krohne
,
C.
,
Heuser
,
A.
,
Erdmann
,
B.
,
Klaassen
,
S.
,
Hudson
,
B.
,
Magarin
,
M.
,
Kirchner
,
F.
,
Todiras
,
M.
 et al.  (
2009
).
Stress-induced dilated cardiomyopathy in a knock-in mouse model mimicking human titin-based disease
.
J. Mol. Cell. Cardiol.
47
,
352
-
358
.
Granzier
,
H. L.
and
Labeit
,
S.
(
2004
).
The giant protein titin: a major player in myocardial mechanics, signaling, and disease
.
Circ. Res.
94
,
284
-
295
.
Granzier
,
H. L.
,
Hutchinson
,
K. R.
,
Tonino
,
P.
,
Methawasin
,
M.
,
Li
,
F. W.
,
Slater
,
R. E.
,
Bull
,
M. M.
,
Saripalli
,
C.
,
Pappas
,
C. T.
,
Gregorio
,
C. C.
 et al.  (
2014
).
Deleting titin's I-band/A-band junction reveals critical roles for titin in biomechanical sensing and cardiac function
.
Proc. Natl. Acad. Sci. USA
111
,
14589
-
14594
.
Guo
,
W.
,
Bharmal
,
S. J.
,
Esbona
,
K.
and
Greaser
,
M. L.
(
2010
).
Titin diversity--alternative splicing gone wild
.
J. Biomed. Biotechnol.
2010
,
753675
.
Hackman
,
P.
,
Vihola
,
A.
,
Haravuori
,
H.
,
Marchand
,
S.
,
Sarparanta
,
J.
,
de Seze
,
J.
,
Labeit
,
S.
,
Witt
,
C.
,
Peltonen
,
L.
,
Richard
,
I.
 et al.  (
2002
).
Tibial muscular dystrophy is a titinopathy caused by mutations in TTN, the gene encoding the giant skeletal-muscle protein titin
.
Am. J. Hum. Genet.
71
,
492
-
500
.
Herman
,
D. S.
,
Lam
,
L.
,
Taylor
,
M. R. G.
,
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
.
Hill
,
C. S.
,
Duran
,
S.
,
Lin
,
Z. X.
,
Weber
,
K.
and
Holtzer
,
H.
(
1986
).
Titin and myosin, but not desmin, are linked during myofibrillogenesis in postmitotic mononucleated myoblasts
.
J. Cell Biol.
103
,
2185
-
2196
.
Hinson
,
J. T.
,
Chopra
,
A.
,
Nafissi
,
N.
,
Polacheck
,
W. J.
,
Benson
,
C. C.
,
Swist
,
S.
,
Gorham
,
J.
,
Yang
,
L.
,
Schafer
,
S.
,
Sheng
,
C. C.
 et al.  (
2015
).
HEART DISEASE. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy
.
Science
349
,
982
-
986
.
Huang
,
W.
,
Zhang
,
R.
and
Xu
,
X.
(
2009
).
Myofibrillogenesis in the developing zebrafish heart: A functional study of tnnt2
.
Dev. Biol.
331
,
237
-
249
.
Kok
,
F. O.
,
Shin
,
M.
,
Ni
,
C.-W.
,
Gupta
,
A.
,
Grosse
,
A. S.
,
van Impel
,
A.
,
Kirchmaier
,
B. C.
,
Peterson-Maduro
,
J.
,
Kourkoulis
,
G.
,
Male
,
I.
 et al.  (
2015
).
Reverse genetic screening reveals poor correlation between morpholino-induced and mutant phenotypes in zebrafish
.
Dev. Cell
32
,
97
-
108
.
Komiyama
,
M.
,
Maruyama
,
K.
and
Shimada
,
Y.
(
1990
).
Assembly of connectin (titin) in relation to myosin and alpha-actinin in cultured cardiac myocytes
.
J. Muscle Res. Cell Motil.
11
,
419
-
428
.
Labeit
,
S.
and
Kolmerer
,
B.
(
1995
).
Titins: giant proteins in charge of muscle ultrastructure and elasticity
.
Science
270
,
293
-
296
.
Labeit
,
S.
,
Gautel
,
M.
,
Lakey
,
A.
and
Trinick
,
J.
(
1992
).
Towards a molecular understanding of titin
.
EMBO J.
11
,
1711
-
1716
.
LeWinter
,
M. M.
and
Granzier
,
H. L.
(
2013
).
Titin is a major human disease gene
.
Circulation
127
,
938
-
944
.
McKeown
,
K. A.
,
Downes
,
G. B.
and
Hutson
,
L. D.
(
2009
).
Modular laboratory exercises to analyze the development of zebrafish motor behavior
.
Zebrafish
6
,
179
-
185
.
Myhre
,
J. L.
,
Hills
,
J. A.
,
Prill
,
K.
,
Wohlgemuth
,
S. L.
and
Pilgrim
,
D. B.
(
2014
).
The titin A-band rod domain is dispensable for initial thick filament assembly in zebrafish
.
Dev. Biol.
387
,
93
-
108
.
Norton
,
N.
,
Li
,
D.
,
Rampersaud
,
E.
,
Morales
,
A.
,
Martin
,
E. R.
,
Zuchner
,
S.
,
Guo
,
S.
,
Gonzalez
,
M.
,
Hedges
,
D. J.
,
Robertson
,
P. D.
, et al. 
(
2013
).
Exome sequencing and genome-wide linkage analysis in 17 families illustrate the complex contribution of TTN truncating variants to dilated cardiomyopathy
.
Circ. Cardiovasc. Genet.
6
,
144
-
153
.
Otten
,
C.
and
Abdelilah-Seyfried
,
S.
(
2013
).
Laser-inflicted injury of zebrafish embryonic skeletal muscle
.
J. Vis. Exp.
17
,
e4351
.
Radke
,
M. H.
,
Peng
,
J.
,
Wu
,
Y.
,
McNabb
,
M.
,
Nelson
,
O. L.
,
Granzier
,
H.
and
Gotthardt
,
M.
(
2007
).
Targeted deletion of titin N2B region leads to diastolic dysfunction and cardiac atrophy
.
Proc. Natl. Acad. Sci. USA
104
,
3444
-
3449
.
Roberts
,
A. M.
,
Ware
,
J. S.
,
Herman
,
D. S.
,
Schafer
,
S.
,
Baksi
,
J.
,
Bick
,
A. G.
,
Buchan
,
R. J.
,
Walsh
,
R.
,
John
,
S.
,
Wilkinson
,
S.
 et al.  (
2015
).
Integrated allelic, transcriptional, and phenomic dissection of the cardiac effects of titin truncations in health and disease
.
Sci. Transl. Med.
7
,
270ra6
.
Rossi
,
A.
,
Kontarakis
,
Z.
,
Gerri
,
C.
,
Nolte
,
H.
,
Hölper
,
S.
,
Krüger
,
M.
and
Stainier
,
D. Y. R.
(
2015
).
Genetic compensation induced by deleterious mutations but not gene knockdowns
.
Nature
524
,
230
-
233
.
Sanger
,
J. W.
,
Wang
,
J.
,
Holloway
,
B.
,
Du
,
A.
and
Sanger
,
J. M.
(
2009
).
Myofibrillogenesis in skeletal muscle cells in zebrafish
.
Cell Motil. Cytoskelet.
66
,
556
-
566
.
Schoenebeck
,
J. J.
and
Yelon
,
D.
(
2007
).
Illuminating cardiac development: advances in imaging add new dimensions to the utility of zebrafish genetics
.
Semin. Cell Dev. Biol.
18
,
27
-
35
.
Seeley
,
M.
,
Huang
,
W.
,
Chen
,
Z.
,
Wolff
,
W. O.
,
Lin
,
X.
and
Xu
,
X.
(
2007
).
Depletion of zebrafish titin reduces cardiac contractility by disrupting the assembly of Z-discs and A-bands
.
Circ. Res.
100
,
238
-
245
.
Steffen
,
L. S.
,
Guyon
,
J. R.
,
Vogel
,
E. D.
,
Howell
,
M. H.
,
Zhou
,
Y.
,
Weber
,
G. J.
,
Zon
,
L. I.
and
Kunkel
,
L. M.
(
2007
).
The zebrafish runzel muscular dystrophy is linked to the titin gene
.
Dev. Biol.
309
,
180
-
192
.
Straight
,
A. F.
,
Cheung
,
A.
,
Limouze
,
J.
,
Chen
,
I.
,
Westwood
,
N. J.
,
Sellers
,
J. R.
and
Mitchison
,
T. J.
(
2003
).
Dissecting temporal and spatial control of cytokinesis with a myosin II Inhibitor
.
Science
299
,
1743
-
1747
.
Trinick
,
J.
(
1996
).
Cytoskeleton: titin as a scaffold and spring
.
Curr. Biol.
6
,
258
-
260
.
Udd
,
B.
,
Vihola
,
A.
,
Sarparanta
,
J.
,
Richard
,
I.
and
Hackman
,
P.
(
2005
).
Titinopathies and extension of the M-line mutation phenotype beyond distal myopathy and LGMD2J
.
Neurology
64
,
636
-
642
.
Wang
,
S. M.
,
Greaser
,
M. L.
,
Schultz
,
E.
,
Bulinski
,
J. C.
,
Lin
,
J. J.
and
Lessard
,
J. L.
(
1988
).
Studies on cardiac myofibrillogenesis with antibodies to titin, actin, tropomyosin, and myosin
.
J. Cell Biol.
107
,
1075
-
1083
.
Warren
,
C. M.
,
Krzesinski
,
P. R.
and
Greaser
,
M. L.
(
2003
).
Vertical agarose gel electrophoresis and electroblotting of high-molecular-weight proteins
.
Electrophoresis
24
,
1695
-
1702
.
Weinert
,
S.
,
Bergmann
,
N.
,
Luo
,
X.
,
Erdmann
,
B.
and
Gotthardt
,
M.
(
2006
).
M line-deficient titin causes cardiac lethality through impaired maturation of the sarcomere
.
J. Cell Biol.
173
,
559
-
570
.
Whiting
,
A.
,
Wardale
,
J.
and
Trinick
,
J.
(
1989
).
Does titin regulate the length of muscle thick filaments?
J. Mol. Biol.
205
,
263
-
268
.
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 an alternatively spliced exon of titin
.
Nat. Genet.
30
,
205
-
209
.
Yang
,
J.
and
Xu
,
X.
(
2012
).
alpha-Actinin2 is required for the lateral alignment of Z discs and ventricular chamber enlargement during zebrafish cardiogenesis
.
FASEB J.
26
,
4230
-
4242
.
Yang
,
J.
,
Hartjes
,
K. A.
,
Nelson
,
T. J.
and
Xu
,
X.
(
2014a
).
Cessation of contraction induces cardiomyocyte remodeling during zebrafish cardiogenesis
.
Am. J. Physiol. Heart Circ. Physiol.
306
,
H382
-
H395
.
Yang
,
J.
,
Shih
,
Y.-H.
and
Xu
,
X.
(
2014b
).
Understanding cardiac sarcomere assembly with zebrafish genetics
.
Anat. Rec.
297
,
1681
-
1693
.
Zhang
,
R.
,
Yang
,
J.
,
Zhu
,
J.
and
Xu
,
X.
(
2009
).
Depletion of zebrafish Tcap leads to muscular dystrophy via disrupting sarcomere-membrane interaction, not sarcomere assembly
.
Hum. Mol. Genet.
18
,
4130
-
4140
.
Zou
,
J.
,
Tran
,
D.
,
Baalbaki
,
M.
,
Tang
,
L. F.
,
Poon
,
A.
,
Pelonero
,
A.
,
Titus
,
E. W.
,
Yuan
,
C.
,
Shi
,
C.
,
Patchava
,
S.
 et al.  (
2015
).
An internal promoter underlies the difference in disease severity between N- and C-terminal truncation mutations of Titin in zebrafish
.
Elife
4
,
e09406
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information